1
|
Nagarajan A, Scoggin K, Adams LG, Threadgill D, Andrews-Polymenis H. Identification of a genetic region linked to tolerance to MRSA infection using Collaborative Cross mice. PLoS Genet 2024; 20:e1011378. [PMID: 39178306 PMCID: PMC11407622 DOI: 10.1371/journal.pgen.1011378] [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: 10/10/2023] [Revised: 09/17/2024] [Accepted: 07/29/2024] [Indexed: 08/25/2024] Open
Abstract
Staphylococcus aureus (S. aureus) colonizes humans asymptomatically but can also cause opportunistic infections, ranging from mild skin infections to severe life-threatening conditions. Resistance and tolerance are two ways a host can survive an infection. Resistance is limiting the pathogen burden, while tolerance is limiting the health impact of a given pathogen burden. In previous work, we established that collaborative cross (CC) mouse line CC061 is highly susceptible to Methicillin-resistant S. aureus infection (MRSA, USA300), while CC024 is tolerant. To identify host genes involved in tolerance after S. aureus infection, we crossed CC061 mice and CC024 mice to generate F1 and F2 populations. Survival after MRSA infection in the F1 and F2 generations was 65% and 55% and followed a complex dominant inheritance pattern for the CC024 increased survival phenotype. Colonization in F2 animals was more extreme than in their parents, suggesting successful segregation of genetic factors. We identified a Quantitative Trait Locus (QTL) peak on chromosome 7 for survival and weight change after infection. In this QTL, the WSB/EiJ (WSB) allele was present in CC024 mice and contributed to their MRSA tolerant phenotype. Two genes, C5ar1 and C5ar2, have high-impact variants in this region. C5ar1 and C5ar2 are receptors for the complement factor C5a, an anaphylatoxin that can trigger a massive immune response by binding to these receptors. We hypothesize that C5a may have altered binding to variant receptors in CC024 mice, reducing damage caused by the cytokine storm and resulting in the ability to tolerate a higher pathogen burden and longer survival.
Collapse
Affiliation(s)
- Aravindh Nagarajan
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - Kristin Scoggin
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| | - L Garry Adams
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, United States of America
| | - David Threadgill
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Molecular and Cellular Medicine, Texas A&M University, College Station, Texas, United States of America
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, Texas, United States of America
- Department of Biochemistry & Biophysics and Department of Nutrition, Texas A&M University, College Station, Texas, United States of America
| | - Helene Andrews-Polymenis
- Interdisciplinary Program in Genetics and Genomics, Texas A&M University, College Station, Texas, United States of America
- Department of Microbial Pathogenesis and Immunology, Texas A&M University, College Station, Texas, United States of America
| |
Collapse
|
2
|
Cheetham CJ, McKelvey MC, McAuley DF, Taggart CC. Neutrophil-Derived Proteases in Lung Inflammation: Old Players and New Prospects. Int J Mol Sci 2024; 25:5492. [PMID: 38791530 PMCID: PMC11122108 DOI: 10.3390/ijms25105492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Neutrophil-derived proteases are critical to the pathology of many inflammatory lung diseases, both chronic and acute. These abundant enzymes play roles in key neutrophil functions, such as neutrophil extracellular trap formation and reactive oxygen species release. They may also be released, inducing tissue damage and loss of tissue function. Historically, the neutrophil serine proteases (NSPs) have been the main subject of neutrophil protease research. Despite highly promising cell-based and animal model work, clinical trials involving the inhibition of NSPs have shown mixed results in lung disease patients. As such, the cutting edge of neutrophil-derived protease research has shifted to proteases that have had little-to-no research in neutrophils to date. These include the cysteine and serine cathepsins, the metzincins and the calpains, among others. This review aims to outline the previous work carried out on NSPs, including the shortcomings of some of the inhibitor-orientated clinical trials. Our growing understanding of other proteases involved in neutrophil function and neutrophilic lung inflammation will then be discussed. Additionally, the potential of targeting these more obscure neutrophil proteases will be highlighted, as they may represent new targets for inhibitor-based treatments of neutrophil-mediated lung inflammation.
Collapse
Affiliation(s)
- Coby J. Cheetham
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Michael C. McKelvey
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| | - Daniel F. McAuley
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK;
| | - Clifford C. Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine and Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK; (C.J.C.); (M.C.M.)
| |
Collapse
|
3
|
Duda HC, von Toerne C, Korbonits L, Didier A, Scholz AM, Märtlbauer E, Hauck SM, Deeg CA. Cathepsin S Is More Abundant in Serum of Mycobacterium avium subsp. paratuberculosis-Infected Dairy Cows. Metabolites 2024; 14:215. [PMID: 38668343 PMCID: PMC11051907 DOI: 10.3390/metabo14040215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Mycobacterium avium subsp. paratuberculosis (MAP) is the causative agent of bovine paratuberculosis, a chronic granulomatous enteritis leading to economic losses and posing a risk to human health due to its zoonotic potential. The pathogen cannot reliably be detected by standard methods, and immunological procedures during the infection are not well understood. Therefore, the aim of our study was to explore host-pathogen interactions in MAP-infected dairy cows and to improve diagnostic tests. Serum proteomics analysis using quantitative label-free LC-MS/MS revealed 60 differentially abundant proteins in MAP-infected dairy cows compared to healthy controls from the same infected herd and 90 differentially abundant proteins in comparison to another control group from an uninfected herd. Pathway enrichment analysis provided new insights into the immune response to MAP and susceptibility to the infection. Furthermore, we found a higher abundance of Cathepsin S (CTSS) in the serum of MAP-infected dairy cows, which is involved in multiple enriched pathways associated with the immune system. Confirmed with Western blotting, we identified CTSS as a potential biomarker for bovine paratuberculosis. This study enabled a better understanding of procedures in the host-pathogen response to MAP and improved detection of paratuberculosis-diseased cattle.
Collapse
Affiliation(s)
- Heidi C. Duda
- Chair of Physiology, Department of Veterinary Sciences, Ludwig Maximilian University of Munich, D-82152 Martinsried, Germany
| | - Christine von Toerne
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-85763 Neuherberg, Germany (S.M.H.)
| | - Lucia Korbonits
- Chair of Physiology, Department of Veterinary Sciences, Ludwig Maximilian University of Munich, D-82152 Martinsried, Germany
| | - Andrea Didier
- Chair of Hygiene and Technology of Milk, Department of Veterinary Sciences, Ludwig Maximilian University of Munich, D-85764 Oberschleißheim, Germany; (A.D.)
| | - Armin M. Scholz
- Livestock Center of the Faculty of Veterinary Medicine, Ludwig Maximilian University of Munich, D-85764 Oberschleißheim, Germany;
| | - Erwin Märtlbauer
- Chair of Hygiene and Technology of Milk, Department of Veterinary Sciences, Ludwig Maximilian University of Munich, D-85764 Oberschleißheim, Germany; (A.D.)
| | - Stefanie M. Hauck
- Metabolomics and Proteomics Core, Helmholtz Center Munich, German Research Center for Environmental Health, D-85763 Neuherberg, Germany (S.M.H.)
| | - Cornelia A. Deeg
- Chair of Physiology, Department of Veterinary Sciences, Ludwig Maximilian University of Munich, D-82152 Martinsried, Germany
| |
Collapse
|
4
|
Capitani N, Baldari CT. The Immunological Synapse: An Emerging Target for Immune Evasion by Bacterial Pathogens. Front Immunol 2022; 13:943344. [PMID: 35911720 PMCID: PMC9325968 DOI: 10.3389/fimmu.2022.943344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Similar to other pathogens, bacteria have developed during their evolution a variety of mechanisms to overcome both innate and acquired immunity, accounting for their ability to cause disease or chronic infections. The mechanisms exploited for this critical function act by targeting conserved structures or pathways that regulate the host immune response. A strategic potential target is the immunological synapse (IS), a highly specialized structure that forms at the interface between antigen presenting cells (APC) and T lymphocytes and is required for the establishment of an effective T cell response to the infectious agent and for the development of long-lasting T cell memory. While a variety of bacterial pathogens are known to impair or subvert cellular processes essential for antigen processing and presentation, on which IS assembly depends, it is only recently that the possibility that IS may be a direct target of bacterial virulence factors has been considered. Emerging evidence strongly supports this notion, highlighting IS targeting as a powerful, novel means of immune evasion by bacterial pathogens. In this review we will present a brief overview of the mechanisms used by bacteria to affect IS assembly by targeting APCs. We will then summarize what has emerged from the current handful of studies that have addressed the direct impact of bacterial virulence factors on IS assembly in T cells and, based on the strategic cellular processes targeted by these factors in other cell types, highlight potential IS-related vulnerabilities that could be exploited by these pathogens to evade T cell mediated immunity.
Collapse
Affiliation(s)
- Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | | |
Collapse
|
5
|
Kowalewicz-Kulbat M, Locht C. Recombinant BCG to Enhance Its Immunomodulatory Activities. Vaccines (Basel) 2022; 10:827. [PMID: 35632582 PMCID: PMC9143156 DOI: 10.3390/vaccines10050827] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
The bacillus Calmette-Guérin (BCG) is an attenuated Mycobacterium bovis derivative that has been widely used as a live vaccine against tuberculosis for a century. In addition to its use as a tuberculosis vaccine, BCG has also been found to have utility in the prevention or treatment of unrelated diseases, including cancer. However, the protective and therapeutic efficacy of BCG against tuberculosis and other diseases is not perfect. For three decades, it has been possible to genetically modify BCG in an attempt to improve its efficacy. Various immune-modulatory molecules have been produced in recombinant BCG strains and tested for protection against tuberculosis or treatment of several cancers or inflammatory diseases. These molecules include cytokines, bacterial toxins or toxin fragments, as well as other protein and non-protein immune-modulatory molecules. The deletion of genes responsible for the immune-suppressive properties of BCG has also been explored for their effect on BCG-induced innate and adaptive immune responses. Most studies limited their investigations to the description of T cell immune responses that were modified by the genetic modifications of BCG. Some studies also reported improved protection by recombinant BCG against tuberculosis or enhanced therapeutic efficacy against various cancer forms or allergies. However, so far, these investigations have been limited to mouse models, and the prophylactic or therapeutic potential of recombinant BCG strains has not yet been illustrated in other species, including humans, with the exception of a genetically modified BCG strain that is now in late-stage clinical development as a vaccine against tuberculosis. In this review, we provide an overview of the different molecular engineering strategies adopted over the last three decades in order to enhance the immune-modulatory potential of BCG.
