1
|
Kireev FD, Lopatnikova JA, Alshevskaya AA, Sennikov SV. Role of Tumor Necrosis Factor in Tuberculosis. Biomolecules 2025; 15:709. [PMID: 40427602 PMCID: PMC12108764 DOI: 10.3390/biom15050709] [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: 03/18/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Tumor necrosis factor (TNF) is a key immunoregulatory cytokine with a dual role in the host response to Mycobacterium tuberculosis. While essential for granuloma formation, macrophage activation, and containment of latent infection, TNF can also contribute to tissue damage and immune pathology. This review systematically analyzes over 300 peer-reviewed studies published between 1980 and 2024, highlighting the molecular and cellular mechanisms of TNF action in tuberculosis (TB). Particular attention is given to TNF receptor signaling pathways, the balance between protective and pathological immune responses, and the modulation of TNF activity during anti-TNF therapy in patients with autoimmune diseases. We discuss how different TNF inhibitors vary in their capacity to interfere with host defense mechanisms, with monoclonal antibodies carrying a higher reactivation risk than receptor-based agents. To enhance conceptual clarity, we provide newly developed schematic representations that integrate current knowledge on TNF-driven immune dynamics, including its interaction with other cytokines, effects on granuloma stability, and role in intracellular bacterial control. Understanding the pleiotropic functions of TNF in tuberculosis pathogenesis is crucial for developing safe immunomodulatory strategies and optimizing the clinical management of patients at risk of latent TB reactivation.
Collapse
Affiliation(s)
- Fedor D. Kireev
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution “Research Institute of Fundamental and Clinical Immunology” (RIFCI), 630099 Novosibirsk, Russia; (F.D.K.); (J.A.L.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| | - Julia A. Lopatnikova
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution “Research Institute of Fundamental and Clinical Immunology” (RIFCI), 630099 Novosibirsk, Russia; (F.D.K.); (J.A.L.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| | - Alina A. Alshevskaya
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| | - Sergey V. Sennikov
- Laboratory of Molecular Immunology, Federal State Budgetary Scientific Institution “Research Institute of Fundamental and Clinical Immunology” (RIFCI), 630099 Novosibirsk, Russia; (F.D.K.); (J.A.L.)
- Laboratory of Immune Engineering, Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” under the Ministry of Health of the Russian Federation (Sechenov University), 119048 Moscow, Russia;
| |
Collapse
|
2
|
Ma Y, Jiang T, Zhu X, Xu Y, Wan K, Zhang T, Xie M. Efferocytosis in dendritic cells: an overlooked immunoregulatory process. Front Immunol 2024; 15:1415573. [PMID: 38835772 PMCID: PMC11148234 DOI: 10.3389/fimmu.2024.1415573] [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: 04/10/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation. While macrophages are primarily known for this task, dendritic cells (DCs) also play a significant role. This review delves into the unique contributions of various DC subsets to efferocytosis, highlighting the distinctions in how DCs and macrophages recognize and handle apoptotic cells. It further explores how efferocytosis influences DC maturation, thereby affecting immune tolerance. This underscores the pivotal role of DCs in orchestrating immune responses and sustaining immune equilibrium, providing new insights into their function in immune regulation.
Collapse
Affiliation(s)
- Yanyan Ma
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tangxing Jiang
- Department of Emergency Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xun Zhu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yizhou Xu
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ke Wan
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tingxuan Zhang
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Miaorong Xie
- Department of Emergency and Critical Care Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Mott D, Yang J, Baer C, Papavinasasundaram K, Sassetti CM, Behar SM. High Bacillary Burden and the ESX-1 Type VII Secretion System Promote MHC Class I Presentation by Mycobacterium tuberculosis-Infected Macrophages to CD8 T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1531-1542. [PMID: 37000471 PMCID: PMC10159937 DOI: 10.4049/jimmunol.2300001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/21/2023] [Indexed: 04/01/2023]
Abstract
We used a mouse model to study how Mycobacterium tuberculosis subverts host defenses to persist in macrophages despite immune pressure. CD4 T cells can recognize macrophages infected with a single bacillus in vitro. Under identical conditions, CD8 T cells inefficiently recognize infected macrophages and fail to restrict M. tuberculosis growth, although they can inhibit M. tuberculosis growth during high-burden intracellular infection. We show that high intracellular M. tuberculosis numbers cause macrophage death, leading other macrophages to scavenge cellular debris and cross-present the TB10.4 Ag to CD8 T cells. Presentation by infected macrophages requires M. tuberculosis to have a functional ESX-1 type VII secretion system. These data indicate that phagosomal membrane damage and cell death promote MHC class I presentation of the immunodominant Ag TB10.4 by macrophages. Although this mode of Ag presentation stimulates cytokine production that we presume would be host beneficial, killing of uninfected cells could worsen immunopathology. We suggest that shifting the focus of CD8 T cell recognition to uninfected macrophages would limit the interaction of CD8 T cells with infected macrophages and impair CD8 T cell-mediated resolution of tuberculosis.
Collapse
Affiliation(s)
- Daniel Mott
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Jason Yang
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Christina Baer
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Kadamba Papavinasasundaram
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Christopher M. Sassetti
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Samuel M. Behar
- Immunology and Microbiology Program, Graduate School of Biomedical Science, Worcester, Massachusetts, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
4
|
Meng C, Liu J, Kang X, Xu Z, Xu S, Li X, Pan Z, Chen X, Jiao X. Discrepancy in Response of Mouse Dendritic Cells against BCG: Weak Immune Effects of Plasmacytoid Dendritic Cells Compared to Classical Dendritic Cells despite the Uptake of Bacilli. Trop Med Infect Dis 2023; 8:tropicalmed8030140. [PMID: 36977141 PMCID: PMC10057906 DOI: 10.3390/tropicalmed8030140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/25/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Tuberculosis (TB), a zoonosis characterized by chronic respiratory infections, is mainly caused by Mycobacterium tuberculosis and is associated with one of the heaviest disease burdens in the world. Dendritic cells (DCs) play a key role and act as a bridge between innate and adaptive immune responses against TB. DCs are divided into distinct subsets. Currently, the response of DCs to mycobacterial infections is poorly understood. Herein, we aimed to evaluate the responses of splenic conventional DCs (cDC) and plasmacytoid DCs (pDC), subsets to Bacillus Calmette–Guérin (BCG) infection in mice. Splenic pDC had a significantly higher infection rate and intracellular bacterial count than cDC and the CD8+ and CD8− cDC subsets after BCG infection. However, the expression levels of CD40, CD80, CD86, and MHC-II molecules were significantly upregulated in splenic cDC and the CD8 cDC subsets compared to pDC during BCG infection. Splenic cDC had a higher expression of IFN-γ and IL-12p70 than pDC, whereas pDC had higher levels of TNF-α and MCP-1 than cDC in mice infected with BCG. At early stages of immunization with BCG containing the Ag85A protein, splenic cDC and pDC could present the Ag85A peptide to a specific T hybridoma; however, cDC had a stronger antigen presenting activity than pDC. In summary, splenic cDC and pDC extensively participate in mouse immune responses against BCG infection in vivo. Although pDC had a higher BCG uptake, cDC induced stronger immunological effects, including activation and maturation, cytokine production, and antigen presentation.
Collapse
Affiliation(s)
- Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Jun Liu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Xilong Kang
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Shuangyuan Xu
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
| | - Xin Li
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (X.C.); (X.J.)
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou 225009, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture and Rural Affairs, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Correspondence: (X.C.); (X.J.)
| |
Collapse
|
5
|
Mundra A, Yegiazaryan A, Karsian H, Alsaigh D, Bonavida V, Frame M, May N, Gargaloyan A, Abnousian A, Venketaraman V. Pathogenicity of Type I Interferons in Mycobacterium tuberculosis. Int J Mol Sci 2023; 24:3919. [PMID: 36835324 PMCID: PMC9965986 DOI: 10.3390/ijms24043919] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Tuberculosis (TB) is a leading cause of mortality due to infectious disease and rates have increased during the emergence of COVID-19, but many of the factors determining disease severity and progression remain unclear. Type I Interferons (IFNs) have diverse effector functions that regulate innate and adaptive immunity during infection with microorganisms. There is well-documented literature on type I IFNs providing host defense against viruses; however, in this review, we explore the growing body of work that indicates high levels of type I IFNs can have detrimental effects to a host fighting TB infection. We report findings that increased type I IFNs can affect alveolar macrophage and myeloid function, promote pathological neutrophil extracellular trap responses, inhibit production of protective prostaglandin 2, and promote cytosolic cyclic GMP synthase inflammation pathways, and discuss many other relevant findings.
Collapse
Affiliation(s)
- Akaash Mundra
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Aram Yegiazaryan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Haig Karsian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Dijla Alsaigh
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Victor Bonavida
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Mitchell Frame
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Nicole May
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Areg Gargaloyan
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Arbi Abnousian
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA 91768, USA
| |
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
|
Zheng N, Fleming J, Hu P, Jiao J, Zhang G, Yang R, Li C, Liu Y, Bi L, Zhang H. CD84 is a Suppressor of T and B Cell Activation during Mycobacterium tuberculosis Pathogenesis. Microbiol Spectr 2022; 10:e0155721. [PMID: 35196822 PMCID: PMC8865571 DOI: 10.1128/spectrum.01557-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 01/21/2022] [Indexed: 11/20/2022] Open
Abstract
Interest in host-directed therapies as alternatives/adjuncts to antibiotic treatment has resurged with the increasing prevalence of antibiotic-resistant tuberculosis (TB). Immunotherapies that reinvigorate immune responses by targeting immune checkpoints like PD-1/PD-L1 have proved successful in cancer therapy. Immune cell inhibitory receptors that trigger Mycobacterium tuberculosis-specific immunosuppression, however, are unknown. Here, we show that the levels of CD84, a SLAM family receptor, increase in T and B cells in lung tissues from M. tuberculosis-infected C57BL/6 mice and in peripheral blood mononuclear cells (PBMCs) from pulmonary TB patients. M. tuberculosis challenge experiments using CD84-deficient C57BL/6 mice suggest that CD84 expression likely leads to T and B cell immunosuppression during M. tuberculosis pathogenesis and also plays an inhibitory role in B cell activation. Importantly, CD84-deficient mice showed improved M. tuberculosis clearance and longer survival than M. tuberculosis-infected wild-type (WT) mice. That CD84 is a putative M. tuberculosis infection-specific inhibitory receptor suggests it may be a suitable target for the development of TB-specific checkpoint immunotherapies. IMPORTANCE Immune checkpoint therapies, such as targeting checkpoints like PD-1/PD-L1, have proved successful in cancer therapy and can reinvigorate immune responses. The potential of this approach for treating chronic infectious diseases like TB has been recognized, but a lack of suitable immunotherapeutic targets, i.e., immune cell inhibitory receptors that trigger immunosuppression specifically during Mycobacterium tuberculosis pathogenesis, has limited the application of this strategy in the development of new TB therapies. Our focus in this study was to address this gap and search for an M. tuberculosis-specific checkpoint target. Our results suggest that CD84 is a putative inhibitory receptor that may be a suitable target for the development of TB-specific checkpoint immunotherapies.
