1
|
Barbero AM, Hernández Del Pino RE, Fuentes F, Barrionuevo P, Pasquinelli V. Platelets promote human macrophages-mediated macropinocytosis of Clostridioides difficile. Front Cell Infect Microbiol 2024; 13:1252509. [PMID: 38249298 PMCID: PMC10796631 DOI: 10.3389/fcimb.2023.1252509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/11/2023] [Indexed: 01/23/2024] Open
Abstract
Clostridioides difficile is the main causative agent of hospital-acquired diarrhea and the potentially lethal disease, C. difficile infection. The cornerstone of the current therapy is the use of antibiotics, which is not fully effective. The molecular mechanisms, inflammatory conditions and host-immune responses that could benefit the persistence or elimination of C. difficile remain unclear. Macrophages perform different ways of endocytosis as part of their immune surveillance functions and platelets, classically known for their coagulatory role, are also important modulators of the immune system. The aim of this study was to evaluate the endocytosis of vegetative C. difficile by human macrophages and the involvement of platelets in this process. Our results showed that both macrophages and platelets interact with live and heat-killed C. difficile. Furthermore, platelets form complexes with human monocytes in healthy donor's fresh blood and the presence of C. difficile increased these cell-cell interactions. Using flow cytometry and confocal microscopy, we show that macrophages can internalize C. difficile and that platelets improve this uptake. By using inhibitors of different endocytic pathways, we demonstrate that macropinocytosis is the route of entry of C. difficile into the cell. Taken together, our findings are the first evidence for the internalization of vegetative non-toxigenic and hypervirulent C. difficile by human macrophages and highlight the role of platelets in innate immunity during C. difficile infection. Deciphering the crosstalk of C. difficile with immune cells could provide new tools for understanding the pathogenesis of C. difficile infection and for the development of host-directed therapies.
Collapse
Affiliation(s)
- Angela María Barbero
- Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA), Buenos Aires, Argentina
- Centro de Investigaciones y Transferencias del Noroeste de la Provincia de Buenos Aires (CIT NOBA), UNNOBA-Universidad Nacional de San Antonio de Areco (UNSAdA)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Rodrigo Emanuel Hernández Del Pino
- Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA), Buenos Aires, Argentina
- Centro de Investigaciones y Transferencias del Noroeste de la Provincia de Buenos Aires (CIT NOBA), UNNOBA-Universidad Nacional de San Antonio de Areco (UNSAdA)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Federico Fuentes
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Paula Barrionuevo
- Instituto de Medicina Experimental (CONICET-Academia Nacional de Medicina), Buenos Aires, Argentina
| | - Virginia Pasquinelli
- Centro de Investigaciones Básicas y Aplicadas (CIBA), Universidad Nacional del Noroeste de la Provincia de Buenos Aires (UNNOBA), Buenos Aires, Argentina
- Centro de Investigaciones y Transferencias del Noroeste de la Provincia de Buenos Aires (CIT NOBA), UNNOBA-Universidad Nacional de San Antonio de Areco (UNSAdA)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
2
|
Yang Q, Qi F, Ye T, Li J, Xu G, He X, Deng G, Zhang P, Liao M, Qiao K, Zhang Z. The interaction of macrophages and CD8 T cells in bronchoalveolar lavage fluid is associated with latent tuberculosis infection. Emerg Microbes Infect 2023:2239940. [PMID: 37470432 PMCID: PMC10399483 DOI: 10.1080/22221751.2023.2239940] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/21/2023]
Abstract
Mycobacterium tuberculosis (Mtb) infection, including active tuberculosis (TB) and latent Mtb infection (LTBI), leads to diverse outcomes owing to different host immune responses. However, the immune mechanisms that govern the progression from LTBI to TB remain poorly defined in humans. Here, we profiled the lung immune cell populations within the bronchoalveolar lavage fluid (BALF) from patients with LTBI or TB using single-cell RNA sequencing (scRNA-seq). We found that Mtb infection substantially changed the immune cell compartments in the BALF, especially for the three subsets of macrophages, monocyte macrophage (MM)-CCL23, MM-FCN1, and MM-SPP1, which were found to be associated with the disease status of TB infection. Notably, MM-CCL23 cells derived from monocytes after stimulation with Mtb were characterized by high levels of chemokine (CCL23 and CXCL5) production and might serve as a marker for Mtb infection. The MM-CCL23 population mainly recruited CD8-CCR6 T cells through CCL20/CCR6, which was a prominent feature associated with protection immunity in LTBI. This study improves our understanding of the lung immune landscape during Mtb infection, which may inform future vaccine design for protective immunity.