Collapse
Affiliation(s)
- Magdalena Kowalewicz-Kulbat
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| | - Camille Locht
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
- CHU Lille, Institut Pasteur de Lille, U1019–UMR9017–CIIL–Center for Infection and Immunity of Lille, University Lille, CNRS, Inserm, F-59000 Lille, France
| |
Collapse
|
6
|
Involvement of Cathepsins Protein in Mycobacterial Infection and Its Future Prospect as a Therapeutic Target. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10385-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
7
|
Dwivedi V, Gautam S, Headley CA, Piergallini T, Torrelles JB, Turner J. IL-10 Receptor Blockade Delivered Simultaneously with Bacillus Calmette-Guérin Vaccination Sustains Long-Term Protection against Mycobacterium tuberculosis Infection in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1406-1416. [PMID: 35181640 PMCID: PMC11075079 DOI: 10.4049/jimmunol.2100900] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/12/2022] [Indexed: 11/19/2022]
Abstract
Mycobacterium bovis bacillus Calmette-Guérin (BCG) immunization still remains the best vaccination strategy available to control the development of active tuberculosis. Protection afforded by BCG vaccination gradually wanes over time and although booster strategies have promise, they remain under development. An alternative approach is to improve BCG efficacy through host-directed therapy. Building upon prior knowledge that blockade of IL-10R1 during early Mycobacterium tuberculosis infection improves and extends control of M. tuberculosis infection in mice, we employed a combined anti-IL-10R1/BCG vaccine strategy. An s.c. single vaccination of BCG/anti-IL10-R1 increased the numbers of CD4+ and CD8+ central memory T cells and reduced Th1 and Th17 cytokine levels in the lung for up to 7 wk postvaccination. Subsequent M. tuberculosis challenge in mice showed both an early (4 wk) and sustained long-term (47 wk) control of infection, which was associated with increased survival. In contrast, protection of BCG/saline-vaccinated mice waned 8 wk after M. tuberculosis infection. Our findings demonstrate that a single and simultaneous vaccination with BCG/anti-IL10-R1 sustains long-term protection, identifying a promising approach to enhance and extend the current BCG-mediated protection against TB.
Collapse
Affiliation(s)
- Varun Dwivedi
- Disease Intervention and Prevention Program, Texas Biomedical Research Institute, San Antonio, TX
| | - Shalini Gautam
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| | - Colwyn A Headley
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| | - Tucker Piergallini
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| | - Jordi B Torrelles
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX
| | - Joanne Turner
- Host Pathogen Interactions Program, Texas Biomedical Research Institute, San Antonio, TX; and
| |
Collapse
|
8
|
Naik P, Naik MN, Mishra DK, Joseph J. Methicillin resistance in Staphylococcus aureus modulates the transcriptome and disease pathology in a murine model of endophthalmitis. Exp Eye Res 2022; 218:109016. [DOI: 10.1016/j.exer.2022.109016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/05/2022] [Accepted: 02/24/2022] [Indexed: 11/16/2022]
|
9
|
Pires D, Calado M, Velez T, Mandal M, Catalão MJ, Neyrolles O, Lugo-Villarino G, Vérollet C, Azevedo-Pereira JM, Anes E. Modulation of Cystatin C in Human Macrophages Improves Anti-Mycobacterial Immune Responses to Mycobacterium tuberculosis Infection and Coinfection With HIV. Front Immunol 2021; 12:742822. [PMID: 34867965 PMCID: PMC8637326 DOI: 10.3389/fimmu.2021.742822] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/21/2021] [Indexed: 11/25/2022] Open
Abstract
Tuberculosis owes its resurgence as a major global health threat mostly to the emergence of drug resistance and coinfection with HIV. The synergy between HIV and Mycobacterium tuberculosis (Mtb) modifies the host immune environment to enhance both viral and bacterial replication and spread. In the lung immune context, both pathogens infect macrophages, establishing favorable intracellular niches. Both manipulate the endocytic pathway in order to avoid destruction. Relevant players of the endocytic pathway to control pathogens include endolysosomal proteases, cathepsins, and their natural inhibitors, cystatins. Here, a mapping of the human macrophage transcriptome for type I and II cystatins during Mtb, HIV, or Mtb-HIV infection displayed different profiles of gene expression, revealing cystatin C as a potential target to control mycobacterial infection as well as HIV coinfection. We found that cystatin C silencing in macrophages significantly improves the intracellular killing of Mtb, which was concomitant with an increased general proteolytic activity of cathepsins. In addition, downmodulation of cystatin C led to an improved expression of the human leukocyte antigen (HLA) class II in macrophages and an increased CD4+ T-lymphocyte proliferation along with enhanced IFN-γ secretion. Overall, our results suggest that the targeting of cystatin C in human macrophages represents a promising approach to improve the control of mycobacterial infections including multidrug-resistant (MDR) TB.
Collapse
Affiliation(s)
- David Pires
- Host-Pathogen Interactions, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Marta Calado
- Host-Pathogen Interactions, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Tomás Velez
- Host-Pathogen Interactions, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Manoj Mandal
- Host-Pathogen Interactions, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Maria João Catalão
- Host-Pathogen Interactions, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Olivier Neyrolles
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Toulouse, France
| | - Christel Vérollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, Centre National de la Recherche Scientifique (CNRS), Toulouse, France
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Elsa Anes
- Host-Pathogen Interactions, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
10
|
Anes E, Azevedo-Pereira JM, Pires D. Cathepsins and Their Endogenous Inhibitors in Host Defense During Mycobacterium tuberculosis and HIV Infection. Front Immunol 2021; 12:726984. [PMID: 34421929 PMCID: PMC8371317 DOI: 10.3389/fimmu.2021.726984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/22/2021] [Indexed: 01/15/2023] Open
Abstract
The moment a very old bacterial pathogen met a young virus from the 80's defined the beginning of a tragic syndemic for humanity. Such is the case for the causative agent of tuberculosis and the human immunodeficiency virus (HIV). Syndemic is by definition a convergence of more than one disease resulting in magnification of their burden. Both pathogens work synergistically contributing to speed up the replication of each other. Mycobacterium tuberculosis (Mtb) and HIV infections are in the 21st century among the leaders of morbidity and mortality of humankind. There is an urgent need for development of new approaches for prevention, better diagnosis, and new therapies for both infections. Moreover, these approaches should consider Mtb and HIV as a co-infection, rather than just as separate problems, to prevent further aggravation of the HIV-TB syndemic. Both pathogens manipulate the host immune responses to establish chronic infections in intracellular niches of their host cells. This includes manipulation of host relevant antimicrobial proteases such as cathepsins or their endogenous inhibitors. Here we discuss recent understanding on how Mtb and HIV interact with cathepsins and their inhibitors in their multifactorial functions during the pathogenesis of both infections. Particularly we will address the role on pathogen transmission, during establishment of intracellular chronic niches and in granuloma clinical outcome and tuberculosis diagnosis. This area of research will open new avenues for the design of innovative therapies and diagnostic interventions so urgently needed to fight this threat to humanity.
Collapse
Affiliation(s)
- Elsa Anes
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - José Miguel Azevedo-Pereira
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - David Pires
- Host-Pathogen Interactions Unit, Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
11
|
Bouzeyen R, Chugh S, Gosain TP, Barbouche MR, Haoues M, Rao KVS, Essafi M, Singh R. Co-Administration of Anticancer Candidate MK-2206 Enhances the Efficacy of BCG Vaccine Against Mycobacterium tuberculosis in Mice and Guinea Pigs. Front Immunol 2021; 12:645962. [PMID: 34122406 PMCID: PMC8190480 DOI: 10.3389/fimmu.2021.645962] [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: 12/24/2020] [Accepted: 04/29/2021] [Indexed: 01/19/2023] Open
Abstract
The failure of M. bovis BCG to induce long-term protection has been endowed to its inability to escape the phagolysosome, leading to mild activation of CD8+ mediated T cell response. Induction of apoptosis in host cells plays an important role in potentiating dendritic cells-mediated priming of CD8+ T cells, a process defined as “cross-priming.” Moreover, IL-10 secretion by infected cells has been reported to hamper BCG-induced immunity against Tuberculosis (TB). Previously, we have reported that apoptosis of BCG-infected macrophages and inhibition of IL-10 secretion is FOXO3 dependent, a transcription factor negatively regulated by the pro-survival activated threonine kinase, Akt. We speculate that FOXO3-mediated induction of apoptosis and abrogation of IL-10 secretion along with M. bovis BCG immunization might enhance the protection imparted by BCG. Here, we have assessed whether co-administration of a known anti-cancer Akt inhibitor, MK-2206, enhances the protective efficacy of M. bovis BCG in mice model of infection. We observed that in vitro MK-2206 treatment resulted in FOXO3 activation, enhanced BCG-induced apoptosis of macrophages and inhibition of IL-10 secretion. Co-administration of M. bovis BCG along with MK-2206 also increased apoptosis of antigen-presenting cells in draining lymph nodes of immunized mice. Further, MK-2206 administration improved BCG-induced CD4+ and CD8+ effector T cells responses and its ability to induce both effector and central memory T cells. Finally, we show that co-administration of MK-2206 enhanced the protection imparted by M. bovis BCG against Mtb in aerosol infected mice and guinea pigs. Taken together, we provide evidence that MK-2206-mediated activation of FOXO3 potentiates BCG-induced immunity and imparts protection against Mtb through enhanced innate immune response.
Collapse
Affiliation(s)
- Rania Bouzeyen
- Institut Pasteur de Tunis, LTCII, LR11 IPT02, Tunis, Tunisia
| | - Saurabh Chugh
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | - Meriam Haoues
- Institut Pasteur de Tunis, LTCII, LR11 IPT02, Tunis, Tunisia
| | - Kanury V S Rao
- Translational Health Science and Technology Institute, Faridabad, India
| | - Makram Essafi
- Institut Pasteur de Tunis, LTCII, LR11 IPT02, Tunis, Tunisia
| | - Ramandeep Singh
- Translational Health Science and Technology Institute, Faridabad, India
| |
Collapse
|
12
|
Covián C, Ríos M, Berríos-Rojas RV, Bueno SM, Kalergis AM. Induction of Trained Immunity by Recombinant Vaccines. Front Immunol 2021; 11:611946. [PMID: 33584692 PMCID: PMC7873984 DOI: 10.3389/fimmu.2020.611946] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 11/24/2020] [Indexed: 01/24/2023] Open
Abstract
Vaccines represent an important strategy to protect humans against a wide variety of pathogens and have even led to eradicating some diseases. Although every vaccine is developed to induce specific protection for a particular pathogen, some vaccine formulations can also promote trained immunity, which is a non-specific memory-like feature developed by the innate immune system. It is thought that trained immunity can protect against a wide variety of pathogens other than those contained in the vaccine formulation. The non-specific memory of the trained immunity-based vaccines (TIbV) seems beneficial for the immunized individual, as it may represent a powerful strategy that contributes to the control of pathogen outbreaks, reducing morbidity and mortality. A wide variety of respiratory viruses, including respiratory syncytial virus (hRSV) and metapneumovirus (hMPV), cause serious illness in children under 5 years old and the elderly. To address this public health problem, we have developed recombinant BCG vaccines that have shown to be safe and immunogenic against hRSV or hMPV. Besides the induction of specific adaptive immunity against the viral antigens, these vaccines could generate trained immunity against other respiratory pathogens. Here, we discuss some of the features of trained immunity induced by BCG and put forward the notion that recombinant BCGs expressing hRSV or hMPV antigens have the capacity to simultaneously induce specific adaptive immunity and non-specific trained immunity. These recombinant BCG vaccines could be considered as TIbV capable of inducing simultaneously the development of specific protection against hRSV or hMPV, as well as non-specific trained-immunity-based protection against other pathogenic viruses.