Collapse
Affiliation(s)
- Nan Zheng
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Joy Fleming
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Peilei Hu
- Hunan Chest Hospital, Changsha, Hunan Province, China
| | - Jianjian Jiao
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Guoqin Zhang
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ruifang Yang
- Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing, China
| | - Chuanyou Li
- Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing, China
| | - Yi Liu
- Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Key Laboratory for Drug Resistant Tuberculosis Research, Beijing, China
| | - Lijun Bi
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- CAS Center of Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- Guangdong Province Key Laboratory of TB Systems Biology and Translational Medicine, Foshan, Guangdong Province, China
| | - Hongtai Zhang
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
8
|
Strömdahl AC, Ignatowicz L, Petruk G, Butrym M, Wasserstrom S, Schmidtchen A, Puthia M. Peptide-coated polyurethane material reduces wound infection and inflammation. Acta Biomater 2021; 128:314-331. [PMID: 33951491 DOI: 10.1016/j.actbio.2021.04.045] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 01/24/2023]
Abstract
There is an urgent need for treatments that not only reduce bacterial infection that occurs during wounding but that also target the accompanying excessive inflammatory response. TCP-25, a thrombin-derived antibacterial peptide, scavenges toll-like receptor agonists such as endotoxins and lipoteichoic acid and prevents toll-like receptor-4 dimerization to reduce infection-related inflammation in vivo. Using a combination of biophysical, cellular, and microbiological assays followed by experimental studies in mouse and pig models, we show that TCP-25, when delivered from a polyurethane (PU) material, exerts anti-infective and anti-inflammatory effects in vitro and in vivo. Specifically, TCP-25 killed the common wound pathogens, Pseudomonas aeruginosa and Staphylococcus aureus, in both in vitro and in vivo assays. Furthermore, after its release from the PU material, the peptide retained its capacity to induce its helical conformation upon endotoxin interaction, yielding reduced activation of NF-κB in THP-1 reporter cells, and diminished accumulation of inflammatory cells and subsequent release of IL-6 and TNF-α in subcutaneous implant models in vivo. Moreover, in a porcine partial thickness wound infection model, TCP-25 treated infection with S. aureus, and reduced the concomitant inflammatory response. Taken together, these findings demonstrate a combined antibacterial and anti-inflammatory effect of TCP-25 delivered from PU in vitro, and in mouse and porcine in vivo models of localized infection-inflammation. STATEMENT OF SIGNIFICANCE: Local wound infections may result in systemic complications and can be difficult to treat due to increasing antimicrobial resistance. Surgical site infections and biomaterial-related infections present a major challenge for hospitals. In recent years, various antimicrobial coatings have been developed for infection prevention and current concepts focus on various matrices with added anti-infective components, including various antibiotics and antiseptics. We have developed a dual action wound dressing concept where the host defense peptide TCP-25, when delivered from a PU material, targets both bacterial infection and the accompanying inflammation. TCP-25 PU showed efficacy in in vitro and experimental wound models in mouse and minipigs.
Collapse
|
9
|
Schorey JS, Cheng Y, McManus WR. Bacteria- and host-derived extracellular vesicles - two sides of the same coin? J Cell Sci 2021; 134:268991. [PMID: 34081134 DOI: 10.1242/jcs.256628] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Intracellular bacterial pathogens spend portions of their life cycle both inside and outside host cells. While in these two distinct environments, they release or shed bacterial components, including virulence factors that promote their survival and replication. Some of these components are released through extracellular vesicles, which are either derived from the bacteria themselves or from the host cells. Bacteria- and host-derived vesicles have been studied almost exclusively in isolation from each other, with little discussion of the other type of secreted vesicles, despite the fact that both are generated during an in vivo infection and both are likely play a role in bacterial pathogenesis and host immunity. In this Review, we aim to bridge this gap and discuss what we know of bacterial membrane vesicles in their generation and composition. We will compare and contrast this with the composition of host-derived vesicles with regard to bacterial components. We will also compare host cell responses to the different vesicles, with a focus on how these vesicles modulate the immune response, using Mycobacterium, Listeria and Salmonella as specific examples for these comparisons.
Collapse
Affiliation(s)
- Jeffrey S Schorey
- Department of Biological Sciences, Galvin Life Science Center, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Yong Cheng
- Department of Biochemistry and Molecular Biology, Noble Research Center, Oklahoma State University, Stillwater, OK 74078, USA
| | - William R McManus
- Department of Biological Sciences, Galvin Life Science Center, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
10
|
Shariq M, Quadir N, Sharma N, Singh J, Sheikh JA, Khubaib M, Hasnain SE, Ehtesham NZ. Mycobacterium tuberculosis RipA Dampens TLR4-Mediated Host Protective Response Using a Multi-Pronged Approach Involving Autophagy, Apoptosis, Metabolic Repurposing, and Immune Modulation. Front Immunol 2021; 12:636644. [PMID: 33746976 PMCID: PMC7969667 DOI: 10.3389/fimmu.2021.636644] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/03/2021] [Indexed: 12/26/2022] Open
Abstract
Reductive evolution has endowed Mycobacterium tuberculosis (M. tb) with moonlighting in protein functions. We demonstrate that RipA (Rv1477), a peptidoglycan hydrolase, activates the NFκB signaling pathway and elicits the production of pro-inflammatory cytokines, TNF-α, IL-6, and IL-12, through the activation of an innate immune-receptor, toll-like receptor (TLR)4. RipA also induces an enhanced expression of macrophage activation markers MHC-II, CD80, and CD86, suggestive of M1 polarization. RipA harbors LC3 (Microtubule-associated protein 1A/1B-light chain 3) motifs known to be involved in autophagy regulation and indeed alters the levels of autophagy markers LC3BII and P62/SQSTM1 (Sequestosome-1), along with an increase in the ratio of P62/Beclin1, a hallmark of autophagy inhibition. The use of pharmacological agents, rapamycin and bafilomycin A1, reveals that RipA activates PI3K-AKT-mTORC1 signaling cascade that ultimately culminates in the inhibition of autophagy initiating kinase ULK1 (Unc-51 like autophagy activating kinase). This inhibition of autophagy translates into efficient intracellular survival, within macrophages, of recombinant Mycobacterium smegmatis expressing M. tb RipA. RipA, which also localizes into mitochondria, inhibits the production of oxidative phosphorylation enzymes to promote a Warburg-like phenotype in macrophages that favors bacterial replication. Furthermore, RipA also inhibited caspase-dependent programed cell death in macrophages, thus hindering an efficient innate antibacterial response. Collectively, our results highlight the role of an endopeptidase to create a permissive replication niche in host cells by inducing the repression of autophagy and apoptosis, along with metabolic reprogramming, and pointing to the role of RipA in disease pathogenesis.
Collapse
Affiliation(s)
- Mohd Shariq
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India
| | - Neha Quadir
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India.,Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Neha Sharma
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India.,Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Jasdeep Singh
- Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Javaid A Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Mohd Khubaib
- Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
| | - Seyed E Hasnain
- Jamia Hamdard Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India.,Dr. Reddy's Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, India.,Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D) Hauz Khas, New Delhi, India
| | - Nasreen Z Ehtesham
- Indian Council of Medical Research-National Institute of Pathology, New Delhi, India
| |
Collapse
|
11
|
Maphasa RE, Meyer M, Dube A. The Macrophage Response to Mycobacterium tuberculosis and Opportunities for Autophagy Inducing Nanomedicines for Tuberculosis Therapy. Front Cell Infect Microbiol 2021; 10:618414. [PMID: 33628745 PMCID: PMC7897680 DOI: 10.3389/fcimb.2020.618414] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/18/2020] [Indexed: 12/23/2022] Open
Abstract
The major causative agent of tuberculosis (TB), i.e., Mycobacterium tuberculosis (Mtb), has developed mechanisms to evade host defense responses and persist within host cells for prolonged periods of time. Mtb is also increasingly resistant to existing anti-TB drugs. There is therefore an urgent need to develop new therapeutics for TB and host directed therapies (HDTs) hold potential as effective therapeutics for TB. There is growing interest in the induction of autophagy in Mtb host cells using autophagy inducing compounds (AICs). Nanoparticles (NPs) can enhance the effect of AICs, thus improving stability, enabling cell targeting and providing opportunities for multimodal therapy. In this review, we focus on the macrophage responses to Mtb infection, in particular, the mechanistic aspects of autophagy and the evasion of autophagy by intracellular Mtb. Due to the overlap between the onset of autophagy and apoptosis; we also focus on the relationship between apoptosis and autophagy. We will also review known AICs in the context of Mtb infection. Finally, we discuss the applications of NPs in inducing autophagy with the intention of sharing insights to encourage further research and development of nanomedicine HDTs for TB therapy.
Collapse
Affiliation(s)
- Retsepile E Maphasa
- Infectious Disease Nanomedicine Research Group, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| | - Mervin Meyer
- DST/Mintek Nanotechnology Innovation Centre, Biolabels Node, Department of Biotechnology, University of the Western Cape, Cape Town, South Africa
| | - Admire Dube
- Infectious Disease Nanomedicine Research Group, School of Pharmacy, University of the Western Cape, Cape Town, South Africa
| |
Collapse
|
12
|
Nore KG, Jørgensen MJ, Dyrhol-Riise AM, Jenum S, Tonby K. Elevated Levels of Anti-Inflammatory Eicosanoids and Monocyte Heterogeneity in Mycobacterium tuberculosis Infection and Disease. Front Immunol 2020; 11:579849. [PMID: 33304347 PMCID: PMC7693556 DOI: 10.3389/fimmu.2020.579849] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022] Open
Abstract
Eicosanoids modulate both innate and adaptive immune responses in Mycobacterium tuberculosis (Mtb) infection and have been suggested as possible Host Directed Therapy (HDT) targets, but more knowledge of eicosanoid dynamics in Mtb infection is required. We investigated the levels and ratios of eicosanoid mediators and their cellular sources, monocyte subsets and CD4 T cells in Tuberculosis (TB) patients with various clinical states of Mtb infection. Patients consenting to prospective enrolment in a TB quality registry and biorepository, 16 with pulmonary TB (before and at-end-of treatment), 14 with extrapulmonary TB and 17 latently infected (LTBI) were included. Plasma levels of Prostaglandin E2 (PGE2), Lipoxin A4 (LXA4), and Leukotriene B4 (LTB4) were measured by enzyme-linked immunosorbent assay. Monocyte subsets and CD4 T cells and their expression of Cyclooxygenase-2 (COX-2), Prostaglandin receptor EP2 (EP2), and 5-Lipoxygenase (5-LOX) were analyzed by flow cytometry with and without Purified Protein Derivate (PPD)-stimulation. Pulmonary TB patients had elevated levels of the anti-inflammatory mediator LXA4 at diagnosis compared to LTBI (p < 0.01), while levels of PGE2 and LTB4 showed no difference between clinical states of Mtb infection. LTB4 was the only mediator to be reduced upon treatment (p < 0.05), along with the ratio LTB4/LXA4 (p < 0.01). Pulmonary TB patients had higher levels of total monocytes at diagnosis compared to end-of-treatment and LTBI (both p < 0.05), and a relative increase in the classical monocyte subset. All monocyte subsets had low basal expression of COX-2 and 5-LOX, which were markedly increased upon PPD stimulation. By contrast, the expression of EP2 was reduced upon stimulation. CD4 T cells expressed low basal COX-2 activity that increased modestly upon stimulation, whereas their basal expression of 5-LOX was considerable. In conclusion, the level of eicosanoids in plasma seem to vary between clinical states of Mtb infection. Mediators in the eicosanoid system are present in monocytes and CD4 T cells. The expression of eicosanoids in monocytes are responsive to mycobacterial stimulation independent of Mtb disease state, but subsets are heterogeneous with regard to eicosanoid-mediator expression. Further exploration of eicosanoid mediators as targets for HDT in TB are warranted.