Collapse
Affiliation(s)
- Qianting Yang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| | - Furong Qi
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Taosheng Ye
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
- Department of Respiratory endoscopy, Shenzhen Third People's Hospital, Shenzhen, China
| | - Jinpei Li
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
- Department of Respiratory endoscopy, Shenzhen Third People's Hospital, Shenzhen, China
| | - Gang Xu
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Xiaomeng He
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Guofang Deng
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
- Department of Pulmonary Medicine & Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, China
| | - Peize Zhang
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
- Department of Pulmonary Medicine & Tuberculosis, Shenzhen Third People's Hospital, Shenzhen, China
| | - Mingfeng Liao
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| | - Kun Qiao
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
- Department of Thoracic Surgery, Shenzhen Third People's Hospital, Shenzhen, China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital; The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
- Shenzhen Clinical Research Center for Tuberculosis, Shenzhen, China
| |
Collapse
|
3
|
Urbán-Solano A, Flores-Gonzalez J, Cruz-Lagunas A, Pérez-Rubio G, Buendia-Roldan I, Ramón-Luing LA, Chavez-Galan L. High levels of PF4, VEGF-A, and classical monocytes correlate with the platelets count and inflammation during active tuberculosis. Front Immunol 2022; 13:1016472. [PMID: 36325331 PMCID: PMC9618821 DOI: 10.3389/fimmu.2022.1016472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
Platelets play a major role in coagulation and hemostasis; evidence supports the hypothesis that they also contribute to immunological processes. Increased platelet counts have been associated with poor prognosis in tuberculosis (TB). Platelet–monocyte aggregates have been reported in patients with TB, but it is still unclear if only one monocyte subpopulation is correlated to the platelet count; moreover, the platelet–monocyte axis has not been studied during latent tuberculosis (LTB). In this study, mononuclear cells and plasma were obtained from patients diagnosed with active drug-sensitive TB (DS-TB, n = 10) and LTB (n = 10); cytokines and growth factors levels associated to platelets were evaluated, and correlations with monocyte subpopulations were performed to identify a relationship between them, as well as an association with the degree of lung damage. Our data showed that, compared to LTB, DS-TB patients had an increased frequency of platelets, monocytes, and neutrophils. Although DS-TB patients showed no significant difference in the frequency of classical and non-classical monocytes, the classical monocytes had increased CD14 intensity of expression and frequency of TLR-2+. Furthermore, the plasma levels of angiogenic factors such as vascular endothelial growth factor (VEGF-A), platelet-derived growth factor (PDGF-BB), and platelet factor-4 (PF4), and pro-inflammatory cytokines like interleukin 6 (IL-6), interleukin 1 beta (IL-1β), and interferon-γ-inducible protein 10 (IP-10) were increased in DS-TB patients. In addition, PF-4 and VEGF-A correlated positively with the frequency of classical monocytes and the platelet count. Using a principal component analysis, we identified four groups of DS-TB patients according to their levels of pro-inflammatory cytokines, angiogenic factors, and degree of lung damage. This study establishes that there is a correlation between VEGF-A and PF4 with platelets and classical monocytes during active TB, suggesting that those cell subpopulations are the major contributors of these molecules, and together, they control the severity of lung damage by amplification of the inflammatory environment.
Collapse
Affiliation(s)
- Alexia Urbán-Solano
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Julio Flores-Gonzalez
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetic, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Gloria Pérez-Rubio
- HLA Laboratory, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Ivette Buendia-Roldan
- Translational Research Laboratory on Aging and Pulmonary Fibrosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Lucero A. Ramón-Luing
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Leslie Chavez-Galan
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- *Correspondence: Leslie Chavez-Galan, ;
| |
Collapse
|
4
|
Ahmad F, Rani A, Alam A, Zarin S, Pandey S, Singh H, Hasnain SE, Ehtesham NZ. Macrophage: A Cell With Many Faces and Functions in Tuberculosis. Front Immunol 2022; 13:747799. [PMID: 35603185 PMCID: PMC9122124 DOI: 10.3389/fimmu.2022.747799] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 03/30/2022] [Indexed: 01/16/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) is the causative agent of human tuberculosis (TB) which primarily infects the macrophages. Nearly a quarter of the world's population is infected latently by Mtb. Only around 5%-10% of those infected develop active TB disease, particularly during suppressed host immune conditions or comorbidity such as HIV, hinting toward the heterogeneity of Mtb infection. The aerosolized Mtb first reaches the lungs, and the resident alveolar macrophages (AMs) are among the first cells to encounter the Mtb infection. Evidence suggests that early clearance of Mtb infection is associated with robust innate immune responses in resident macrophages. In addition to lung-resident macrophage subsets, the recruited monocytes and monocyte-derived macrophages (MDMs) have been suggested to have a protective role during Mtb infection. Mtb, by virtue of its unique cell surface lipids and secreted protein effectors, can evade killing by the innate immune cells and preferentially establish a niche within the AMs. Continuous efforts to delineate the determinants of host defense mechanisms have brought to the center stage the crucial role of macrophage phenotypical variations for functional adaptations in TB. The morphological and functional heterogeneity and plasticity of the macrophages aid in confining the dissemination of Mtb. However, during a suppressed or hyperactivated immune state, the Mtb virulence factors can affect macrophage homeostasis which may skew to favor pathogen growth, causing active TB. This mini-review is aimed at summarizing the interplay of Mtb pathomechanisms in the macrophages and the implications of macrophage heterogeneity and plasticity during Mtb infection.