Collapse
Affiliation(s)
- Camila Covián
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Ríos
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roslye V. Berríos-Rojas
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
13
|
Szulc-Dąbrowska L, Bossowska-Nowicka M, Struzik J, Toka FN. Cathepsins in Bacteria-Macrophage Interaction: Defenders or Victims of Circumstance? Front Cell Infect Microbiol 2020; 10:601072. [PMID: 33344265 PMCID: PMC7746538 DOI: 10.3389/fcimb.2020.601072] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/05/2020] [Indexed: 02/06/2023] Open
Abstract
Macrophages are the first encounters of invading bacteria and are responsible for engulfing and digesting pathogens through phagocytosis leading to initiation of the innate inflammatory response. Intracellular digestion occurs through a close relationship between phagocytic/endocytic and lysosomal pathways, in which proteolytic enzymes, such as cathepsins, are involved. The presence of cathepsins in the endo-lysosomal compartment permits direct interaction with and killing of bacteria, and may contribute to processing of bacterial antigens for presentation, an event necessary for the induction of antibacterial adaptive immune response. Therefore, it is not surprising that bacteria can control the expression and proteolytic activity of cathepsins, including their inhibitors – cystatins, to favor their own intracellular survival in macrophages. In this review, we summarize recent developments in defining the role of cathepsins in bacteria-macrophage interaction and describe important strategies engaged by bacteria to manipulate cathepsin expression and activity in macrophages. Particularly, we focus on specific bacterial species due to their clinical relevance to humans and animal health, i.e., Mycobacterium, Mycoplasma, Staphylococcus, Streptococcus, Salmonella, Shigella, Francisella, Chlamydia, Listeria, Brucella, Helicobacter, Neisseria, and other genera.
Collapse
Affiliation(s)
- Lidia Szulc-Dąbrowska
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Magdalena Bossowska-Nowicka
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Justyna Struzik
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland
| | - Felix N Toka
- Division of Immunology, Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Sciences-Szkoła Główna Gospodarstwa Wejskiego, Warsaw, Poland.,Center for Integrative Mammalian Research, Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
| |
Collapse
|
14
|
Abstract
Mycobacterium tuberculosis remains the leading cause of death attributed to a single infectious organism. Bacillus Calmette-Guerin (BCG), the standard vaccine against M. tuberculosis, is thought to prevent only 5% of all vaccine-preventable deaths due to tuberculosis, thus an alternative vaccine is required. One of the principal barriers to vaccine development against M. tuberculosis is the complexity of the immune response to infection, with uncertainty as to what constitutes an immunological correlate of protection. In this paper, we seek to give an overview of the immunology of M. tuberculosis infection, and by doing so, investigate possible targets of vaccine development. This encompasses the innate, adaptive, mucosal and humoral immune systems. Though MVA85A did not improve protection compared with BCG alone in a large-scale clinical trial, the correlates of protection this has revealed, in addition to promising results from candidate such as VPM1002, M72/ASO1E and H56:IC31 point to a brighter future in the field of TB vaccine development.
Collapse
Affiliation(s)
- Benedict Brazier
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| | - Helen McShane
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7DQ UK
| |
Collapse
|
15
|
Brown R, Nath S, Lora A, Samaha G, Elgamal Z, Kaiser R, Taggart C, Weldon S, Geraghty P. Cathepsin S: investigating an old player in lung disease pathogenesis, comorbidities, and potential therapeutics. Respir Res 2020; 21:111. [PMID: 32398133 PMCID: PMC7216426 DOI: 10.1186/s12931-020-01381-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
Dysregulated expression and activity of cathepsin S (CTSS), a lysosomal protease and a member of the cysteine cathepsin protease family, is linked to the pathogenesis of multiple diseases, including a number of conditions affecting the lungs. Extracellular CTSS has potent elastase activity and by processing cytokines and host defense proteins, it also plays a role in the regulation of inflammation. CTSS has also been linked to G-coupled protein receptor activation and possesses an important intracellular role in major histocompatibility complex class II antigen presentation. Modulated CTSS activity is also associated with pulmonary disease comorbidities, such as cancer, cardiovascular disease, and diabetes. CTSS is expressed in a wide variety of immune cells and is biologically active at neutral pH. Herein, we review the significance of CTSS signaling in pulmonary diseases and associated comorbidities. We also discuss CTSS as a plausible therapeutic target and describe recent and current clinical trials examining CTSS inhibition as a means for treatment.
Collapse
Affiliation(s)
- Ryan Brown
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sridesh Nath
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Alnardo Lora
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Ghassan Samaha
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Ziyad Elgamal
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Ryan Kaiser
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA
| | - Clifford Taggart
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Sinéad Weldon
- Airway Innate Immunity Research (AiiR) Group, Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, Northern Ireland, UK
| | - Patrick Geraghty
- Division of Pulmonary & Critical Care Medicine, Department of Medicine, State University of New York Downstate Medical Centre, Brooklyn, NY, USA.
- Department of Cell Biology, State University of New York Downstate Medical Centre, Brooklyn, NY, USA.
| |
Collapse
|
16
|
Bettencourt P, Müller J, Nicastri A, Cantillon D, Madhavan M, Charles PD, Fotso CB, Wittenberg R, Bull N, Pinpathomrat N, Waddell SJ, Stylianou E, Hill AVS, Ternette N, McShane H. Identification of antigens presented by MHC for vaccines against tuberculosis. NPJ Vaccines 2020; 5:2. [PMID: 31908851 PMCID: PMC6941960 DOI: 10.1038/s41541-019-0148-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/25/2019] [Indexed: 11/09/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb) is responsible for more deaths globally than any other pathogen. The only available vaccine, bacillus Calmette-Guérin (BCG), has variable efficacy throughout the world. A more effective vaccine is urgently needed. The immune response against tuberculosis relies, at least in part, on CD4+ T cells. Protective vaccines require the induction of antigen-specific CD4+ T cells via mycobacterial peptides presented by MHC class-II in infected macrophages. In order to identify mycobacterial antigens bound to MHC, we have immunoprecipitated MHC class-I and class-II complexes from THP-1 macrophages infected with BCG, purified MHC class-I and MHC class-II peptides and analysed them by liquid chromatography tandem mass spectrometry. We have successfully identified 94 mycobacterial peptides presented by MHC-II and 43 presented by MHC-I, from 76 and 41 antigens, respectively. These antigens were found to be highly expressed in infected macrophages. Gene ontology analysis suggests most of these antigens are associated with membranes and involved in lipid biosynthesis and transport. The sequences of selected peptides were confirmed by spectral match validation and immunogenicity evaluated by IFN-gamma ELISpot against peripheral blood mononuclear cell from volunteers vaccinated with BCG, M.tb latently infected subjects or patients with tuberculosis disease. Three antigens were expressed in viral vectors, and evaluated as vaccine candidates alone or in combination in a murine aerosol M.tb challenge model. When delivered in combination, the three candidate vaccines conferred significant protection in the lungs and spleen compared with BCG alone, demonstrating proof-of-concept for this unbiased approach to identifying new candidate antigens. Protective vaccines against Mycobacterium tuberculosis (M.tb), such as bacillus Calmette-Guérin (BCG), trigger strong CD4 T-cell responses specific to mycobacterium peptides, but their efficacy is variable. Paulo Bettencourt and colleagues now identify a set of mycobacterium peptides presented by BCG-infected macrophages via major compatibility complexes (MHC), and show that three of these antigens can be combined to formulate a vaccine that confers improved protection to Mtb infection in mice. After identifying 94 MHC-II-associated and 43 MHC-I-associated mycobacterium peptides, the researchers performed immunogenicity assays with peripheral blood mononuclear cells from BCG-vaccinated donors, latent Mtb-infected patients and patients with tuberculosis, and show that a set of these peptides was recognised by the immune cells, validating their potential as possible components for new Mtb vaccine formulations. These findings further support the value of immunopeptidomics for the identification of new antigens for effective vaccine alternatives.
Collapse
Affiliation(s)
| | - Julius Müller
- 1Jenner Institute, University of Oxford, Oxford, OX3 7DQ UK
| | - Annalisa Nicastri
- 2Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ UK
| | - Daire Cantillon
- 3Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX UK
| | - Meera Madhavan
- 1Jenner Institute, University of Oxford, Oxford, OX3 7DQ UK
| | - Philip D Charles
- 2Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ UK
| | - Carine B Fotso
- 1Jenner Institute, University of Oxford, Oxford, OX3 7DQ UK
| | | | - Naomi Bull
- 1Jenner Institute, University of Oxford, Oxford, OX3 7DQ UK
| | | | - Simon J Waddell
- 3Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX UK
| | | | | | - Nicola Ternette
- 1Jenner Institute, University of Oxford, Oxford, OX3 7DQ UK.,2Target Discovery Institute, University of Oxford, Oxford, OX3 7FZ UK
| | - Helen McShane
- 1Jenner Institute, University of Oxford, Oxford, OX3 7DQ UK
| |
Collapse
|
17
|
Lewis MS, Danelishvili L, Rose SJ, Bermudez LE. MAV_4644 Interaction with the Host Cathepsin Z Protects Mycobacterium avium subsp. hominissuis from Rapid Macrophage Killing. Microorganisms 2019; 7:microorganisms7050144. [PMID: 31117286 PMCID: PMC6560410 DOI: 10.3390/microorganisms7050144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/03/2019] [Accepted: 05/10/2019] [Indexed: 01/15/2023] Open
Abstract
Mycobacterium avium subspecies hominissuis (MAH) is an opportunistic pathogen that is ubiquitous in the environment and often isolated from faucets and showerheads. MAH mostly infects humans with an underlying disease, such as chronic pulmonary disorder, cystic fibrosis, or individuals that are immunocompromised. In recent years, MAH infections in patients without concurrent disease are increasing in prevalence as well. This pathogen is resistant to many antibiotics due to the impermeability of its envelope and due to the phenotypic resistance established within the host macrophages, making difficult to treat MAH infections. By screening a MAH transposon library for mutants that are susceptible to killing by reactive nitrogen intermediaries, we identified the MAV_4644 (MAV_4644:Tn) gene knockout clone that was also significantly attenuated in growth within the host macrophages. Complementation of the mutant restored the wild-type phenotype. The MAV_4644 gene encodes a dual-function protein with a putative pore-forming function and ADP-ribosyltransferase activity. Protein binding assay suggests that MAV_4644 interacts with the host lysosomal peptidase cathepsin Z (CTSZ), a key regulator of the cell signaling and inflammation. Pathogenic mycobacteria have been shown to suppress the action of many cathepsins to establish their intracellular niche. Our results demonstrate that knocking-down the cathepsin Z in human macrophages rescues the attenuated phenotype of MAV_4644:Tn clone. Although, the purified cathepsin Z by itself does not have any killing effect on MAH, it contributes to bacterial killing in the presence of the nitric oxide (NO). Our data suggest that the cathepsin Z is involved in early macrophage killing of MAH, and the virulence factor MAV_4644 protects the pathogen from this process.
Collapse
Affiliation(s)
- Matthew S Lewis
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA.
| | - Lia Danelishvili
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA.
| | - Sasha J Rose
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA.
| | - Luiz E Bermudez
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA.