Collapse
Affiliation(s)
- Kristin Grotle Nore
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marthe Jøntvedt Jørgensen
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Synne Jenum
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Kristian Tonby
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
13
|
Sutiwisesak R, Hicks ND, Boyce S, Murphy KC, Papavinasasundaram K, Carpenter SM, Boucau J, Joshi N, Le Gall S, Fortune SM, Sassetti CM, Behar SM. A natural polymorphism of Mycobacterium tuberculosis in the esxH gene disrupts immunodomination by the TB10.4-specific CD8 T cell response. PLoS Pathog 2020; 16:e1009000. [PMID: 33075106 PMCID: PMC7597557 DOI: 10.1371/journal.ppat.1009000] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/29/2020] [Accepted: 09/23/2020] [Indexed: 12/20/2022] Open
Abstract
CD8 T cells provide limited protection against Mycobacterium
tuberculosis (Mtb) infection in the mouse model. As Mtb causes
chronic infection in mice and humans, we hypothesize that Mtb impairs T cell
responses as an immune evasion strategy. TB10.4 is an immunodominant antigen in
people, nonhuman primates, and mice, which is encoded by the
esxH gene. In C57BL/6 mice, 30–50% of pulmonary CD8 T cells
recognize the TB10.44−11 epitope. However, TB10.4-specific CD8 T
cells fail to recognize Mtb-infected macrophages. We speculate that Mtb elicits
immunodominant CD8 T cell responses to antigens that are inefficiently presented
by infected cells, thereby focusing CD8 T cells on nonprotective antigens. Here,
we leverage naturally occurring polymorphisms in esxH, which
frequently occur in lineage 1 strains, to test this “decoy hypothesis”. Using
the clinical isolate 667, which contains an EsxHA10T polymorphism, we
observe a drastic change in the hierarchy of CD8 T cells. Using isogenic
Erd.EsxHA10T and Erd.EsxHWT strains, we prove that
this polymorphism alters the hierarchy of immunodominant CD8 T cell responses.
Our data are best explained by immunodomination, a mechanism by which
competition for APC leads to dominant responses suppressing subdominant
responses. These results were surprising as the variant epitope can bind to
H2-Kb and is recognized by TB10.4-specific CD8 T cells. The
dramatic change in TB10.4-specific CD8 responses resulted from increased
proteolytic degradation of A10T variant, which destroyed the
TB10.44-11epitope. Importantly, this polymorphism affected T cell
priming and recognition of infected cells. These data support a model in which
nonprotective CD8 T cells become immunodominant and suppress subdominant
responses. Thus, polymorphisms between clinical Mtb strains, and BCG or H37Rv
sequence-based vaccines could lead to a mismatch between T cells that are primed
by vaccines and the epitopes presented by infected cells. Reprograming host
immune responses should be considered in the future design of vaccines. An important question for vaccine developers is the relative potency of CD4 vs.
CD8 T cells against Mtb, as strategies differ for eliciting these different T
cell subsets. Despite robust antigen-specific pulmonary CD8 T cell responses,
CD4 T cells mediate more protection than CD8 T cells in the murine model. Most
CD8 T cells recognize a single antigen, TB10.4, which is encoded by the
esxH gene. Based on finding that
TB10.44−11-specific CD8 T cells poorly recognize Mtb-infected
macrophages, we hypothesized that Mtb evades detection by CD8 T cells and
focuses the CD8 T cell response on non-protective antigen. We termed these
antigens “decoy antigens.” To test this hypothesis, we took advantage of a
natural variant of the esxH gene, which contains an A10T
polymorphism within the TB10.44−11 epitope. This polymorphism
drastically alters the hierarchy of CD8 T cell responses elicited by Mtb. These
data suggest that immunodomination by the TB10.4 epitope acts to suppress
subdominant CD8 T cell responses to other Mtb antigens, impairing the CD8 T cell
response to other Mtb antigens, some of which might be presented by Mtb-infected
macrophages and be targets of protective immunity. Importantly, this single
amino acid polymorphism, which does not significantly alter MHC-binding or T
cell recognition, alters the half-life of the epitope and consequently, has a
profound effect on CD8 T cell priming and recognition of infected cells. These
data also provide a mechanism that could be exploited to manipulate the
hierarchy of immunodominant responses.
Collapse
Affiliation(s)
- Rujapak Sutiwisesak
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Nathan D. Hicks
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts, United States of
America
| | - Shayla Boyce
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Kenan C. Murphy
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Kadamba Papavinasasundaram
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Stephen M. Carpenter
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Julie Boucau
- Ragon Institute of Massachusetts General Hospital, Massachusetts
Institute of Technology and Harvard University, Cambridge, MA, United States of
America
| | - Neelambari Joshi
- Ragon Institute of Massachusetts General Hospital, Massachusetts
Institute of Technology and Harvard University, Cambridge, MA, United States of
America
| | - Sylvie Le Gall
- Ragon Institute of Massachusetts General Hospital, Massachusetts
Institute of Technology and Harvard University, Cambridge, MA, United States of
America
| | - Sarah M. Fortune
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan
School of Public Health, Boston, Massachusetts, United States of
America
| | - Christopher M. Sassetti
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
| | - Samuel M. Behar
- Immunology and Microbiology Program, Graduate School of Biomedical
Science, University of Massachusetts Medical School, Worcester, Massachusetts,
United States of America
- Department of Microbiology and Physiological Systems, University of
Massachusetts Medical School, Worcester, Massachusetts, United States of
America
- * E-mail:
| |
Collapse
|
14
|
Patankar YR, Sutiwisesak R, Boyce S, Lai R, Lindestam Arlehamn CS, Sette A, Behar SM. Limited recognition of Mycobacterium tuberculosis-infected macrophages by polyclonal CD4 and CD8 T cells from the lungs of infected mice. Mucosal Immunol 2020; 13:140-148. [PMID: 31636345 PMCID: PMC7161428 DOI: 10.1038/s41385-019-0217-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 02/04/2023]
Abstract
Immune responses following Mycobacterium tuberculosis (Mtb) infection or vaccination are frequently assessed by measuring T-cell recognition of crude Mtb antigens, recombinant proteins, or peptide epitopes. We previously showed that not all Mtb-specific T cells recognize Mtb-infected macrophages. Thus, an important question is what proportion of T cells elicited by Mtb infection recognize Mtb-infected macrophages. We address this question by developing a modified elispot assay using viable Mtb-infected macrophages, a low multiplicity of infection and purified T cells. In C57BL/6 mice, CD4 and CD8 T cells were classically MHC restricted. Comparable frequencies of T cells that recognize Mtb-infected macrophages were determined using interferon-γ elispot and intracellular cytokine staining, and lung CD4 T cells more sensitively recognized Mtb-infected macrophages than lung CD8 T cells. Compared to the relatively high frequencies of T cells specific for antigens such as ESAT-6 and TB10.4, low frequencies of total pulmonary T cells elicited by aerosolized Mtb infection recognize Mtb-infected macrophages. Finally, we demonstrate that BCG vaccination elicits T cells that recognize Mtb-infected macrophages. We propose that the frequency of T cells that recognize infected macrophages could correlate with protective immunity and may be an alternative approach to measuring T-cell responses to Mtb antigens.
Collapse
Affiliation(s)
- Yash R. Patankar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rujapak Sutiwisesak
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Shayla Boyce
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Rocky Lai
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Cecilia S. Lindestam Arlehamn
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Alessandro Sette
- 0000 0004 0461 3162grid.185006.aDepartment of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA 92037 USA ,0000 0001 2107 4242grid.266100.3Department of Medicine, University of California San Diego, La Jolla, CA 92093 USA
| | - Samuel M. Behar
- 0000 0001 0742 0364grid.168645.8Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| |
Collapse
|
15
|
Stamm CE, Pasko BL, Chaisavaneeyakorn S, Franco LH, Nair VR, Weigele BA, Alto NM, Shiloh MU. Screening Mycobacterium tuberculosis Secreted Proteins Identifies Mpt64 as a Eukaryotic Membrane-Binding Bacterial Effector. mSphere 2019; 4:e00354-19. [PMID: 31167949 PMCID: PMC6553557 DOI: 10.1128/msphere.00354-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 05/19/2019] [Indexed: 02/07/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis, is one of the most successful human pathogens. One reason for its success is that Mtb can reside within host macrophages, a cell type that normally functions to phagocytose and destroy infectious bacteria. However, Mtb is able to evade macrophage defenses in order to survive for prolonged periods of time. Many intracellular pathogens secrete virulence factors targeting host membranes and organelles to remodel their intracellular environmental niche. We hypothesized that Mtb secreted proteins that target host membranes are vital for Mtb to adapt to and manipulate the host environment for survival. Thus, we characterized 200 secreted proteins from Mtb for their ability to associate with eukaryotic membranes using a unique temperature-sensitive yeast screen and to manipulate host trafficking pathways using a modified inducible secretion screen. We identified five Mtb secreted proteins that both associated with eukaryotic membranes and altered the host secretory pathway. One of these secreted proteins, Mpt64, localized to the endoplasmic reticulum during Mtb infection of murine and human macrophages and impaired the unfolded protein response in macrophages. These data highlight the importance of secreted proteins in Mtb pathogenesis and provide a basis for further investigation into their molecular mechanisms.IMPORTANCE Advances have been made to identify secreted proteins of Mycobacterium tuberculosis during animal infections. These data, combined with transposon screens identifying genes important for M. tuberculosis virulence, have generated a vast resource of potential M. tuberculosis virulence proteins. However, the function of many of these proteins in M. tuberculosis pathogenesis remains elusive. We have integrated three cell biological screens to characterize nearly 200 M. tuberculosis secreted proteins for eukaryotic membrane binding, host subcellular localization, and interactions with host vesicular trafficking. In addition, we observed the localization of one secreted protein, Mpt64, to the endoplasmic reticulum (ER) during M. tuberculosis infection of macrophages. Interestingly, although Mpt64 is exported by the Sec pathway, its delivery into host cells was dependent upon the action of the type VII secretion system. Finally, we observed that Mpt64 impairs the ER-mediated unfolded protein response in macrophages.