Collapse
Affiliation(s)
- Faraz Ahmad
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Anshu Rani
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Anwar Alam
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Sheeba Zarin
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| | - Saurabh Pandey
- Department of Biochemistry, Jamia Hamdard, New Delhi, India
| | - Hina Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
| | - Seyed Ehtesham Hasnain
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi (IIT-D), New Delhi, India
- Department of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, India
| | - Nasreen Zafar Ehtesham
- Laboratory of Infection Biology and Cell Signaling, Indian Council of Medical Research (ICMR)-National Institute of Pathology, New Delhi, India
| |
Collapse
|
5
|
La Manna MP, Orlando V, Badami GD, Tamburini B, Azgomi MS, Presti EL, Del Nonno F, Petrone L, Belmonte B, Falasca L, Carlo PD, Dieli F, Goletti D, Caccamo N. Platelets accumulate in lung lesions of tuberculosis patients and inhibit T-cell responses and Mycobacterium tuberculosis replication in macrophages. Eur J Immunol 2022; 52:784-799. [PMID: 35338775 PMCID: PMC9325462 DOI: 10.1002/eji.202149549] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/24/2021] [Accepted: 03/23/2022] [Indexed: 12/05/2022]
Abstract
Platelets regulate human inflammatory responses that lead to disease. However, the role of platelets in tuberculosis (TB) pathogenesis is still unclear. Here, we show that patients with active TB have a high number of platelets in peripheral blood and a low number of lymphocytes leading to a high platelets to lymphocytes ratio (PL ratio). Moreover, the serum concentration of different mediators promoting platelet differentiation or associated with platelet activation is increased in active TB. Immunohistochemistry analysis shows that platelets localise around the lung granuloma lesions in close contact with T lymphocytes and macrophages. Transcriptomic analysis of caseous tissue of human pulmonary TB granulomas, followed by Gene Ontology analysis, shows that 53 platelet activation‐associated genes are highly expressed compared to the normal lung tissue. In vitro activated platelets (or their supernatants) inhibit BCG‐induced T‐ lymphocyte proliferation and IFN‐γ production. Likewise, platelets inhibit the growth of intracellular macrophages of Mycobacterium (M.) tuberculosis. Soluble factors released by activated platelets mediate both immunological and M. tuberculosis replication activities. Furthermore, proteomic and neutralisation studies (by mAbs) identify TGF‐β and PF4 as the factors responsible for inhibiting T‐cell response and enhancing the mycobactericidal activity of macrophages, respectively. Altogether these results highlight the importance of platelets in TB pathogenesis.
Collapse
Affiliation(s)
- Marco P La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Giusto D Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Elena Lo Presti
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Franca Del Nonno
- Pathology Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Linda Petrone
- Translational research Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - Laura Falasca
- Pathology Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Paola Di Carlo
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Delia Goletti
- Translational research Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| |
Collapse
|
6
|
Cronan MR. In the Thick of It: Formation of the Tuberculous Granuloma and Its Effects on Host and Therapeutic Responses. Front Immunol 2022; 13:820134. [PMID: 35320930 PMCID: PMC8934850 DOI: 10.3389/fimmu.2022.820134] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The defining pathology of tuberculosis is the granuloma, an organized structure derived from host immune cells that surrounds infecting Mycobacterium tuberculosis. As the location of much of the bacteria in the infected host, the granuloma is a central point of interaction between the host and the infecting bacterium. This review describes the signals and cellular reprogramming that drive granuloma formation. Further, as a central point of host-bacterial interactions, the granuloma shapes disease outcome by altering host immune responses and bacterial susceptibility to antibiotic treatment, as discussed herein. This new understanding of granuloma biology and the signaling behind it highlights the potential for host-directed therapies targeting the granuloma to enhance antibiotic access and tuberculosis-specific immune responses.