- Department of Microbiology, College of Science, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
18
|
Ernst JD. Mechanisms of M. tuberculosis Immune Evasion as Challenges to TB Vaccine Design. Cell Host Microbe 2019; 24:34-42. [PMID: 30001523 DOI: 10.1016/j.chom.2018.06.004] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tuberculosis (TB) is a large global health problem, in part because of the long period of coevolution of the pathogen, Mycobacterium tuberculosis, and its human host. A major factor that sustains the global epidemic of TB is the lack of a sufficiently effective vaccine. While basic mechanisms of immunity that protect against TB have been identified, attempts to improve immunity to TB by vaccination have been disappointing. This Review discusses the mechanisms used by M. tuberculosis to evade innate and adaptive immunity and that likely limit the efficacy of vaccines developed to date. Despite multiple mechanisms of immune evasion, recent trials have indicated that effective TB vaccines remain an attainable goal. This Review discusses how knowledge from other systems can inform improvements on current vaccine approaches.
Collapse
Affiliation(s)
- Joel D Ernst
- Division of Experimental Medicine, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
19
|
Visser JG, Van Staden ADP, Smith C. Harnessing Macrophages for Controlled-Release Drug Delivery: Lessons From Microbes. Front Pharmacol 2019; 10:22. [PMID: 30740053 PMCID: PMC6355695 DOI: 10.3389/fphar.2019.00022] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/09/2019] [Indexed: 01/15/2023] Open
Abstract
With the effectiveness of therapeutic agents ever decreasing and the increased incidence of multi-drug resistant pathogens, there is a clear need for administration of more potent, potentially more toxic, drugs. Alternatively, biopharmaceuticals may hold potential but require specialized protection from premature in vivo degradation. Thus, a paralleled need for specialized drug delivery systems has arisen. Although cell-mediated drug delivery is not a completely novel concept, the few applications described to date are not yet ready for in vivo application, for various reasons such as drug-induced carrier cell death, limited control over the site and timing of drug release and/or drug degradation by the host immune system. Here, we present our hypothesis for a new drug delivery system, which aims to negate these limitations. We propose transport of nanoparticle-encapsulated drugs inside autologous macrophages polarized to M1 phenotype for high mobility and treated to induce transient phagosome maturation arrest. In addition, we propose a significant shift of existing paradigms in the study of host-microbe interactions, in order to study microbial host immune evasion and dissemination patterns for their therapeutic utilization in the context of drug delivery. We describe a system in which microbial strategies may be adopted to facilitate absolute control over drug delivery, and without sacrificing the host carrier cells. We provide a comprehensive summary of the lessons we can learn from microbes in the context of drug delivery and discuss their feasibility for in vivo therapeutic application. We then describe our proposed "synthetic microbe drug delivery system" in detail. In our opinion, this multidisciplinary approach may hold the solution to effective, controlled drug delivery.
Collapse
Affiliation(s)
- Johan Georg Visser
- Department of Physiological Sciences, Stellenbosch University, Matieland, South Africa
| | | | - Carine Smith
- Department of Physiological Sciences, Stellenbosch University, Matieland, South Africa
| |
Collapse
|
20
|
Xu Z, Xia A, Li X, Zhu Z, Shen Y, Jin S, Lan T, Xie Y, Wu H, Meng C, Sun L, Yin Y, Chen X, Jiao X. Rapid loss of early antigen-presenting activity of lymph node dendritic cells against Ag85A protein following Mycobacterium bovis BCG infection. BMC Immunol 2018; 19:19. [PMID: 29940854 PMCID: PMC6019797 DOI: 10.1186/s12865-018-0258-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 06/18/2018] [Indexed: 02/07/2023] Open
Abstract
Background Control of Mycobacterium tuberculosis (Mtb) infection requires CD4+ T-cell responses and major histocompatibility complex class II (MHC II) presentation of Mtb antigens (Ags). Dendritic cells (DCs) are the most potent of the Ag-presenting cells and are central to the initiation of T-cell immune responses. Much research has indicated that DCs play an important role in anti-mycobacterial immune responses at early infection time points, but the kinetics of Ag presentation by these cells during these events are incompletely understood. Results In the present study, we evaluated in vivo dynamics of early Ag presentation by murine lymph-node (LN) DCs in response to Mycobacterium bovis bacillus Calmette–Guérin (BCG) Ag85A protein. Results showed that the early Ag-presenting activity of murine DCs induced by M. bovis BCG Ag85A protein in vivo was transient, appearing at 4 h and being barely detectable at 72 h. The transcription levels of CIITA, MHC II and the expression of MHC II molecule on the cell surface increased following BCG infection. Moreover, BCG was found to survive within the inguinal LN DC pool, representing a continuing source of mycobacterial Ag85A protein, with which LN DCs formed Ag85A peptide-MHCII complexes in vivo. Conclusions Our results demonstrate that a decrease in Ag85A peptide production as a result of the inhibition of Ag processing to is largely responsible for the short duration of Ag presentation by LN DCs during BCG infection in vivo.
Collapse
Affiliation(s)
- Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Aihong Xia
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Xin Li
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Zhaocheng Zhu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Yechi Shen
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA of China, Yangzhou University, Yangzhou, China
| | - Shanshan Jin
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA of China, Yangzhou University, Yangzhou, China
| | - Tian Lan
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA of China, Yangzhou University, Yangzhou, China
| | - Yuqing Xie
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA of China, Yangzhou University, Yangzhou, China
| | - Han Wu
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA of China, Yangzhou University, Yangzhou, China
| | - Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Lin Sun
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China
| | - Yuelan Yin
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA of China, Yangzhou University, Yangzhou, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, No. 48 Wenhui East Road, Yangzhou, 225009, Jiangsu, China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| | - Xinan Jiao
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, MOA of China, Yangzhou University, Yangzhou, China. .,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| |
Collapse
|
21
|
Talukder MAS, Balboula AZ, Shirozu T, Kim SW, Kunii H, Suzuki T, Ito T, Kimura K, Takahashi M. Activation of lysosomal cathepsins in pregnant bovine leukocytes. Reproduction 2018; 155:515-528. [PMID: 29626104 DOI: 10.1530/rep-18-0078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 04/06/2018] [Indexed: 11/08/2022]
Abstract
In ruminants, interferon-tau (IFNT)-mediated expression of interferon-stimulated genes in peripheral blood leukocytes (PBLs) can indicate pregnancy. Recently, type 1 IFN-mediated activation of lysosomes and lysosomal cathepsins (CTSs) was observed in immune cells. This study investigated the status of lysosomal CTSs and lysosomes in PBLs collected from pregnant (P) and non-pregnant (NP) dairy cows, and conducted in vitro IFNT stimulation of NP blood leukocytes. Blood samples were collected 0, 7, 14 and 18 days post-artificial insemination, and the peripheral blood mononuclear cells (PBMCs) and polymorphonuclear granulocytes (PMNs) separated. The fluorescent activity of CTSB and CTSK in PMNs significantly increased with the progress of pregnancy, especially on day 18. In vitro supplementation of IFNT significantly increased the activities of CTSB and CTSK in NP PBMCs and PMNs. CTSB expression was significantly higher in PBMCs and PMNs collected from P day-18 cows than from NP cows, whereas there was no difference in CTSK expression. IFNT increased CTSB expression but did not affect CTSK expression. Immunodetection showed an increase of CTSB in P day-18 PBMCs and PMNs. In vitro stimulation of IFNT increased CTSB in NP PBMCs and PMNs. Lysosomal acidification showed a significant increase in P day-18 PBMCs and PMNs. IFNT also stimulated lysosomal acidification. Expressions of lysosome-associated membrane protein (LAMP) 1 and LAMP2 were significantly higher in P day-18 PBMCs and PMNs. The results suggest that pregnancy-specific activation of lysosomal functions by CTS activation in blood leukocytes is highly associated with IFNT during maternal and fetal recognition of pregnancy.
Collapse
Affiliation(s)
- Md Abdus Shabur Talukder
- Laboratory of Animal Genetics and ReproductionResearch Faculty of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Ahmed Zaky Balboula
- Department of TheriogenologyFaculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Takahiro Shirozu
- Laboratory of Animal Genetics and ReproductionResearch Faculty of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Sung Woo Kim
- Animal Genetic Resources Research CenterNational Institute of Animal Science, Namwon, Korea
| | - Hiroki Kunii
- Laboratory of Animal Genetics and ReproductionResearch Faculty of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Toshiyuki Suzuki
- Laboratory of Animal Genetics and ReproductionResearch Faculty of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Tsukino Ito
- Laboratory of Animal Genetics and ReproductionResearch Faculty of Agriculture, Hokkaido University, Hokkaido, Japan
| | - Koji Kimura
- Graduate School of Environmental and Life ScienceOkayama University, Okayama, Japan
| | - Masashi Takahashi
- Laboratory of Animal Genetics and ReproductionResearch Faculty of Agriculture, Hokkaido University, Hokkaido, Japan .,Global Station for FoodLand and Water Resources, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo, Japan
| |
Collapse
|
22
|
Chuang YM, Pinn ML, Karakousis PC, Hung CF. Intranasal Immunization with DnaK Protein Induces Protective Mucosal Immunity against Tuberculosis in CD4-Depleted Mice. Front Cell Infect Microbiol 2018; 8:31. [PMID: 29473022 PMCID: PMC5809501 DOI: 10.3389/fcimb.2018.00031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 01/23/2018] [Indexed: 01/21/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) remains a global health challenge due to the limited efficacy of the Mtb vaccine in current use, Bacillus Calmette-Guérin (BCG). To date, there is no available vaccine for immunocompromised individuals. Thus, there is an urgent need to develop a new vaccine candidate which can induce mucosal immunity in hosts with different immune statuses. DnaK (HSP70) has been shown to induce protective immunity against Mtb infection when administered by DNA vaccine; however, the protection is inferior to that induced by the BCG vaccine. In our study, we vaccinated C57BL/6J mice with DnaK protein alone. Subcutaneous or intranasal vaccination with DnaK generated IFNγ-secreting CD4+ T cells in the spleen, but only intranasal vaccination generated IL-17-releasing CD4+ T cells in the lungs, even when circulating CD4+ T cells were diminished. Furthermore, intranasal vaccination with DnaK generated tissue resident CD4+ T cells in the lungs. Vaccination with DnaK alone resulted in protective immunity comparable to BCG vaccination against tuberculosis in mice. Our results demonstrate that intranasal vaccination with DnaK can generate mucosal immunity in immunocompromised or immunocompetent mice and DnaK vaccination can generate protection against Mtb similar to BCG, underscoring its potential utility as an Mtb vaccine candidate in humans.