Collapse
Affiliation(s)
- Chelsea E Stamm
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Breanna L Pasko
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sujittra Chaisavaneeyakorn
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Luis H Franco
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Center for Autophagy Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vidhya R Nair
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bethany A Weigele
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Neal M Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Michael U Shiloh
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
16
|
Song M, Xu S, Zhong A, Zhang J. Crosstalk between macrophage and T cell in atherosclerosis: Potential therapeutic targets for cardiovascular diseases. Clin Immunol 2019; 202:11-17. [DOI: 10.1016/j.clim.2019.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 12/03/2018] [Accepted: 03/01/2019] [Indexed: 01/05/2023]
|
17
|
Niu W, Sun B, Li M, Cui J, Huang J, Zhang L. TLR-4/microRNA-125a/NF-κB signaling modulates the immune response to Mycobacterium tuberculosis infection. Cell Cycle 2018; 17:1931-1945. [PMID: 30153074 DOI: 10.1080/15384101.2018.1509636] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis, could lead to kinds of clinical disorders and remains a leading global health problem, resulting in great morbidity and mortality worldwide. Previous studies have firmly demonstrated that M. tuberculosis (M.tb) has evolved to utilize different mechanisms to evade or attenuate the host immune response, such as regulation of immune-related genes by modulation of miRNAs of host or bacteria. However, the knowledge of functions of miRNAs during M.tb infection remains limited. Here, we reported that a host microRNA, miR-125a, was significantly up-regulated by M.tb infection in both RAW264.7 and THP-1cells, in a TLR4 signaling-dependent manner. Subsequently, our results demonstrated that miR-125a was a negative regulator of NF-kB pathway by directly targeting TRAF6, resulting in the suppression of cytokines, attenuation of immune response and promotion of M.tb survival. Taken together, our findings provide a novel detailed molecular mechanism in which miR-125a was enhanced to inhibit inflammatory cytokines secretion and attenuate the immune response during M.tb infection in RAW264.7 and THP-1 cells, and suggest an intrinsic a promising anti-M.tb therapeutic target.
Collapse
Affiliation(s)
- Wenyi Niu
- a Department of Tuberculosis , The First Affliated Hospital of Xinxiang Medical University , Weihui , China
| | - Bing Sun
- a Department of Tuberculosis , The First Affliated Hospital of Xinxiang Medical University , Weihui , China
| | - Mingying Li
- a Department of Tuberculosis , The First Affliated Hospital of Xinxiang Medical University , Weihui , China
| | - Junwei Cui
- a Department of Tuberculosis , The First Affliated Hospital of Xinxiang Medical University , Weihui , China
| | - Jian Huang
- a Department of Tuberculosis , The First Affliated Hospital of Xinxiang Medical University , Weihui , China
| | - Ligong Zhang
- a Department of Tuberculosis , The First Affliated Hospital of Xinxiang Medical University , Weihui , China
| |
Collapse
|
18
|
Shapouri-Moghaddam A, Mohammadian S, Vazini H, Taghadosi M, Esmaeili SA, Mardani F, Seifi B, Mohammadi A, Afshari JT, Sahebkar A. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol 2018; 233:6425-6440. [PMID: 29319160 DOI: 10.1002/jcp.26429] [Citation(s) in RCA: 3154] [Impact Index Per Article: 450.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 01/05/2018] [Indexed: 12/12/2022]
Abstract
Macrophages are heterogeneous and their phenotype and functions are regulated by the surrounding micro-environment. Macrophages commonly exist in two distinct subsets: 1) Classically activated or M1 macrophages, which are pro-inflammatory and polarized by lipopolysaccharide (LPS) either alone or in association with Th1 cytokines such as IFN-γ, GM-CSF, and produce pro-inflammatory cytokines such as interleukin-1β (IL-1β), IL-6, IL-12, IL-23, and TNF-α; and 2) Alternatively activated or M2 macrophages, which are anti-inflammatory and immunoregulatory and polarized by Th2 cytokines such as IL-4 and IL-13 and produce anti-inflammatory cytokines such as IL-10 and TGF-β. M1 and M2 macrophages have different functions and transcriptional profiles. They have unique abilities by destroying pathogens or repair the inflammation-associated injury. It is known that M1/M2 macrophage balance polarization governs the fate of an organ in inflammation or injury. When the infection or inflammation is severe enough to affect an organ, macrophages first exhibit the M1 phenotype to release TNF-α, IL-1β, IL-12, and IL-23 against the stimulus. But, if M1 phase continues, it can cause tissue damage. Therefore, M2 macrophages secrete high amounts of IL-10 and TGF-β to suppress the inflammation, contribute to tissue repair, remodeling, vasculogenesis, and retain homeostasis. In this review, we first discuss the basic biology of macrophages including origin, differentiation and activation, tissue distribution, plasticity and polarization, migration, antigen presentation capacity, cytokine and chemokine production, metabolism, and involvement of microRNAs in macrophage polarization and function. Secondly, we discuss the protective and pathogenic role of the macrophage subsets in normal and pathological pregnancy, anti-microbial defense, anti-tumor immunity, metabolic disease and obesity, asthma and allergy, atherosclerosis, fibrosis, wound healing, and autoimmunity.
Collapse
Affiliation(s)
- Abbas Shapouri-Moghaddam
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeed Mohammadian
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Vazini
- Nursing Department, Basic Sciences Faculty, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Mahdi Taghadosi
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed-Alireza Esmaeili
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Mardani
- Faculty of Medicine, Student Research Committee, Immunology Research Center, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bita Seifi
- Department of Anatomy, Islamic Azad University, Mashhad Branch, Iran
| | - Asadollah Mohammadi
- Inflammation and Inflammatory Disease Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil T Afshari
- Faculty of Medicine, Department of Immunology, BuAli Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Leisching G, Wiid I, Baker B. OAS1, 2, and 3: Significance During Active Tuberculosis? J Infect Dis 2018; 217:1517-1521. [DOI: 10.1093/infdis/jiy084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 02/12/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Gina Leisching
- South African Medical Research Council Centre for Tuberculosis Research, Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, Stellenbosch University
| | - Ian Wiid
- South African Medical Research Council Centre for Tuberculosis Research, Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, Stellenbosch University
| | - Bienyameen Baker
- South African Medical Research Council Centre for Tuberculosis Research, Department of Science and Technology/National Research Foundation Centre of Excellence for Biomedical Tuberculosis Research, Faculty of Medicine and Health Sciences, Stellenbosch University
| |
Collapse
|
20
|
Kumar P. IFNγ-producing CD4 + T lymphocytes: the double-edged swords in tuberculosis. Clin Transl Med 2017; 6:21. [PMID: 28646367 PMCID: PMC5482791 DOI: 10.1186/s40169-017-0151-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/07/2017] [Indexed: 11/30/2022] Open
Abstract
IFNγ-producing CD4+ T cells (IFNγ+CD4+ T cells) are the key orchestrators of protective immunity against Mycobacterium tuberculosis (Mtb). Primarily, these cells act by enabling Mtb-infected macrophages to enforce phagosome-lysosome fusion, produce reactive nitrogen intermediates (RNIs), and activate autophagy pathways. However, TB is a heterogeneous disease and a host of clinical and experimental findings has also implicated IFNγ+CD4+ T cells in TB pathogenesis. High frequency of IFNγ+CD4+ T cells is the most invariable feature of the active disease. Active TB patients mount a heightened IFNγ+CD4+ T cell response to mycobacterial antigens and demonstrate an IFNγ-inducible transcriptomic signature. IFNγ+CD4+ T cells have also been shown to mediate TB-associated immune reconstitution inflammatory syndrome (TB-IRIS) observed in a subset of antiretroviral therapy (ART)-treated HIV- and Mtb-coinfected people. The pathological face of IFNγ+CD4+ T cells during mycobacterial infection is further uncovered by studies in the animal model of TB-IRIS and in Mtb-infected PD-1-/- mice. This manuscript encompasses the evidence supporting the dual role of IFNγ+CD4+ T cells during Mtb infection and sheds light on immune mechanisms involved in protection versus pathogenesis.
Collapse
Affiliation(s)
- Pawan Kumar
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India.
| |
Collapse
|
21
|
Meng L, Tong J, Wang H, Tao C, Wang Q, Niu C, Zhang X, Gao Q. PPE38 Protein of Mycobacterium tuberculosis Inhibits Macrophage MHC Class I Expression and Dampens CD8 + T Cell Responses. Front Cell Infect Microbiol 2017; 7:68. [PMID: 28348981 PMCID: PMC5346565 DOI: 10.3389/fcimb.2017.00068] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/22/2017] [Indexed: 12/23/2022] Open
Abstract
Suppression of CD8+ T cell activation is a critical mechanism used by Mycobacterium tuberculosis (MTB) to escape protective host immune responses. PPE38 belongs to the unique PPE family of MTB and in our previous study, PPE38 protein was speculated to participate in manipulating macrophage MHC class I pathway. To test this hypothesis, the function of mycobacterial PPE38 protein was assessed here using macrophage and mouse infection models. Decreased amount of MHC class I was observed on the surface of macrophages infected with PPE38-expressing mycobacteria. The transcript of genes encoding MHC class I was also inhibited by PPE38. After infection of C57BL/6 mice with Mycobacterium smegmatis expressing PPE38 (Msmeg-PPE38), decreased number of CD8+ T cells was found in spleen, liver, and lungs through immunohistochemical analysis, comparing to the control strain harboring empty vector (Msmeg-V). Consistently, flow cytometry assay showed that fewer effector/memory CD8+ T cells (CD44highCD62Llow) were activated in spleen from Msmeg-PPE38 infected mice. Moreover, Msmeg-PPE38 confers a growth advantage over Msmeg-V in C57BL/6 mice, indicating an effect of PPE38 to favor mycobacterial persistence in vivo. Overall, this study shows a unique biological function of PPE38 protein to facilitate mycobacteria to escape host immunity, and provides hints for TB vaccine development.
Collapse
Affiliation(s)
- Lu Meng
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Jingfeng Tong
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Hui Wang
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan UniversityShanghai, China; The State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, School of Medicine, Shenzhen UniversityGuangdong, China
| | - Chengwu Tao
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences Shanghai, China
| | - Qinglan Wang
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Chen Niu
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| | - Xiaoming Zhang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences Shanghai, China
| | - Qian Gao
- Key laboratory of Medical Molecular Virology, Institute of Biomedical Sciences and Institute of Medical Microbiology, Shanghai Medical College, Fudan University Shanghai, China
| |
Collapse
|
22
|
Schaaf K, Smith SR, Duverger A, Wagner F, Wolschendorf F, Westfall AO, Kutsch O, Sun J. Mycobacterium tuberculosis exploits the PPM1A signaling pathway to block host macrophage apoptosis. Sci Rep 2017; 7:42101. [PMID: 28176854 PMCID: PMC5296758 DOI: 10.1038/srep42101] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/30/2016] [Indexed: 12/19/2022] Open
Abstract
The ability to suppress host macrophage apoptosis is essential for M. tuberculosis (Mtb) to replicate intracellularly while protecting it from antibiotic treatment. We recently described that Mtb infection upregulated expression of the host phosphatase PPM1A, which impairs the antibacterial response of macrophages. Here we establish PPM1A as a checkpoint target used by Mtb to suppress macrophage apoptosis. Overproduction of PPM1A suppressed apoptosis of Mtb-infected macrophages by a mechanism that involves inactivation of the c-Jun N-terminal kinase (JNK). Targeted depletion of PPM1A by shRNA or inhibition of PPM1A activity by sanguinarine restored JNK activation, resulting in increased apoptosis of Mtb-infected macrophages. We also demonstrate that activation of JNK by subtoxic concentrations of anisomycin induced selective apoptotic killing of Mtb-infected human macrophages, which was completely blocked in the presence of a specific JNK inhibitor. Finally, selective killing of Mtb-infected macrophages and subsequent bacterial release enabled rifampicin to effectively kill Mtb at concentrations that were insufficient to act against intracellular Mtb, providing proof of principle for the efficacy of a "release and kill" strategy. Taken together, these findings suggest that drug-induced selective apoptosis of Mtb-infected macrophages is achievable.
Collapse
Affiliation(s)
- Kaitlyn Schaaf
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Samuel R. Smith
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alexandra Duverger
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Frederic Wagner
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Frank Wolschendorf
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Andrew O. Westfall
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Olaf Kutsch
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jim Sun
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
23
|
Pires D, Marques J, Pombo JP, Carmo N, Bettencourt P, Neyrolles O, Lugo-Villarino G, Anes E. Role of Cathepsins in Mycobacterium tuberculosis Survival in Human Macrophages. Sci Rep 2016; 6:32247. [PMID: 27572605 PMCID: PMC5004184 DOI: 10.1038/srep32247] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 08/04/2016] [Indexed: 02/07/2023] Open
Abstract
Cathepsins are proteolytic enzymes that function in the endocytic pathway, especially in lysosomes, where they contribute directly to pathogen killing or indirectly, by their involvement in the antigen presentation pathways. Mycobacterium tuberculosis (MTB) is a facultative intracellular pathogen that survives inside the macrophage phagosomes by inhibiting their maturation to phagolysosomes and thus avoiding a low pH and protease-rich environment. We previously showed that mycobacterial inhibition of the proinflammatory transcription factor NF-κB results in impaired delivery of lysosomal enzymes to phagosomes and reduced pathogen killing. Here, we elucidate how MTB also controls cathepsins and their inhibitors, cystatins, at the level of gene expression and proteolytic activity. MTB induced a general down-regulation of cathepsin expression in infected cells, and inhibited IFNγ-mediated increase of cathepsin mRNA. We further show that a decrease in cathepsins B, S and L favours bacterial survival within human primary macrophages. A siRNA knockdown screen of a large set of cathepsins revealed that almost half of these enzymes have a role in pathogen killing, while only cathepsin F coincided with MTB resilience. Overall, we show that cathepsins are important for the control of MTB infection, and as a response, it manipulates their expression and activity to favour its intracellular survival.
Collapse
Affiliation(s)
- David Pires
- Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Portugal
| | - Joana Marques
- Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - João Palma Pombo
- Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Nuno Carmo
- Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Portugal
| | - Paulo Bettencourt
- Research Institute for Medicines, iMed-ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Portugal
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Portugal
| | - Olivier Neyrolles
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Geanncarlo Lugo-Villarino
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Elsa Anes
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Portugal
| |
Collapse
|
24
|
Meng C, Wang X, Xu Z, Hu M, Liu J, Pan Z, Chen X, Jiao X. Murine Flt3 ligand-generated plasmacytoid and conventional dendritic cells display functional differentiation in activation, inflammation, and antigen presentation during BCG infection in vitro. In Vitro Cell Dev Biol Anim 2016; 53:67-76. [PMID: 27496194 DOI: 10.1007/s11626-016-0076-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 07/22/2016] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) are composed of distinct subsets. Their immunologic functions (especially in pathogenic infection, such as with mycobacteria) are poorly understood, largely because of their rarity and difficulty of preparation. We used the murine Fms-like tyrosine kinase 3 (Flt3) ligand to generate conventional DCs (FL-cDCs) and plasmacytoid DCs (FL-pDCs) and further evaluated their immunological responses to bacillus Calmette-Guérin (BCG) infection in vitro. BCG cells were observed inside both FL-cDCs and FL-pDCs by confocal microscopy, as confirmed by flow cytometric analysis showing a low infection rate of approximately 6 %, which was similar to in vivo results. The CD40, CD80, CD86, and MHC-II proteins were significantly upregulated in both FL-cDCs and -pDCs beginning at 4 h post-BCG exposure. FL-pDCs secreted TNF-α and IL-6 earlier and at significantly higher levels in the first 12 h following infection, but demonstrated delayed and weak activation and maturation compared to FL-cDCs. Although both subsets proved capable of presenting a mycobacterial antigen, FL-pDCs exhibited weaker activity in this respect than did FL-cDCs. In summary, the existence of FL-generated cDCs and pDCs imply functional differentiation in activation, inflammation, and antigen presentation, although both cells types participated extensively in the immune response to BCG infection.
Collapse
Affiliation(s)
- Chuang Meng
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiaoyan Wang
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhengzhong Xu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Maozhi Hu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.,Testing Center, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jiaying Liu
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhiming Pan
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xiang Chen
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Xinan Jiao
- Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
25
|
da Silva ALG, Bresciani MJ, Karnopp TE, Weber AF, Ellwanger JH, Henriques JAP, Valim ARDM, Possuelo LG. DNA damage and cellular abnormalities in tuberculosis, lung cancer and chronic obstructive pulmonary disease. Multidiscip Respir Med 2015; 10:38. [PMID: 26688728 PMCID: PMC4684909 DOI: 10.1186/s40248-015-0034-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/18/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Tuberculosis (TB), Lung Cancer (LC) and Chronic Obstructive Pulmonary Diseases (COPD) affect millions of individuals worldwide. Monitoring of DNA damage in pathological situations has been investigated because it can add a new dimension to clinical expression and may represent a potential target for therapeutic intervention. The aim of this study was to evaluate DNA damage and the frequency of cellular abnormalities in TB, LC and COPD patients by comparing them to healthy subjects. METHODS The detection of DNA damage by a buccal micronucleus cytome assay was investigated in patients with COPD (n = 28), LC (n = 18) and TB (n = 22) and compared to control individuals (n = 17). RESULTS The COPD group had a higher frequency of apoptotic cells compared to TB and LC group. The TB group showed a higher frequency of DNA damage, defect in cytokinesis, apoptotic and necrotic cells. Patients with LC had low frequency of chromosomal aberrations than TB and COPD patients. CONCLUSION COPD patients showed cellular abnormalities that corresponded to cell death by apoptosis and necrosis, while patients with TB presented defects in cytokinesis and dysfunctions in DNA repair that resulted in the formation of micronucleus (MN) besides apoptotic and necrotic cells. Patients with COPD, TB and LC had a low frequency of permanent DNA damage.
Collapse
Affiliation(s)
- Andréa Lúcia Gonçalves da Silva
- />Department of Health and Physical Education, University of Santa Cruz do Sul – UNISC, Avenida Independência, 2293, Bairro Universitário, Santa Cruz do Sul, RS CEP 96815-900 Brazil
| | - Maribel Josimara Bresciani
- />Department of Biology and Pharmacy, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
| | - Thaís Evelyn Karnopp
- />Department of Biology and Pharmacy, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
| | - Augusto Ferreira Weber
- />Department of Biology and Pharmacy, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
| | - Joel Henrique Ellwanger
- />Department of Biology and Pharmacy, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
- />Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre, RS Brazil
| | - João Antonio Pêgas Henriques
- />Graduate Program in Cellular and Molecular Biology, Federal University of Rio Grande do Sul - UFRGS, Porto Alegre, RS Brazil
| | - Andréia Rosane de Moura Valim
- />Department of Biology and Pharmacy, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
- />Graduate Program in Health Promotion, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
| | - Lia Gonçalves Possuelo
- />Department of Biology and Pharmacy, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
- />Graduate Program in Health Promotion, University of Santa Cruz do Sul - UNISC, Santa Cruz do Sul, RS Brazil
| |
Collapse
|
26
|
Evidence of a pro-apoptotic effect of specific antibodies in a bovine macrophage model of infection with Mycobacterium avium subsp. paratuberculosis. Vet Immunol Immunopathol 2015; 169:47-53. [PMID: 26827838 DOI: 10.1016/j.vetimm.2015.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 08/26/2015] [Accepted: 12/05/2015] [Indexed: 12/12/2022]
Abstract
Mycobacterium avium subspecies paratuberculosis (MAP) is the causative agent of Johne's disease (JD), a chronic granulomatous enteritis in ruminants. Understanding the protective immune response following infection is crucial to improve the diagnosis and the development of vaccines against this disease. The goal of this work was to assess whether specific antibodies were able to modulate the macrophage response to MAP infection by evaluating apoptosis and TNF-α secretion in an in vitro model. Sera from healthy (n=2), MAP-infected (n=3) and lipoarabinomannan (LAM)-immunized (n=3) bovines were evaluated. LAM was chosen as immunogen due to its relevant role in mycobacterial pathogenesis. We demonstrated by two different techniques (Acridine Orange/Ethidium Bromide microscopy and Annexin V/7-Amino-Actinomycin D flow cytometry) that the immune sera from both, MAP-infected and LAM-immunized bovines, significantly increased macrophage apoptosis in infected cultures. Comparable levels of apoptosis were detected when MAP was pre-incubated with purified specific antibodies instead of whole serum. Furthermore, this effect was accompanied by a significantly higher secretion of TNF-α. These results strongly suggest that specific antibodies could limit the impact of MAP on the apoptosis of bovine cells. This work would contribute to elucidate the role of the specific antibody response in bovine JD and its prevention.