Collapse
Affiliation(s)
- Yu-Min Chuang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael L Pinn
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Petros C Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
23
|
Pires D, Bernard EM, Pombo JP, Carmo N, Fialho C, Gutierrez MG, Bettencourt P, Anes E. Mycobacterium tuberculosis Modulates miR-106b-5p to Control Cathepsin S Expression Resulting in Higher Pathogen Survival and Poor T-Cell Activation. Front Immunol 2017; 8:1819. [PMID: 29326705 PMCID: PMC5741618 DOI: 10.3389/fimmu.2017.01819] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/04/2017] [Indexed: 01/20/2023] Open
Abstract
The success of tuberculosis (TB) bacillus, Mycobacterium tuberculosis (Mtb), relies on the ability to survive in host cells and escape to immune surveillance and activation. We recently demonstrated that Mtb manipulation of host lysosomal cathepsins in macrophages leads to decreased enzymatic activity and pathogen survival. In addition, while searching for microRNAs (miRNAs) involved in posttranscriptional gene regulation during mycobacteria infection of human macrophages, we found that selected miRNAs such as miR-106b-5p were specifically upregulated by pathogenic mycobacteria. Here, we show that miR-106b-5p is actively manipulated by Mtb to ensure its survival in macrophages. Using an in silico prediction approach, we identified miR-106b-5p with a potential binding to the 3'-untranslated region of cathepsin S (CtsS) mRNA. We demonstrated by luminescence-based methods that miR-106b-5p indeed targets CTSS mRNA resulting in protein translation silencing. Moreover, miR-106b-5p gain-of-function experiments lead to a decreased CtsS expression favoring Mtb intracellular survival. By contrast, miR-106b-5p loss-of-function in infected cells was concomitant with increased CtsS expression, with significant intracellular killing of Mtb and T-cell activation. Modulation of miR-106b-5p did not impact necrosis, apoptosis or autophagy arguing that miR-106b-5p directly targeted CtsS expression as a way for Mtb to avoid exposure to degradative enzymes in the endocytic pathway. Altogether, our data suggest that manipulation of miR-106b-5p as a potential target for host-directed therapy for Mtb infection.
Collapse
Affiliation(s)
- David Pires
- Host-Pathogen Interactions Unit, Faculty of Pharmacy, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Elliott M. Bernard
- Host-Pathogen Interactions in Tuberculosis Laboratory, The Francis Crick Institute, London, United Kingdom
| | - João Palma Pombo
- Host-Pathogen Interactions Unit, Faculty of Pharmacy, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Nuno Carmo
- Host-Pathogen Interactions Unit, Faculty of Pharmacy, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Catarina Fialho
- Host-Pathogen Interactions Unit, Faculty of Pharmacy, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | | | - Paulo Bettencourt
- Host-Pathogen Interactions Unit, Faculty of Pharmacy, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| | - Elsa Anes
- Host-Pathogen Interactions Unit, Faculty of Pharmacy, Research Institute for Medicines, iMed-ULisboa, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
24
|
Thirunavukkarasu S, Plain KM, Purdie AC, Whittington RJ, de Silva K. IFN-γ fails to overcome inhibition of selected macrophage activation events in response to pathogenic mycobacteria. PLoS One 2017; 12:e0176400. [PMID: 28505170 PMCID: PMC5432162 DOI: 10.1371/journal.pone.0176400] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/10/2017] [Indexed: 01/25/2023] Open
Abstract
According to most models of mycobacterial infection, inhibition of the pro-inflammatory macrophage immune responses contributes to the persistence of bacteria. Mycobacterium avium subsp. paratuberculosis (MAP) is a highly successful pathogen in cattle and sheep and is also implicated as the causative agent of Crohn's disease in humans. Pathogenic mycobacteria such as MAP have developed multiple strategies to evade host defence mechanisms including interfering with the macrophages' capacity to respond to IFN-γ, a feature which might be lacking in non-pathogenic mycobacteria such as M. smegmatis. We hypothesized that pre-sensitisation of macrophages with the pro-inflammatory cytokine IFN-γ would help in overcoming the inhibitory effect of MAP or its antigens on macrophage inflammatory responses. Herein we have compared a series of macrophage activation parameters in response to MAP and M. smegmatis as well as mycobacterial antigens. While IFN-γ did overcome the inhibition in immune suppressive mechanisms in response to MAP antigen as well as M. smegmatis, we could not find a clear role for IFN-γ in overcoming the inhibition of macrophage inflammatory responses to the pathogenic mycobacterium, MAP. We demonstrate that suppression of macrophage defence mechanisms by pathogenic mycobacteria is unlikely to be overcome by prior sensitization with IFN-γ alone. This indicates that IFN-γ signaling pathway-independent mechanisms may exist for overcoming inhibition of macrophage effector functions in response to pathogenic mycobacteria. These findings have important implications in understanding the survival mechanisms of pathogenic mycobacteria directed towards finding better therapeutics and vaccination strategies.
Collapse
Affiliation(s)
- Shyamala Thirunavukkarasu
- The University of Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Karren M. Plain
- The University of Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Auriol C. Purdie
- The University of Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Richard J. Whittington
- The University of Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, Australia
| | - Kumudika de Silva
- The University of Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Sydney, Australia
| |
Collapse
|
25
|
Moliva JI, Turner J, Torrelles JB. Immune Responses to Bacillus Calmette-Guérin Vaccination: Why Do They Fail to Protect against Mycobacterium tuberculosis? Front Immunol 2017; 8:407. [PMID: 28424703 PMCID: PMC5380737 DOI: 10.3389/fimmu.2017.00407] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 03/22/2017] [Indexed: 12/11/2022] Open
Abstract
Mycobacterium tuberculosis (M.tb), the causative agent of tuberculosis (TB), is the current leading cause of death due to a single infectious organism. Although curable, the broad emergence of multi-, extensive-, extreme-, and total-drug resistant strains of M.tb has hindered eradication efforts of this pathogen. Furthermore, computational models predict a quarter of the world’s population is infected with M.tb in a latent state, effectively serving as the largest reservoir for any human pathogen with the ability to cause significant morbidity and mortality. The World Health Organization has prioritized new strategies for improved vaccination programs; however, the lack of understanding of mycobacterial immunity has made it difficult to develop new successful vaccines. Currently, Mycobacterium bovis bacillus Calmette–Guérin (BCG) is the only vaccine approved for use to prevent TB. BCG is highly efficacious at preventing meningeal and miliary TB, but is at best 60% effective against the development of pulmonary TB in adults and wanes as we age. In this review, we provide a detailed summary on the innate immune response of macrophages, dendritic cells, and neutrophils in response to BCG vaccination. Additionally, we discuss adaptive immune responses generated by BCG vaccination, emphasizing their specific contributions to mycobacterial immunity. The success of future vaccines against TB will directly depend on our understanding of mycobacterial immunity.
Collapse
Affiliation(s)
- Juan I Moliva
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Joanne Turner
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| | - Jordi B Torrelles
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH, USA.,Center for Microbial Interface Biology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
26
|
Lau A, Singh V, Soualhine H, Hmama Z. Expression of Cathepsin S in BCG converts it into a pro-apoptotic and highly immunogenic strain. Vaccine 2017; 35:2060-2068. [PMID: 28318770 DOI: 10.1016/j.vaccine.2017.02.065] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/16/2017] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND BCG vaccine, introduced almost 100years ago, is the only option to prevent TB disease. It effectively protects newborns from meningeal TB but fails to prevent adult pulmonary TB. TB kills 1.3million people annually in areas where BCG vaccination is widely practiced. Thus, more effective TB vaccines are urgently needed. Others and we have shown that BCG mimics features of virulent M. tuberculosis, in particular attenuation of essential macrophage functions such as phagosome maturation and antigen presentation. One of these studies revealed that defect in antigen presentation is largely due to down-regulation of the cysteine protease Cathepsin S (CatS), which prevents MHC II molecule maturation and proper antigen peptide loading. Recent studies also suggested a potential role for cysteine proteases in the regulation of apoptosis, a key cellular process used by the macrophage to (i) contain and process ingested bacteria and (ii) facilitate cross-talk antigen presentation between the macrophage and dendritic cells. METHOD To reverse the phenotype of vaccine-mediated macrophage attenuation, we engineered a novel BCG strain that expresses and secretes active CatS (rBCG-CatS) to examine its pro-apoptotic properties in vitro, and subsequently, immunogenicity in mice. RESULTS Transcriptomic profiling of macrophages infected with rBCG-CatS, but not BCG, revealed upregulation of key pro-apoptotic genes and downregulation of anti-apoptotic genes, which were further confirmed by RT-qPCR analyses of expression of selected genes. Macrophages infected with rBCG-CatS undergo apoptosis as indicated by increased levels of annexin V staining and intracellular caspase-3 cleavage. Consistent with these findings, mice vaccinated with rBCG-CatS showed increased antigen-specific CD4+ T-cell responses, as well as enhanced cytokine production and proliferation in CD4+ upon ex vivo re-stimulation. CONCLUSION Collectively, this study shows that a pro-apoptotic BCG strain alleviates adverse traits of the wild-type strain, resulting in a highly immunogenic TB vaccine.
Collapse
Affiliation(s)
- Alice Lau
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Vijender Singh
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Hafid Soualhine
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Zakaria Hmama
- Division of Infectious Diseases, Department of Medicine and Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC V6H 3Z6, Canada.
| |
Collapse
|
27
|
Speth MT, Repnik U, Griffiths G. Layer-by-layer nanocoating of live Bacille-Calmette-Guérin mycobacteria with poly(I:C) and chitosan enhances pro-inflammatory activation and bactericidal capacity in murine macrophages. Biomaterials 2016; 111:1-12. [PMID: 27716523 DOI: 10.1016/j.biomaterials.2016.09.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/28/2016] [Accepted: 09/30/2016] [Indexed: 12/11/2022]
Abstract
Tuberculosis (TB) is a major disease burden globally causing more than 1.5 million deaths per year. The attenuated live vaccine strain Bacille Calmette-Guérin (BCG), although providing protection against childhood TB, is largely ineffective against adult pulmonary TB. A major aim therefore is to increase the potency of the BCG vaccine to generate stronger and more sustained immunity against TB. Here, we investigated the use of layer-by-layer (LbL) nanocoating of the surface of live BCG with several layers of polyinosinic-polycytidylic acid (poly(I:C)), a strong inducer of cell-mediated immunity, and the biodegradable polysaccharide chitosan to enhance BCG immunogenicity. Nanocoating of live BCG did not affect bacterial viability or growth in vitro but induced killing of the BCG in infected mouse bone marrow-derived macrophages and enhanced macrophage production of pro-inflammatory cytokines and expression of surface co-stimulatory molecules relative to uncoated BCG. In addition, poly(I:C) surface-coated BCG, but not BCG alone or together with soluble poly(I:C), induced high production of nitric oxide (NO) and IL-12. These results argue that BCG and surface absorbed poly(I:C) act in a synergistic manner to elicit pro-inflammatory macrophage activation. In conclusion, nanocoating of live BCG with the immunostimulatory agent poly(I:C) may be an appropriate strategy to enhance and modulate host responses to the BCG vaccine.