Collapse
|
27
|
Amaral EP, Lasunskaia EB, D'Império-Lima MR. Innate immunity in tuberculosis: how the sensing of mycobacteria and tissue damage modulates macrophage death. Microbes Infect 2015; 18:11-20. [PMID: 26369715 DOI: 10.1016/j.micinf.2015.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 12/16/2022]
Abstract
The success of Mycobacterium tuberculosis as a human pathogen has been attributed to the ability of the bacillus to proliferate inside macrophages and to induce cell death. This review describes how the sensors of the innate immune system modulate the cell death pathways in infected macrophages and, consequently, the pathogenesis of tuberculosis.
Collapse
Affiliation(s)
- Eduardo P Amaral
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Elena B Lasunskaia
- Laboratory of Recognition Biology, Center of Biosciences and Biotechnology, State University of North Fluminense, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | | |
Collapse
|
28
|
Travar M, Petkovic M, Verhaz A. Type I, II, and III Interferons: Regulating Immunity to Mycobacterium tuberculosis Infection. Arch Immunol Ther Exp (Warsz) 2015; 64:19-31. [PMID: 26362801 DOI: 10.1007/s00005-015-0365-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 07/22/2015] [Indexed: 01/18/2023]
Abstract
Interferons (IFNs) are cytokines released by host cells in response to the presence of pathogens or tumor cells. The aim of this review was to present the previously known and new findings about the role of interferons type I and II, and recently discovered type III in Mycobacterium tuberculosis (M. tuberculosis) infection control. Infection of various cell types with M. tuberculosis induce both IFN-α and IFN-β synthesis. The majority of the studies support the findings that IFN type I actually promotes infection with M. tuberculosis. It has been well establish that IFN-γ has protective function against M. tuberculosis and the other mycobacteria and that the primary source of this cytokine are CD4(+) and CD8(+) T cells. Recently, it has been shown that also the innate lymphocytes, γδ T cells, natural killer (NK) T cells, and NK cells can also be the source of IFN-γ in response to mycobacterial infection. Several studies have shown that CD4(+) T cells protect mice against M. tuberculosis independently of IFN-γ. The balance between IFN-γ and different cytokines such as IL-10 and other Th2 cell cytokines is likely to influence disease outcome. Type I IFN appears to be detrimental through at least three separate, but overlapping, type I IFN-mediated mechanisms: induction of excessive apoptosis, specific suppression of Th1 and IFN-γ responses, and dampening of the immune response by strong IL-10 induction. Recently it has been found that M. tuberculosis infection in A549 lung epithelial cells stimulate up-regulation of IFN-λ genes in vitro. IFN-λs also have a role in modulation of Th1/Th2 response. IFN-λs are not essential for M. tuberculosis infection control, but can give some contribution in immune response to this pathogen.
Collapse
Affiliation(s)
- Maja Travar
- Department of Microbiology, University Hospital Clinical Centre Banja Luka, Banja Luka, Republic of Srpska, Bosnia and Herzegovina. .,Department of Microbiology and Immunology, Faculty of Medicine, Banja Luka University, Banja Luka, Republic of Srpska, Bosnia and Herzegovina.
| | - Miroslav Petkovic
- Department of Microbiology and Immunology, Faculty of Medicine, Banja Luka University, Banja Luka, Republic of Srpska, Bosnia and Herzegovina
| | - Antonija Verhaz
- Clinic for Infectious Diseases, University Hospital Clinical Centre Banja Luka, Banja Luka, Republic of Srpska, Bosnia and Herzegovina
| |
Collapse
|
29
|
He D, Zhang X, Gao Q, Huang R, Deng Z, Guo C, Guo Q, Huang J, Zhang H. Correlation between Serum Level of Monocyte Chemoattractant Protein-1 and Postoperative Recurrence of Spinal Tuberculosis in the Chinese Han Population. PLoS One 2015; 10:e0125756. [PMID: 25962150 PMCID: PMC4427401 DOI: 10.1371/journal.pone.0125756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/26/2015] [Indexed: 12/01/2022] Open
Abstract
Objective To correlate serum level of monocyte chemoattractant protein-1 (MCP-1) with postoperative recurrence of spinal tuberculosis in the Chinese Han population. Methods Patients of Han nationality with newly diagnosed spinal tuberculosis were consecutively included in this study. At different time points postoperatively, serum level of MCP-1 was determined using an enzyme linked immunosorbent assay. Recurrence of spinal tuberculosis after surgery and during the follow-up period was recorded. The correlation between serum MCP-1 level and recurrence of spinal tuberculosis was analyzed. Results A total of 169 patients with spinal tuberculosis were included in the study and followed up for an average of2.2±1.3 years (range, 1–5 years). Of these patients, 11 had postoperative recurrence of spinal tuberculosis. The patients’ serum level of MCP-1 increased significantly after postoperative recurrence of spinal tuberculosis. Once the symptoms of recurrence were cured, the serum level of MCP-1 decreased significantly and it did not differ from patients without disease recurrence. Conclusion Postoperative recurrence of spinal tuberculosis is likely to increase the serum level of MCP-1.
Collapse
Affiliation(s)
- Dan He
- Department of Neurology, The First Hospital of Changsha, Changsha, People’s Republic of China
| | - Xiaolu Zhang
- DepartmentofOrthopedics, The second affiliated Hospital, Fujian Medical University, Quanzhou, People’s Republic of China
| | - Qile Gao
- Department of Spine Surgery, Xiangya Spinal Surgery Center, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- * E-mail:
| | - Rongfu Huang
- Clinical Laboratory, The second affiliated Hospital, Fujian Medical University, Quanzhou, People’s Republic of China
| | - Zhansheng Deng
- Department of Spine Surgery, Xiangya Spinal Surgery Center, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Chaofeng Guo
- Department of Spine Surgery, Xiangya Spinal Surgery Center, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Qiang Guo
- Department of Spine Surgery, Xiangya Spinal Surgery Center, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Jia Huang
- Department of Spine Surgery, Xiangya Spinal Surgery Center, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Hongqi Zhang
- Department of Spine Surgery, Xiangya Spinal Surgery Center, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
30
|
Divangahi M, King IL, Pernet E. Alveolar macrophages and type I IFN in airway homeostasis and immunity. Trends Immunol 2015; 36:307-14. [PMID: 25843635 DOI: 10.1016/j.it.2015.03.005] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/10/2015] [Accepted: 03/10/2015] [Indexed: 12/24/2022]
Abstract
Globally, respiratory infections cause more than 4 million deaths per year, with influenza and tuberculosis (TB) in particular being major causes of mortality and morbidity. Although immune cell activation is critical for killing respiratory pathogens, this response must be tightly regulated to effectively control and eliminate invading microorganisms while minimizing immunopathology and maintaining pulmonary function. The distinct microenvironment of the lung is constantly patrolled by alveolar macrophages (Mφ), which are essential for tissue homeostasis, early pathogen recognition, initiation of the local immune response, and resolution of inflammation. Here, we focus on recent advances that have provided insight into the relation between pulmonary Mφ, type I interferon (IFN) signaling, and the delicate balance between protective and pathological immune responses in the lung.
Collapse
Affiliation(s)
- Maziar Divangahi
- Department of Medicine, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Meakins-Christie Laboratories, Montreal, QC, Canada; Department of Microbiology and Immunology, Microbiome and Disease Tolerance Centre, McGill University, Montreal, QC, Canada.
| | - Irah L King
- Department of Microbiology and Immunology, Microbiome and Disease Tolerance Centre, McGill University, Montreal, QC, Canada
| | - Erwan Pernet
- Department of Medicine, Department of Pathology, McGill International TB Centre, McGill University Health Centre, Meakins-Christie Laboratories, Montreal, QC, Canada
| |
Collapse
|
31
|
Venkatasubramanian S, Dhiman R, Paidipally P, Cheekatla SS, Tripathi D, Welch E, Tvinnereim AR, Jones B, Theodorescu D, Barnes PF, Vankayalapati R. A rho GDP dissociation inhibitor produced by apoptotic T-cells inhibits growth of Mycobacterium tuberculosis. PLoS Pathog 2015; 11:e1004617. [PMID: 25659138 PMCID: PMC4450061 DOI: 10.1371/journal.ppat.1004617] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/09/2014] [Indexed: 01/04/2023] Open
Abstract
In this study, we found that a subpopulation of CD4+CD25+ (85% Foxp3+) cells from persons with latent tuberculosis infection (LTBI) inhibits growth of M. tuberculosis (M. tb) in human monocyte-derived macrophages (MDMs). A soluble factor, Rho GDP dissociation inhibitor (D4GDI), produced by apoptotic CD4+CD25+ (85% Foxp3+) cells is responsible for this inhibition of M. tb growth in human macrophages and in mice. M. tb-expanded CD4+CD25+Foxp3+D4GDI+ cells do not produce IL-10, TGF-β and IFN-γ. D4GDI inhibited growth of M. tb in MDMs by enhancing production of IL-1β, TNF-α and ROS, and by increasing apoptosis of M. tb-infected MDMs. D4GDI was concentrated at the site of disease in tuberculosis patients, with higher levels detected in pleural fluid than in serum. However, in response to M. tb, PBMC from tuberculosis patients produced less D4GDI than PBMC from persons with LTBI. M. tb-expanded CD4+CD25+ (85% Foxp3+) cells and D4GDI induced intracellular M. tb to express the dormancy survival regulator DosR and DosR-dependent genes, suggesting that D4GDI induces a non-replicating state in the pathogen. Our study provides the first evidence that a subpopulation of CD4+CD25+ (85% Foxp3+) cells enhances immunity to M. tb, and that production of D4GDI by this subpopulation inhibits M. tb growth. Most people who are infected with Mycobacterium tuberculosis (M. tb) have latent tuberculosis infection (LTBI) with protective immunity. Patients with active tuberculosis have severe disease and ineffective immunity. Understanding how LTBI individuals control infection without developing disease provides important insight into the mechanisms of protective immunity against tuberculosis, and this information is essential for development of an effective vaccine. It is known that a lymphocyte population called T-cells contributes significantly to protective immunity against tuberculosis infection. In the current study, using human and murine models of M. tb infection, we found that a soluble factor, Rho GDP dissociation inhibitor (D4GDI), produced by a subpopulation of T-cells (CD4+CD25+Foxp3+) inhibits M. tb growth. We also found that D4GDI induces M. tb genes that are expressed during the non-replicative state. Our results suggest that D4GDI has a previously undescribed positive effect on immunity by enhancing host antimicrobial activity. These findings also may aid in understanding the factors that induce LTBI. Further, this information will facilitate development of improved vaccines and immunotherapeutic strategies to prevent and treat tuberculosis, respectively.