Collapse
Affiliation(s)
- Martin Tobias Speth
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway.
| |
Collapse
|
28
|
Martin C, Espaillat MP, Santiago-Schwarz F. IL-10 restricts dendritic cell (DC) growth at the monocyte-to-monocyte-derived DC interface by disrupting anti-apoptotic and cytoprotective autophagic molecular machinery. Immunol Res 2016; 63:131-43. [PMID: 26395023 DOI: 10.1007/s12026-015-8700-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
An evolving premise is that cytoprotective autophagy responses are essential to monocyte-macrophage differentiation. Whether autophagy functions similarly during the monocyte-to-dendritic cell (DC) transition is unclear. IL-10, which induces apoptosis in maturing human DCs, has been shown to inhibit starvation-induced autophagy in murine macrophage cell lines. Based on the strict requirement that Bcl-2-mediated anti-apoptotic processes are implemented during the monocyte-to-DC transition, we hypothesized that cytoprotective autophagy responses also operate at the monocyte-DC interface and that IL-10 inhibits both anti-apoptotic and cytoprotective autophagy responses at this critical juncture. In support of our premise, we show that levels of anti-apoptotic Bcl-2 and autophagy-associated LC3 and Beclin-1 proteins are coincidentally upregulated during the monocyte-to-DC transition. Autophagy was substantiated by increased autophagosome visualization after bafilomycin treatment. Moreover, the autophagy inhibitor 3-MA restricted DC differentiation by prompting apoptosis. IL-10 implemented apoptosis that was coincidentally associated with reduced levels of Bcl-2 and widespread disruption of the autophagic flux. During peak apoptosis, IL-10 produced the death of newly committed DCs. However, cells surviving the IL-10 apoptotic schedule were highly phagocytic macrophage-like cells displaying reduced capacity to stimulate allogeneic naïve T cells in a mixed leukocyte reaction, increased levels of LC3, and mature autophagosomes. Thus, IL-10's negative control of DC-driven adaptive immunity at the monocyte-DC interface includes disruption of coordinately regulated molecular networks involved in pro-survival autophagy and anti-apoptotic responses.
Collapse
Affiliation(s)
- Carla Martin
- Biology Department, Farmingdale State College, State University of New York, Farmingdale, NY, 11735, USA
| | - Mel Pilar Espaillat
- Molecular Genetics and Microbiology and Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY, 11794, USA
| | - Frances Santiago-Schwarz
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
29
|
Xu J, Han R, Kim DW, Mo JH, Jin Y, Rha KS, Kim YM. Role of Interleukin-10 on Nasal Polypogenesis in Patients with Chronic Rhinosinusitis with Nasal Polyps. PLoS One 2016; 11:e0161013. [PMID: 27584662 PMCID: PMC5008817 DOI: 10.1371/journal.pone.0161013] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/28/2016] [Indexed: 01/28/2023] Open
Abstract
Background and Objectives Interleukin 10 (IL-10) is a potent anti-inflammatory cytokine. The dysregulation of IL-10 is associated with an enhanced immunopathologic response to infection, as well as with an increased risk for developing numerous autoimmune diseases. In this study, we investigated IL-10 expression in chronic rhinosinusitis with nasal polyps (CRSwNP) and assessed the possible role of IL-10 in the pathogenesis of CRSwNP. Materials and Methods Thirty-five patients with CRSwNP, 12 patients with chronic rhinosinusitis without NP (CRSsNP) and 10 control subjects were enrolled in this study. NP tissues and uncinated tissues (UT) were collected for analysis. Dispersed NP cells (DNPCs) were cultured in the presence or absence of IL-25 and IL-10, and a flow cytometric assay was performed to identify the constitutive cell populations of the DNPCs. Murine NP (n = 18) models were used for the in vivo experiments. Real-time PCR, immunohistochemistry, western blotting analysis and ELISA were performed to measure the expression levels of the selected inflammatory cytokines and inflammation-associated molecules. Results The mRNA expression levels of IL-10, IL-5, IL-17A, IL-25 and interferon gamma (IFN-γ) were significantly higher in the NP tissues than in the UT tissues. Strong positive correlations were observed between IL-10 and a variety of inflammatory cytokines (IL-5, IL-17A, IL-25, IFN-γ) and inflammation-associated molecules (B-cell activating factor; BAFF, CD19). Other than the IL-25 to IL-10 ratio, the expression ratios of the other measured inflammatory cytokines to IL-10 were significantly lower in the CRSwNP group than in the CRSsNP or control groups. Administrating IL-25 into the cultured DNPCs significantly increased the production of IL-10, but administrating IL-10 had no effect on the production of IL-25. Conclusion Increased expression of IL-10, IL-10 related inflammatory cytokine, and IL-10 related B cell activation indicated that IL-10, a potent anti-inflammatory cytokine, has a pivotal role in the pathogenesis of CRSwNPs.
Collapse
Affiliation(s)
- Jun Xu
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Yanbian University Hospital, Yanji, China
| | - Ruining Han
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
- Department of Otorhinolaryngology-Head and Neck Surgery, Yanbian University Hospital, Yanji, China
| | - Dae Woo Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea
| | - Ji-Hun Mo
- Department of Otorhinolaryngology-Head and Neck Surgery, Dankook University College of Medicine, Chonan, Korea
| | - Yongde Jin
- Department of Otorhinolaryngology-Head and Neck Surgery, Yanbian University Hospital, Yanji, China
| | - Ki-Sang Rha
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
| | - Yong Min Kim
- Department of Otorhinolaryngology-Head and Neck Surgery, Research Institute for Medical Science, Chungnam National University School of Medicine, Daejeon, Korea
- * E-mail:
| |
Collapse
|
30
|
Abstract
Through thousands of years of reciprocal coevolution, Mycobacterium tuberculosis has become one of humanity's most successful pathogens, acquiring the ability to establish latent or progressive infection and persist even in the presence of a fully functioning immune system. The ability of M. tuberculosis to avoid immune-mediated clearance is likely to reflect a highly evolved and coordinated program of immune evasion strategies that interfere with both innate and adaptive immunity. These include the manipulation of their phagosomal environment within host macrophages, the selective avoidance or engagement of pattern recognition receptors, modulation of host cytokine production, and the manipulation of antigen presentation to prevent or alter the quality of T-cell responses. In this article we review an extensive array of published studies that have begun to unravel the sophisticated program of specific mechanisms that enable M. tuberculosis and other pathogenic mycobacteria to persist and replicate in the face of considerable immunological pressure from their hosts. Unraveling the mechanisms by which M. tuberculosis evades or modulates host immune function is likely to be of major importance for the development of more effective new vaccines and targeted immunotherapy against tuberculosis.
Collapse
|
31
|
Hmama Z, Peña-Díaz S, Joseph S, Av-Gay Y. Immunoevasion and immunosuppression of the macrophage by Mycobacterium tuberculosis. Immunol Rev 2015; 264:220-32. [PMID: 25703562 DOI: 10.1111/imr.12268] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
By virtue of their position at the crossroads between the innate and adaptive immune response, macrophages play an essential role in the control of bacterial infections. Paradoxically, macrophages serve as the natural habitat to Mycobacterium tuberculosis (Mtb). Mtb subverts the macrophage's mechanisms of intracellular killing and antigen presentation, leading ultimately to the development of tuberculosis (TB) disease. Here, we describe mechanisms of Mtb uptake by the macrophage and address key macrophage functions that are targeted by Mtb-specific effector molecules enabling this pathogen to circumvent host immune response. The macrophage functions described in this review include fusion between phagosomes and lysosomes, production of reactive oxygen and nitrogen species, antigen presentation and major histocompatibility complex class II expression and trafficking, as well as autophagy and apoptosis. All these are Mtb-targeted key cellular pathways, normally working in concert in the macrophage to recognize, respond, and activate 'proper' immune responses. We further analyze and discuss major molecular interactions between Mtb virulence factors and key macrophage proteins and provide implications for vaccine and drug development.
Collapse
Affiliation(s)
- Zakaria Hmama
- Department of Medicine, Division of Infectious Diseases, Infection and Immunity Research Center, University of British Columbia, Vancouver, BC, Canada
| | | | | | | |
Collapse
|
32
|
Immunomodulation and Disease Tolerance to Staphylococcus aureus. Pathogens 2015; 4:793-815. [PMID: 26580658 PMCID: PMC4693165 DOI: 10.3390/pathogens4040793] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 11/10/2015] [Indexed: 12/12/2022] Open
Abstract
The Gram-positive bacterium Staphylococcus aureus is one of the most frequent pathogens that causes severe morbidity and mortality throughout the world. S. aureus can infect skin and soft tissues or become invasive leading to diseases such as pneumonia, endocarditis, sepsis or toxic shock syndrome. In contrast, S. aureus is also a common commensal microbe and is often part of the human nasal microbiome without causing any apparent disease. In this review, we explore the immunomodulation and disease tolerance mechanisms that promote commensalism to S. aureus.
Collapse
|
33
|
The role of IL-10 in microbiome-associated immune modulation and disease tolerance. Cytokine 2015; 75:291-301. [DOI: 10.1016/j.cyto.2014.11.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/20/2014] [Accepted: 11/21/2014] [Indexed: 02/06/2023]
|
34
|
Kumar P, John V, Marathe S, Das G, Bhaskar S. Mycobacterium indicus pranii induces dendritic cell activation, survival, and Th1/Th17 polarization potential in a TLR-dependent manner. J Leukoc Biol 2015; 97:511-20. [PMID: 25593326 DOI: 10.1189/jlb.1a0714-361r] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
MIP is a nonpathogenic, soil-borne predecessor of Mycobacterium avium. It has been reported previously that MIP possesses strong immunomodulatory properties and confers protection against experimental TB and tumor. DCs, by virtue of their unmatched antigen-presentation potential, play a critical role in activation of antitumor and antimycobacterial immune response. The effect of MIP on the behavior of DCs and the underlying mechanisms, however, have not been investigated so far. In the present study, we showed that MIP induces significant secretion of IL-6, IL-12p40, IL-10, and TNF-α by DCs and up-regulates the expression of costimulatory molecules CD40, CD80, and CD86. MIP(L) induced a significantly higher response compared with MIP(K). PI and Annexin V staining showed that MIP increases DC survival by inhibiting apoptosis. Consistently, higher expression of antiapoptotic proteins Bcl-2 and Bcl-xl was observed in MIP-stimulated DCs. Cytokines, produced by naïve T cells, cocultured with MIP-stimulated DCs, showed that MIP promotes Th1/Th17 polarization potential in DCs. Response to MIP was lost in MyD88(-/-)DCs, underscoring the critical role of TLRs in MIP-induced DC activation. Further studies revealed that TLR2 and TLR9 are involved in DC activation by MIP(L), whereas MIP(K) activates the DCs through TLR2. Our findings establish the DC activation by MIP, define the behavior of MIP-stimulated DCs, and highlight the role of TLRs in MIP-induced DC activation.
Collapse
Affiliation(s)
- Pawan Kumar
- *National Institute of Immunology, New Delhi, India; and International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Vini John
- *National Institute of Immunology, New Delhi, India; and International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Soumitra Marathe
- *National Institute of Immunology, New Delhi, India; and International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Gobardhan Das
- *National Institute of Immunology, New Delhi, India; and International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Sangeeta Bhaskar
- *National Institute of Immunology, New Delhi, India; and International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| |
Collapse
|
35
|
Rottenberg ME, Carow B. SOCS3 and STAT3, major controllers of the outcome of infection with Mycobacterium tuberculosis. Semin Immunol 2014; 26:518-32. [DOI: 10.1016/j.smim.2014.10.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 10/06/2014] [Accepted: 10/07/2014] [Indexed: 01/04/2023]
|
36
|
Rajaram MVS, Ni B, Dodd CE, Schlesinger LS. Macrophage immunoregulatory pathways in tuberculosis. Semin Immunol 2014; 26:471-85. [PMID: 25453226 DOI: 10.1016/j.smim.2014.09.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 09/25/2014] [Accepted: 09/26/2014] [Indexed: 12/17/2022]
Abstract
Macrophages, the major host cells harboring Mycobacterium tuberculosis (M.tb), are a heterogeneous cell type depending on their tissue of origin and host they are derived from. Significant discord in macrophage responses to M.tb exists due to differences in M.tb strains and the various types of macrophages used to study tuberculosis (TB). This review will summarize current concepts regarding macrophage responses to M.tb infection, while pointing out relevant differences in experimental outcomes due to the use of divergent model systems. A brief description of the lung environment is included since there is increasing evidence that the alveolar macrophage (AM) has immunoregulatory properties that can delay optimal protective host immune responses. In this context, this review focuses on selected macrophage immunoregulatory pattern recognition receptors (PRRs), cytokines, negative regulators of inflammation, lipid mediators and microRNAs (miRNAs).
Collapse
Affiliation(s)
- Murugesan V S Rajaram
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Ni
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
| | - Claire E Dodd
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Larry S Schlesinger
- Center for Microbial Interface Biology, Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
37
|
Pulmonary M. tuberculosis infection delays Th1 immunity via immunoadaptor DAP12-regulated IRAK-M and IL-10 expression in antigen-presenting cells. Mucosal Immunol 2014; 7:670-83. [PMID: 24172845 DOI: 10.1038/mi.2013.86] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 09/04/2013] [Accepted: 09/23/2013] [Indexed: 02/04/2023]
Abstract
Interaction of mycobacteria with the host leads to retarded expression of T helper cell type 1 (Th1) immunity in the lung. However, the immune mechanisms remain poorly understood. Using in vivo and in vitro models of Mycobacterium tuberculosis (M. tb) infection, we find the immunoadaptor DAP12 (DNAX-activating protein of 12 kDa) in antigen-presenting cells (APCs) to be critically involved in this process. Upon infection of APCs, DAP12 is required for IRAK-M (interleukin-1 receptor-associated kinase M) expression, which in turn induces interleukin-10 (IL-10) and an immune-suppressed phenotype of APCs, thus leading to suppressed Th1 cell activation. Lack of DAP12 reduces APC IL-10 production and increases their Th1 cell-activating capability, resulting in expedited Th1 responses and enhanced protection. On the other hand, adoptively transferred DAP12-competent APCs suppress Th1 cell activation within DAP12-deficient hosts, and blockade of IL-10 aborts the ability of DAP12-competent APCs to suppress Th1 activation. Our study identifies the DAP12/IRAK-M/IL-10 to be a novel molecular pathway in APCs exploited by mycobacterial pathogens, allowing infection a foothold in the lung.
Collapse
|
38
|
Eisenhut M. Enhanced innate immunity as explanation for reduced Mycobacterium tuberculosis infection in Bacillus Calmette-Guérin-immunized children. Am J Respir Crit Care Med 2013; 188:257-8. [PMID: 23855702 DOI: 10.1164/rccm.201301-0060le] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
39
|
Srivastava S, Ernst JD. Cutting edge: Direct recognition of infected cells by CD4 T cells is required for control of intracellular Mycobacterium tuberculosis in vivo. THE JOURNAL OF IMMUNOLOGY 2013; 191:1016-20. [PMID: 23817429 DOI: 10.4049/jimmunol.1301236] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Effector T cells control intracellular infection by secreting cytokines and through contact-dependent cytolysis. Because cytokines can diffuse and act at a distance, we determined whether cytokine diffusion is sufficient to control Mycobacterium tuberculosis or whether direct recognition of infected cells by CD4 T cells is required. Using MHC class II (MHC II) mixed bone marrow chimeras, we compared the bacterial burdens in lung myeloid cells that were capable (MHC II(+/+)) or not (MHC II(-/-)) of being recognized by CD4 T cells. MHC II(+/+) cells had lower bacterial burdens than did MHC II(-/-) cells. CD4 T cell depletion increased the number of bacteria associated with MHC II(+/+)cells but not MHC II(-/-) cells, indicating that direct recognition of infected cells by CD4 T cells is required for control of intracellular M. tuberculosis. These results show that the effector mechanisms required for CD4 T cell control of distinct intracellular pathogens differ and that long-range cytokine diffusion does not contribute to control of M. tuberculosis.
Collapse
Affiliation(s)
- Smita Srivastava
- Division of Infectious Diseases, Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
40
|
Fol M, Głobińska A, Stączek P, Kowalewicz-Kulbat M, Druszczyńska M, Madiraju MVVS, Rudnicka W. The lack of L-PG production and the repercussions of it in regards to M. Tuberculosis interactions with mononuclear phagocytes. Acta Microbiol Immunol Hung 2013; 60:127-44. [PMID: 23827745 DOI: 10.1556/amicr.60.2013.2.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The lysine connection with phosphatidylglycerol (PG) alters the M. tuberculosis(Mtb) surface charge, and consequently it may decrease the bacterial vulnerability to antimicrobial action of the immune cells. The aim of the study was to assess the significance of PG lysinylation in the Mtb interactions with mononuclear phagocytes. Both the Mtb strain with deletion of lysX gene (Mtb-lysX) which is responsible for PG lysinylation as well as the complemented strain (Mtb-compl) was used to infect human blood monocytes or THP-1 cells. The monocytes were obtained by MACS technique, or THP-1 cells. The Mtb-lysX strain has exhibited the enhanced sensitivity to HNP 1-3. However, it was not susceptible to bactericidal action of cathepsin G. The LysX deletion did not influence the Mtb ability of monocyte induction to IL-10 secretion. The intra- and extracellular expression of MHC-II was similarly reduced after the Mtb-lysX or Mtb-Rv infections. Noticeably significant is that the Mtb strain with deleted lysX has not affected the intensity of the gene expression of cathepsin G compared to the uninfected monocytes. That is the clear contrast to what the Mtb-Rv strain has proved. The obtained results suggest that the Mtb ability to lysinylate PG is a participatory element in mycobacterial strategy of survival inside phagocytic cells. However, the extended studies are needed to determine its influence on the other immune cells and define its role in the developing of Mtb infection.
Collapse
Affiliation(s)
- Marek Fol
- University of Lodz, Department of Immunology and Infectious Biology, Lodz, Poland.
| | | | | | | | | | | | | |
Collapse
|
41
|
Interactions between an M. tuberculosis strain overexpressing mtrA and mononuclear phagocytes. Adv Med Sci 2013; 58:172-83. [PMID: 23640943 DOI: 10.2478/v10039-012-0058-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
PURPOSE It was previously shown that the bacterial two-component regulatory signal transduction (2CR) system MtrAB may be associated with the ability of M. tuberculosis (Mtb) to survive in macrophages. In the present work Mtb mutants: Rv-78 with overexpression of mtrA and Rv-129 with elevated level of phosphorylation-defective MtrA were used for further investigation of the potential influence of the MtrAB system on Mtb interaction with human monocytes. MATERIAL/METHODS Flow cytometry was used to determine the expression of MHC class II molecules. The expression of genes for inducible nitric oxide synthase (iNOS) and cathepsin G was quantified by RT-PCR. The association of Mtb strains with Rab5 and Rab7 positive vacuoles was investigated applying confocal microscopy. IL-10 and IL-12 secretion by monocytes as well as the Mtb susceptibility to cathepsin G were investigated. RESULTS Mutation-carried and wild type Mtb strains inhibited MHC class II expression on monocytes to a similar extent. Monocyte stimulation with mycobacteria led to the increased production of IL-10 but no detectable amounts of IL-12 or NO were observed. Expression of the gene for iNOS was not detected while that for cathepsin G was shown, however its intensity was not associated with MtrA mutation. Mtb mutant strains were more effectively enclosed in phagosomes containing the late endosome marker Rab7 as compared to the control. CONCLUSIONS The results may confirm the importance of the MtrAB system in mycobacterial capacity for successful survival in phagocytes, especially in the context of high degree of colocalization of Mtb Rv-78 to mature phagosomes.
Collapse
|
42
|
Mariotti S, Sargentini V, Pardini M, Giannoni F, De Spirito M, Gagliardi MC, Greco E, Teloni R, Fraziano M, Nisini R. Mycobacterium tuberculosis may escape helper T cell recognition by infecting human fibroblasts. Hum Immunol 2013; 74:722-9. [PMID: 23459076 DOI: 10.1016/j.humimm.2013.02.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 01/29/2013] [Accepted: 02/19/2013] [Indexed: 11/16/2022]
Abstract
The host immune response can limit Mycobacterium tuberculosis (Mtb) spreading in primary tuberculosis (TB) without eradicating all bacilli, which can persist causing latent TB infection and are responsible for reactivation TB. Persistent Mtb is confined to granulomas within phagocytes, but it is also found in other non-immune cells. We focused on fibroblasts since these cells participate to the granuloma formation and were shown to be infected in latent TB infections. We show that in vitro both Mtb and Bacille Calmette-Guérin actively replicate in human fibroblasts. Mycobacterial infection of fibroblasts causes a significant inhibition of interferon (IFN)-γ induced membrane expression of major histocompatibility complex class II molecules in these cells. The functional consequence of in vitro infection is a significant reduction of the fibroblast capacity to present peptides and soluble proteins to autologous specific CD4(+) T cell clones. Moreover, fibroblasts are capable of presenting antigen derived from the processing of heat-killed Mtb, but not from viable Mtb. Data indicate that IFN-γ treated fibroblasts are capable of presenting antigens derived from the processing of whole bacteria in addition to the capacity to present peptides and isolated proteins. Interestingly, Mtb infected fibroblasts lose this capacity, suggesting that Mtb may evade T helper immune surveillance by infecting fibroblasts.
Collapse
Affiliation(s)
- Sabrina Mariotti
- Dipartimento di Malattie Infettive, Parassitarie e Immunomediate, Istituto Superiore di Sanità, Roma, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Schistosoma japonicum soluble egg antigens attenuate IFN-γ-induced MHC class II expression in RAW 264.7 macrophages. PLoS One 2012; 7:e49234. [PMID: 23152879 PMCID: PMC3496699 DOI: 10.1371/journal.pone.0049234] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2012] [Accepted: 10/07/2012] [Indexed: 11/19/2022] Open
Abstract
Innate immune response plays the key role in initiating and guiding the immune response. Elucidating the innate immune related molecular events involved in the interaction between the parasite and the host will aid in the development of an effective vaccine and anti-schistosome pharmaceuticals. In this study, we examined the regulatory effect of Schistosoma japonicum soluble egg antigen (SEA) on MHC class II expression in macrophage cell line RAW 264.7. We demonstrated that SEA possesses the ability to down-regulate IFN-γ-induced MHC class II expression in RAW 264.7 cells. The production of IL-10 and IL-6 in RAW 264.7 cells, induced by SEA, was responsible for mediating the down-regulation of MHC class II. Our findings suggest that in RAW 264.7 cells (1) IFN-γ provides a condition for lower concentrations of SEA to attenuate MHC class II expression; (2) SEA attenuated IFN-γ-induced MHC class II expression and the IL-10 and IL-6 production is mediated at least partly by the interaction of SEA with TLR4; and (3) SEA attenuated IFN-γ-induced MHC class II expression at the transcriptional level.