Collapse
Affiliation(s)
- Sambasivan Venkatasubramanian
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Rohan Dhiman
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Padmaja Paidipally
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Satyanarayana S. Cheekatla
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Deepak Tripathi
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Elwyn Welch
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Amy R. Tvinnereim
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Brenda Jones
- Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Dan Theodorescu
- University of Colorado Comprehensive Cancer Center, Aurora, Colorado, United States of America
| | - Peter F. Barnes
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
| | - Ramakrishna Vankayalapati
- Department of Pulmonary Immunology, Center for Biomedical Research, University of Texas Health Center, Tyler, Texas, United States of America
- * E-mail:
| |
Collapse
|
32
|
Tzelepis F, Verway M, Daoud J, Gillard J, Hassani-Ardakani K, Dunn J, Downey J, Gentile ME, Jaworska J, Sanchez AMJ, Nédélec Y, Vali H, Tabrizian M, Kristof AS, King IL, Barreiro LB, Divangahi M. Annexin1 regulates DC efferocytosis and cross-presentation during Mycobacterium tuberculosis infection. J Clin Invest 2014; 125:752-68. [PMID: 25562320 DOI: 10.1172/jci77014] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 11/13/2014] [Indexed: 01/26/2023] Open
Abstract
The phagocytosis of apoptotic cells and associated vesicles (efferocytosis) by DCs is an important mechanism for both self tolerance and host defense. Although some of the engulfment ligands involved in efferocytosis have been identified and studied in vitro, the contributions of these ligands in vivo remain ill defined. Here, we determined that during Mycobacterium tuberculosis (Mtb) infection, the engulfment ligand annexin1 is an important mediator in DC cross-presentation that increases efferocytosis in DCs and intrinsically enhances the capacity of the DC antigen-presenting machinery. Annexin1-deficient mice were highly susceptible to Mtb infection and showed an impaired Mtb antigen-specific CD8+ T cell response. Importantly, annexin1 expression was greatly downregulated in Mtb-infected human blood monocyte-derived DCs, indicating that reduction of annexin1 is a critical mechanism for immune evasion by Mtb. Collectively, these data indicate that annexin1 is essential in immunity to Mtb infection and mediates the power of DC efferocytosis and cross-presentation.
Collapse
|
33
|
Schorey JS, Cheng Y, Singh PP, Smith VL. Exosomes and other extracellular vesicles in host-pathogen interactions. EMBO Rep 2014; 16:24-43. [PMID: 25488940 DOI: 10.15252/embr.201439363] [Citation(s) in RCA: 547] [Impact Index Per Article: 49.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
An effective immune response requires the engagement of host receptors by pathogen-derived molecules and the stimulation of an appropriate cellular response. Therefore, a crucial factor in our ability to control an infection is the accessibility of our immune cells to the foreign material. Exosomes-which are extracellular vesicles that function in intercellular communication-may play a key role in the dissemination of pathogen- as well as host-derived molecules during infection. In this review, we highlight the composition and function of exosomes and other extracellular vesicles produced during viral, parasitic, fungal and bacterial infections and describe how these vesicles could function to either promote or inhibit host immunity.
Collapse
Affiliation(s)
- Jeffrey S Schorey
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Yong Cheng
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Prachi P Singh
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| | - Victoria L Smith
- Department of Biological Sciences, Eck Institute for Global Health University of Notre Dame, Notre Dame, IN, USA
| |
Collapse
|
34
|
Assis PA, Espíndola MS, Paula-Silva FWG, Rios WM, Pereira PAT, Leão SC, Silva CL, Faccioli LH. Mycobacterium tuberculosis expressing phospholipase C subverts PGE2 synthesis and induces necrosis in alveolar macrophages. BMC Microbiol 2014; 14:128. [PMID: 24886263 PMCID: PMC4057917 DOI: 10.1186/1471-2180-14-128] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 05/06/2014] [Indexed: 01/15/2023] Open
Abstract
Background Phospholipases C (PLCs) are virulence factors found in several bacteria. In Mycobacterium tuberculosis (Mtb) they exhibit cytotoxic effects on macrophages, but the mechanisms involved in PLC-induced cell death are not fully understood. It has been reported that induction of cell necrosis by virulent Mtb is coordinated by subversion of PGE2, an essential factor in cell membrane protection. Results Using two Mtb clinical isolates carrying genetic variations in PLC genes, we show that the isolate 97-1505, which bears plcA and plcB genes, is more resistant to alveolar macrophage microbicidal activity than the isolate 97-1200, which has all PLC genes deleted. The isolate 97-1505 also induced higher rates of alveolar macrophage necrosis, and likewise inhibited COX-2 expression and PGE2 production. To address the direct effect of mycobacterial PLC on cell necrosis and PGE2 inhibition, both isolates were treated with PLC inhibitors prior to macrophage infection. Interestingly, inhibition of PLCs affected the ability of the isolate 97-1505 to induce necrosis, leading to cell death rates similar to those induced by the isolate 97-1200. Finally, PGE2 production by Mtb 97-1505-infected macrophages was restored to levels similar to those produced by 97-1200-infected cells. Conclusions Mycobacterium tuberculosis bearing PLCs genes induces alveolar macrophage necrosis, which is associated to subversion of PGE2 production.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lúcia H Faccioli
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Av Cafe, s/n, Ribeirão Preto, SP 14040-903, Brazil.
| |
Collapse
|
35
|
Host-pathogen interactions during Mycobacterium tuberculosis infections. Curr Top Microbiol Immunol 2014; 374:211-41. [PMID: 23881288 DOI: 10.1007/82_2013_332] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The intimate and persistent connection between Mycobacterium tuberculosis and its human host suggests that the pathogen has evolved extensive mechanisms to evade eradication by the immune system. In particular, the organism has adapted to replicate within phagocytic cells, especially macrophages, which are specialized to kill microbes. Over the past decade of M. tuberculosis research, the means to manipulate both the organism and the host has ushered in an exciting time that has uncovered some of the mechanisms of the innate macrophage-pathogen interactions that lie at the heart of M. tuberculosis pathogenesis, though many interactions likely still await discovery. In this chapter, we will delve into some of these advances, with an emphasis on the interactions that occur on the cellular level when M. tuberculosis cells encounter macrophages. In particular, we focus on two major aspects of M. tuberculosis biology regarding the proximal physical interface between the bacterium and host, namely the interactions with the phagosomal membrane as well as the distinctive mycobacterial cell wall. Importantly, some of the emerging paradigms in M. tuberculosis pathogenesis and host response represent common themes in bacterial pathogenesis, such as the role of host cell membrane perforation in intracellular survival and host response. However, the array of unique bacterial lipid mediators and their interaction with host cells highlights the unique biology of this persistent pathogen.
Collapse
|
36
|
Singh Y, Kaul V, Mehra A, Chatterjee S, Tousif S, Dwivedi VP, Suar M, Van Kaer L, Bishai WR, Das G. Mycobacterium tuberculosis controls microRNA-99b (miR-99b) expression in infected murine dendritic cells to modulate host immunity. J Biol Chem 2013; 288:5056-61. [PMID: 23233675 PMCID: PMC3576108 DOI: 10.1074/jbc.c112.439778] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Indexed: 01/23/2023] Open
Abstract
Mycobacterium tuberculosis resides and replicates within host phagocytes by modulating host microbicidal responses. In addition, it suppresses the production of host protective cytokines to prevent activation of and antigen presentation by M. tuberculosis-infected cells, causing dysregulation of host protective adaptive immune responses. Many cytokines are regulated by microRNAs (miRNAs), a newly discovered class of small noncoding RNAs, which have been implicated in modulating host immune responses in many bacterial and viral diseases. Here, we show that miRNA-99b (miR-99b), an orphan miRNA, plays a key role in the pathogenesis of M. tuberculosis infection. We found that miR-99b expression was highly up-regulated in M. tuberculosis strain H37Rv-infected dendritic cells (DCs) and macrophages. Blockade of miR-99b expression by antagomirs resulted in significantly reduced bacterial growth in DCs. Interestingly, knockdown of miR-99b in DCs significantly up-regulated proinflammatory cytokines such as IL-6, IL-12, and IL-1β. Furthermore, mRNA and membrane-bound protein data indicated that inhibition of miR-99b augments TNF-α and TNFRSF-4 production. Thus, miR-99b targets TNF-α and TNFRSF-4 receptor genes. Treatment of anti-miR-99b-transfected DCs with anti-TNF-α antibody resulted in increased bacterial burden. Thus, our findings unveil a novel host evasion mechanism adopted by M. tuberculosis via miR-99b, which may open up new avenues for designing miRNA-based vaccines and therapies.
Collapse
Affiliation(s)
- Yogesh Singh
- From the Immunology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Vandana Kaul
- From the Immunology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Alka Mehra
- From the Immunology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
- the Division of Infectious Diseases, School of Medicine, New York University, New York, New York 10016
| | - Samit Chatterjee
- From the Immunology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
- the Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland 21218
| | - Sultan Tousif
- From the Immunology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Ved Prakash Dwivedi
- From the Immunology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| | - Mrutyunjay Suar
- the Infection Biology Laboratory, School of Biotechnology, Kalinga Institute of Industrial Technology University, Bhubaneswar 751024, Odisha, India
| | - Luc Van Kaer
- the Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, and
| | - William R. Bishai
- the Kwazulu-Natal Research Institute for Tuberculosis and HIV, Durban 4001, South Africa
| | - Gobardhan Das
- From the Immunology Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| |
Collapse
|
37
|
Divangahi M, Behar SM, Remold H. Dying to live: how the death modality of the infected macrophage affects immunity to tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:103-20. [PMID: 23468106 DOI: 10.1007/978-1-4614-6111-1_6] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Virulent Mycobacterium tuberculosis (Mtb) inhibits apoptosis and triggers necrosis of host macrophages to evade innate delay in the initiation of adaptive immunity. Necrosis is a mechanism used by bacteria to exit macrophage, evade the host defenses, and disseminate while apoptosis is associated with diminished pathogen viability. We have recently demonstrated that eicosanoids regulate cell death program of either human or murine macrophages infected with Mtb. We have defined prostaglandin E2 (PGE2) as a pro-apoptotic host lipid mediator which protects against necrosis. In contrast, lipoxin A4 (LXA4) is a pro-necrotic lipid mediator which suppresses PGE2 synthesis, resulting in mitochondrial damage and inhibition of plasma membrane repair mechanisms; this ultimately leads to the induction of necrosis. Thus, the balance between PGE2 and LXA4 determines whether Mtb-infected macrophages undergo apoptosis or necrosis and this balance determines the outcome of infection.
Collapse
Affiliation(s)
- Maziar Divangahi
- Department of Microbiology and Immunology, McGill University Health Centre, Montreal, Canada.
| | | | | |
Collapse
|
38
|
Behar SM. Antigen-specific CD8(+) T cells and protective immunity to tuberculosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 783:141-63. [PMID: 23468108 DOI: 10.1007/978-1-4614-6111-1_8] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The continuing HIV/AIDS epidemic and the spread of multi-drug resistant Mycobacterium tuberculosis has led to the perpetuation of the worldwide tuberculosis epidemic. While M. bovis BCG is widely used as a vaccine, it lacks efficacy in preventing pulmonary tuberculosis in adults [1]. To combat this ongoing scourge, vaccine development for tuberculosis is a global priority. Most infected individuals develop long-lived protective immunity, which controls and contains M. tuberculosis in a T cell-dependent manner. An effective T cells response determines whether the infection resolves or develops into clinically evident disease. Consequently, there is great interest in determining which T cells subsets mediate anti-mycobacterial immunity, delineating their effector functions, and evaluating whether vaccination can elicit these T cells subsets and induce protective immunity. CD4(+) T cells are critical for resistance to M. tuberculosis in both humans and rodent models. CD4(+) T cells are required to control the initial infection as well as to prevent recrudescence in both humans and mice [2]. While it is generally accepted that class II MHC-restricted CD4(+) T cells are essential for immunity to tuberculosis, M. tuberculosis infection elicits CD8(+) T cells responses in both people and in experimental animals. CD8(+) T cells are also recruited to the lung during M. tuberculosis infection and are found in the granulomas of infected people. Thus, how CD8(+) T cells contribute to overall immunity to tuberculosis and whether antigens recognized by CD8(+) T cells would enhance the efficacy of vaccine strategies continue to be important questions.
Collapse
Affiliation(s)
- Samuel M Behar
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
39
|
Cayabyab MJ, Macovei L, Campos-Neto A. Current and novel approaches to vaccine development against tuberculosis. Front Cell Infect Microbiol 2012; 2:154. [PMID: 23230563 PMCID: PMC3515764 DOI: 10.3389/fcimb.2012.00154] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 11/20/2012] [Indexed: 11/29/2022] Open
Abstract
Antibiotics and vaccines are the two most successful medical countermeasures that humans have created against a number of pathogens. However a select few e.g., Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB) have evaded eradication by vaccines and therapeutic approaches. TB is a global public health problem that kills 1.4 million people per year. The past decade has seen significant progress in developing new vaccine candidates, but the most fundamental questions in understanding disease progression and protective host responses that are responsible for controlling Mtb infection still remain poorly resolved. Current TB treatment requires intense chemotherapy with several antimicrobials, while the only approved vaccine is the classical viable whole-cell based Bacille-Calmette-Guerin (BCG) that protects children from severe forms of TB, but fails to protect adults. Taken together, there is a growing need to conduct basic and applied research to develop novel vaccine strategies against TB. This review is focused on the discussion surrounding current strategies and innovations being explored to discover new protective antigens, adjuvants, and delivery systems in the hopes of creating an efficacious TB vaccine.
Collapse
Affiliation(s)
- Mark J Cayabyab
- Forsyth Institute Cambridge, MA, USA ; Harvard School of Dental Medicine Boston, MA, USA
| | | | | |
Collapse
|
40
|
Prendergast KA, Kirman JR. Dendritic cell subsets in mycobacterial infection: control of bacterial growth and T cell responses. Tuberculosis (Edinb) 2012; 93:115-22. [PMID: 23167967 DOI: 10.1016/j.tube.2012.10.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 12/17/2022]
Abstract
Anti-mycobacterial immunity is guided by specialised antigen presenting cells known as dendritic cells, which are essential for both initiating and maintaining T cell immune responses during infection. The dendritic cell population can be divided into functionally distinct subsets that differ in their ability to present antigen and produce key TH1 cytokines, such as IL-12. This review discusses recent studies, in murine models, investigating which dendritic cell populations are important for mycobacterial control.
Collapse
Affiliation(s)
- Kelly A Prendergast
- Malaghan Institute of Medical Research, PO Box 7060, Newtown, Wellington 6242, New Zealand
| | | |
Collapse
|
41
|
Forrellad MA, Klepp LI, Gioffré A, Sabio y García J, Morbidoni HR, de la Paz Santangelo M, Cataldi AA, Bigi F. Virulence factors of the Mycobacterium tuberculosis complex. Virulence 2012; 4:3-66. [PMID: 23076359 PMCID: PMC3544749 DOI: 10.4161/viru.22329] [Citation(s) in RCA: 411] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The Mycobacterium tuberculosis complex (MTBC) consists of closely related species that cause tuberculosis in both humans and animals. This illness, still today, remains to be one of the leading causes of morbidity and mortality throughout the world. The mycobacteria enter the host by air, and, once in the lungs, are phagocytated by macrophages. This may lead to the rapid elimination of the bacillus or to the triggering of an active tuberculosis infection. A large number of different virulence factors have evolved in MTBC members as a response to the host immune reaction. The aim of this review is to describe the bacterial genes/proteins that are essential for the virulence of MTBC species, and that have been demonstrated in an in vivo model of infection. Knowledge of MTBC virulence factors is essential for the development of new vaccines and drugs to help manage the disease toward an increasingly more tuberculosis-free world.
Collapse
|
42
|
Doan HQ, Gulati N, Levis WR. Ingenol mebutate: potential for further development of cancer immunotherapy. J Drugs Dermatol 2012; 11:1156-1157. [PMID: 23134979 PMCID: PMC4283592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Ingenol mebutate is a diterpene ester derived from the plant Euphorbia peplus and is FDA approved for the topical treatment of actinic keratoses (AK). Shown to be efficacious with as little as a 3-day trial, this compound is being further tested for the topical treatment of other nonmelanoma skin cancers with promising preclinical data. In an effort to elucidate the molecular mechanism of this novel drug, Stahlhut et al. (2012) suggest a role for calcium and apoptosis. Further studies are needed to evaluate the intracellular mechanisms of ingenol mebutate-mediated cytotoxicity. Additionally, studies such as this not only shed light on the mechanism of ingenol mebutate and its derivatives, but also pave the way for evaluating the involvement of the immune system in eliminating drug-treated cells and tissues. This has important implications for the development of novel topical immune modulatory products and the field of topical immunotherapy.
Collapse
Affiliation(s)
- Hung Q Doan
- School of Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | | | | |
Collapse
|
43
|
Aporta A, Arbues A, Aguilo JI, Monzon M, Badiola JJ, de Martino A, Ferrer N, Marinova D, Anel A, Martin C, Pardo J. Attenuated Mycobacterium tuberculosis SO2 vaccine candidate is unable to induce cell death. PLoS One 2012; 7:e45213. [PMID: 23028853 PMCID: PMC3446966 DOI: 10.1371/journal.pone.0045213] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/13/2012] [Indexed: 11/18/2022] Open
Abstract
It has been proposed that Mycobacterium tuberculosis virulent strains inhibit apoptosis and trigger cell death by necrosis of host macrophages to evade innate immunity, while non-virulent strains induce typical apoptosis activating a protective host response. As part of the characterization of a novel tuberculosis vaccine candidate, the M. tuberculosis phoP mutant SO2, we sought to evaluate its potential to induce host cell death. The parental M. tuberculosis MT103 strain and the current vaccine against tuberculosis Bacillus Calmette-Guérin (BCG) were used as comparators in mouse models in vitro and in vivo. Our data reveal that attenuated SO2 was unable to induce apoptotic events neither in mouse macrophages in vitro nor during lung infection in vivo. In contrast, virulent MT103 triggers typical apoptotic events with phosphatidylserine exposure, caspase-3 activation and nuclear condensation and fragmentation. BCG strain behaved like SO2 and did not induce apoptosis. A clonogenic survival assay confirmed that viability of BCG- or SO2-infected macrophages was unaffected. Our results discard apoptosis as the protective mechanism induced by SO2 vaccine and provide evidence for positive correlation between classical apoptosis induction and virulent strains, suggesting apoptosis as a possible virulence determinant during M. tuberculosis infection.
Collapse
Affiliation(s)
- Adriana Aporta
- Grupo Apoptosis, Inmunidad y Cáncer, Dpto. Bioquímica y Biología Molecular y Celular, Fac. Ciencias, Universidad de Zaragoza, Zaragoza, Spain
| | - Ainhoa Arbues
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Fac. Medicina, Universidad de Zaragoza, Zaragoza, Spain
- CIBER Enfermedades respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Juan I. Aguilo
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Fac. Medicina, Universidad de Zaragoza, Zaragoza, Spain
- CIBER Enfermedades respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Monzon
- Research Centre for Encephalopathies and Transmissible Emerging Diseases, Universidad de Zaragoza, Zaragoza, Spain
| | - Juan J. Badiola
- Research Centre for Encephalopathies and Transmissible Emerging Diseases, Universidad de Zaragoza, Zaragoza, Spain
| | | | - Nadia Ferrer
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Fac. Medicina, Universidad de Zaragoza, Zaragoza, Spain
| | - Dessislava Marinova
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Fac. Medicina, Universidad de Zaragoza, Zaragoza, Spain
- CIBER Enfermedades respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Anel
- Grupo Apoptosis, Inmunidad y Cáncer, Dpto. Bioquímica y Biología Molecular y Celular, Fac. Ciencias, Universidad de Zaragoza, Zaragoza, Spain
| | - Carlos Martin
- Grupo de Genética de Micobacterias, Dpto. Microbiología, Medicina Preventiva y Salud Pública, Fac. Medicina, Universidad de Zaragoza, Zaragoza, Spain
- CIBER Enfermedades respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Servicio de Microbiología, Hospital Universitario Miguel Servet, IIS Aragón, Zaragoza, Spain
| | - Julian Pardo
- Fundación Aragón I+D (ARAID), Gobierno de Aragón, Zaragoza, Spain
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza, Zaragoza, Spain
- Grupo Inmunidad Celular Efectora (ICE), Dpto. Bioquímica y Biología Molecular y Celular, Fac. Ciencias, Universidad de Zaragoza/IIS Aragón, Zaragoza, Spain
- * E-mail:
| |
Collapse
|
44
|
Uhlin M, Andersson J, Zumla A, Maeurer M. Adjunct Immunotherapies for Tuberculosis. J Infect Dis 2012; 205 Suppl 2:S325-34. [DOI: 10.1093/infdis/jis197] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|