Collapse
|
44
|
Gowthaman U, Rai PK, Khan N, Jackson DC, Agrewala JN. Lipidated promiscuous peptides vaccine for tuberculosis-endemic regions. Trends Mol Med 2012; 18:607-14. [PMID: 22939171 DOI: 10.1016/j.molmed.2012.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2012] [Revised: 07/10/2012] [Accepted: 07/20/2012] [Indexed: 10/27/2022]
Abstract
Despite nine decades of Bacillus Calmette--Guérin (BCG) vaccination, tuberculosis continues to be a major global health challenge. Clinical trials worldwide have proved the inadequacy of the BCG vaccine in preventing the manifestation of pulmonary tuberculosis in adults. Ironically, the efficacy of BCG is poorest in tuberculosis endemic areas. Factors such as nontuberculous or environmental mycobacteria and helminth infestation have been suggested to limit the efficacy of BCG. Hence, in high TB-burden countries, radically novel strategies of vaccination are urgently required. Here we showcase the properties of lipidated promiscuous peptide vaccines that target and activate cells of the innate and adaptive immune systems by employing a Toll-like receptor-2 agonist, S-[2,3-bis(palmitoyloxy)propyl]cysteine (Pam2Cys). Such a strategy elicits robust protection and enduring memory responses by type 1 T helper cells (Th1). Consequently, lipidated peptides may yield a better vaccine than BCG.
Collapse
Affiliation(s)
- Uthaman Gowthaman
- Immunology Laboratory, CSIR-Institute of Microbial Technology, Chandigarh-160036, India
| | | | | | | | | |
Collapse
|
45
|
Balasubramanian SA, Mohan S, Pye DC, Willcox MDP. Proteases, proteolysis and inflammatory molecules in the tears of people with keratoconus. Acta Ophthalmol 2012; 90:e303-9. [PMID: 22413749 DOI: 10.1111/j.1755-3768.2011.02369.x] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE To investigate the expression of proteases, proteolytic activity and cytokines in the tear film of people with keratoconus. METHODS Basal tears from people with keratoconus, from individuals who had undergone corneal collagen cross-linking for the treatment of keratoconus, and from normal controls were collected using a capillary tube. Corneal curvature of each subject was mapped. The total protein in tears was estimated. Levels and activity of proteases in the tears were analysed using specific antibody arrays and activity assays. RESULTS The total tear protein level was significantly reduced in keratoconus (4.1 ± 0.9 mg/ml) compared with normals (6.7 ± 1.4 mg/ml) (p < 0.0001) or subjects who had undergone corneal collagen cross-linking (5.7 ± 2.3 mg/ml) (p < 0.005). Significantly (p < 0.05) increased tear expression of matrix metalloproteinases (MMP) -1, -3, -7, -13, interleukins (IL) -4, -5, -6, -8 and tumour necrosis factor (TNF) -α, -β were evident in keratoconus. Tear IL-6 was the only cytokine significantly (p < 0.05) increased in tears of keratoconus subjects compared with the collagen cross-linked group. No significant difference in tear proteases were observed between the normal and the cross-linked groups, although the expression of TNF-α was significantly (p < 0.05) increased in the cross-linked group compared with the controls. Elevated gelatinolytic (87.5 ± 33.6 versus 45.8 ± 24.6 FIU, p < 0.0001) and collagenolytic (6.1 ± 3.2 versus 3.6 ± 2.0 FIU, p < 0.05) activities were observed in tears from keratoconus compared with normal subjects. The activity of tear gelatinases (69.6 ± 22.2 FIU) and collagenases (5.7 ± 3.3 FIU) in the collagen cross-linked group was not significantly different compared with either keratoconus or normals. CONCLUSION Tears of people with keratoconus had 1.9 times higher levels of proteolytic activity and over expression of several MMPs and cytokines compared with tears from controls. Further investigations are required to study the possible implications of these changes and whether they can be used to monitor disease progression or determine the success of corneal collagen cross-linking.
Collapse
|
46
|
Cellular and humoral mechanisms involved in the control of tuberculosis. Clin Dev Immunol 2012; 2012:193923. [PMID: 22666281 PMCID: PMC3362816 DOI: 10.1155/2012/193923] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 03/06/2012] [Accepted: 03/06/2012] [Indexed: 02/08/2023]
Abstract
Mycobacterium tuberculosis (Mtb) infection is a major international public health problem. One-third of the world's population is thought to have latent tuberculosis, a condition where individuals are infected by the intracellular bacteria without active disease but are at risk for reactivation, if their immune system fails. Here, we discuss the role of nonspecific inflammatory responses mediated by cytokines and chemokines induced by interaction of innate receptors expressed in macrophages and dendritic cells (DCs). We also review current information regarding the importance of several cytokines including IL-17/IL-23 in the development of protective cellular and antibody-mediated protective responses against Mtb and their influence in containment of the infection. Finally, in this paper, emphasis is placed on the mechanisms of failure of Mtb control, including the immune dysregulation induced by the treatment with biological drugs in different autoimmune diseases. Further functional studies, focused on the mechanisms involved in the early host-Mtb interactions and the interplay between host innate and acquired immunity against Mtb, may be helpful to improve the understanding of protective responses in the lung and in the development of novel therapeutic and prophylactic tools in TB.
Collapse
|
47
|
Moreira J, Aragão-Filho WC, Barillas SG, Barbosa SM, Pedroza LA, Condino-Neto A. Human Leucocytes Response to Viable, Extended Freeze-Drying or Heat-Killed Mycobacterium bovis bacillus Calmette-Guérin. Scand J Immunol 2011; 75:96-101. [DOI: 10.1111/j.1365-3083.2011.02632.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
48
|
Lactoferrin Augmentation of the BCG Vaccine Leads to Increased Pulmonary Integrity. Tuberc Res Treat 2011; 2011:835410. [PMID: 22567270 PMCID: PMC3335707 DOI: 10.1155/2011/835410] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 01/19/2011] [Accepted: 03/01/2011] [Indexed: 11/21/2022] Open
Abstract
The goal of vaccination to prevent tuberculosis disease (TB) is to offer long-term protection to the individual and the community. In addition, the success of any protective TB vaccine should include the ability to limit cavitary formation and disease progression. The current BCG vaccine protects against disseminated TB disease in children by promoting development of antigenic-specific responses. However, its efficacy is limited in preventing postprimary pulmonary disease in adults that is responsible for the majority of disease and transmission. This paper illustrates the use of lactoferrin as an adjuvant to boost efficacy of the BCG vaccine to control organism growth and limit severe manifestation of pulmonary disease. This resulting limitation in pathology may ultimately, limit spread of bacilli and subsequent transmission of organisms between individuals. The current literature is reviewed, and data is presented to support molecular mechanisms underlying lactoferrin's utility as an adjuvant for the BCG vaccine.
Collapse
|
49
|
Coordinated involvement of cathepsins S, D and cystatin C in the commitment of hematopoietic stem cells to dendritic cells. Int J Biochem Cell Biol 2011; 43:775-83. [PMID: 21315176 DOI: 10.1016/j.biocel.2011.02.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 01/29/2011] [Accepted: 02/01/2011] [Indexed: 02/02/2023]
Abstract
The identity of biochemical players which underpin the commitment of CD34(+) hematopoietic stem cells to immunogenic or tolerogenic dendritic cells is largely unknown. To explore this issue, we employed a previously established cell-based system amenable to shift dendritic cell differentiation from the immunogenic into the tolerogenic pathway upon supplementation with a conventional cytokine cocktail containing thrombopoietin (TPO) and IL-16. We show that stringent regulation of cathepsins S and D, two proteases involved in antigen presentation, is crucial to engage cell commitment to either route. In response to TPO+IL-16-dependent signaling, both cathepsins undergo earlier maturation and down-regulation. Additionally, cystatin C orchestrates cathepsin S expression through a tight but reversible interaction that, based on a screen of adult stem cells from disparate origins, CD14(+) cells, primary fibroblasts and the MCF7 cell line, appears unique to CD34(+) stem cells from peripheral and cord blood. As shown by CD4(+) T cell proliferation in mixed-lymphocyte reactions, cell commitment to either pathway is disrupted upon cathepsin knockdown by RNAi. Surprisingly, similar effects were also observed upon gene overexpression, which prompts atypically accelerated maturation of cathepsins S and D in cells of the immunogenic pathway, similar to the tolerogenic route. Furthermore, RNAi studies revealed that cystatin C is a proteolytic target of cathepsin D and has a direct, causal impact on cell differentiation. Together, these findings uncover a novel biochemical cluster that is subject to time-controlled and rigorously balanced expression to mediate specific stem cell commitment at the crossroads towards tolerance or immunity.
Collapse
|
50
|
O'Leary S, O'Sullivan MP, Keane J. IL-10 blocks phagosome maturation in mycobacterium tuberculosis-infected human macrophages. Am J Respir Cell Mol Biol 2010; 45:172-80. [PMID: 20889800 DOI: 10.1165/rcmb.2010-0319oc] [Citation(s) in RCA: 191] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Successful phagolysosomal maturation is an important innate immune response to intracellular infection. However, Mycobacterium tuberculosis (Mtb) can manipulate and inhibit this host response to ensure survival within its niche cell. We investigate the role of the anti-inflammatory cytokine IL-10 on Mtb-phagosome maturation. Blocking IL-10, which was secreted from Mtb-infected macrophages, allowed phagosome maturation to proceed. Macrophage cytokine gene expression profiles were not significantly altered by blocking IL-10 3 hours after infection with Mtb. We demonstrate that IL-10 can regulate this protective phenotype in phorbol myristate acetate (PMA)-treated THP-1 cells, monocyte-derived macrophages (MDMs), and human alveolar macrophages (AMs) infected with Mtb. The regulatory effect of endogenous IL-10 was evident in macrophages infected with virulent Mtb H37Rv, as well as in attenuated strains of mycobacteria. Unlike live Mtb, dead bacilli occupy a mature, acidic phagosome. However, the addition of IL-10 to cells infected with killed Mtb successfully inhibited the maturation of this compartment. Importantly, we demonstrate that the addition of IL-10 to MDMs results in enhanced mycobacterial survival and growth. Our results suggest that IL-10 exerts its effects on this early macrophage response in a partly signal transducer and activator of transcription 3 (STAT3)-dependent manner, and independent of mitogen activated protein kinase p38 (MAPKp38) and extracellular regulated kinase 1/2 (ERK1/2) activity. IL-10 is a feature of human tuberculous granuloma, and these new findings support the hypothesis that this cytokine can promote pathogen persistence by contributing to Mtb-phagosome maturation arrest in human macrophages.
Collapse
Affiliation(s)
- Seónadh O'Leary
- Respiratory Medicine, St. James's Hospital, CResT, Dublin 8, Ireland
| | | | | |
Collapse
|