Collapse
Affiliation(s)
- Mark R. Cronan
-
In Vivo Cell Biology of Infection Group, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
7
|
Kirwan DE, Chong DLW, Friedland JS. Platelet Activation and the Immune Response to Tuberculosis. Front Immunol 2021; 12:631696. [PMID: 34093524 PMCID: PMC8170316 DOI: 10.3389/fimmu.2021.631696] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/04/2021] [Indexed: 12/24/2022] Open
Abstract
In 2019 10 million people developed symptomatic tuberculosis (TB) disease and 1.2 million died. In active TB the inflammatory response causes tissue destruction, which leads to both acute morbidity and mortality. Tissue destruction in TB is driven by host innate immunity and mediated via enzymes, chiefly matrix metalloproteinases (MMPs) which are secreted by leukocytes and stromal cells and degrade the extracellular matrix. Here we review the growing evidence implicating platelets in TB immunopathology. TB patients typically have high platelet counts, which correlate with disease severity, and a hypercoagulable profile. Platelets are present in human TB granulomas and platelet-associated gene transcripts are increased in TB patients versus healthy controls. Platelets most likely drive TB immunopathology through their effect on other immune cells, particularly monocytes, to lead to upregulation of activation markers, increased MMP secretion, and enhanced phagocytosis. Finally, we consider current evidence supporting use of targeted anti-platelet agents in the treatment of TB due to growing interest in developing host-directed therapies to limit tissue damage and improve treatment outcomes. In summary, platelets are implicated in TB disease and contribute to MMP-mediated tissue damage via their cellular interactions with other leukocytes, and are potential targets for novel host-directed therapies.
Collapse
Affiliation(s)
- Daniela E Kirwan
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Deborah L W Chong
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Jon S Friedland
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| |
Collapse
|
8
|
Venous Thromboembolic Disease in Chronic Inflammatory Lung Diseases: Knowns and Unknowns. J Clin Med 2021; 10:jcm10102061. [PMID: 34064992 PMCID: PMC8151562 DOI: 10.3390/jcm10102061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 12/13/2022] Open
Abstract
Persistent inflammation within the respiratory tract underlies the pathogenesis of numerous chronic pulmonary diseases. There is evidence supporting that chronic lung diseases are associated with a higher risk of venous thromboembolism (VTE). However, the relationship between lung diseases and/or lung function with VTE is unclear. Understanding the role of chronic lung inflammation as a predisposing factor for VTE may help determine the optimal management and aid in the development of future preventative strategies. We aimed to provide an overview of the relationship between the most common chronic inflammatory lung diseases and VTE. Asthma, chronic obstructive pulmonary disease, interstitial lung diseases, or tuberculosis increase the VTE risk, especially pulmonary embolism (PE), compared to the general population. However, high suspicion is needed to diagnose a thrombotic event early as the clinical presentation inevitably overlaps with respiratory disorders. PE risk increases with disease severity and exacerbations. Hence, hospitalized patients should be considered for thromboprophylaxis administration. Conversely, all VTE patients should be asked for lung comorbidities before determining anticoagulant therapy duration, as those patients are at increased risk of recurrent PE episodes rather than DVT. Further research is needed to understand the underlying pathophysiology of in-situ thrombosis in those patients.
Collapse
|
9
|
Fu G, Deng M, Neal MD, Billiar TR, Scott MJ. Platelet-Monocyte Aggregates: Understanding Mechanisms and Functions in Sepsis. Shock 2021; 55:156-166. [PMID: 32694394 PMCID: PMC8008955 DOI: 10.1097/shk.0000000000001619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
ABSTRACT Platelets have been shown to play an important immunomodulatory role in the pathogenesis of various diseases through their interactions with other immune and nonimmune cells. Sepsis is a major cause of death in the United States, and many of the mechanisms driving sepsis pathology are still unresolved. Monocytes have recently received increasing attention in sepsis pathogenesis, and multiple studies have associated increased levels of platelet-monocyte aggregates observed early in sepsis with clinical outcomes in sepsis patients. These findings suggest platelet-monocyte aggregates may be an important prognostic indicator. However, the mechanisms leading to platelet interaction and aggregation with monocytes, and the effects of aggregation during sepsis are still poorly defined. There are few studies that have really investigated functions of platelets and monocytes together, despite a large body of research showing separate functions of platelets and monocytes in inflammation and immune responses during sepsis. The goal of this review is to provide insights into what we do know about mechanisms and biological meanings of platelet-monocyte interactions, as well as some of the technical challenges and limitations involved in studying this important potential mechanism in sepsis pathogenesis. Improving our understanding of platelet and monocyte biology in sepsis may result in identification of novel targets that can be used to positively affect outcomes in sepsis.
Collapse
Affiliation(s)
- Guang Fu
- Department of General Surgery, The 3rd Xiangya Hospital, Central South University, Changsha, Hunan, China (visiting scholar in Pittsburgh 2018-09/2020-09)
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meihong Deng
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew D. Neal
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Melanie J. Scott
- Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
- Pittsburgh Trauma Research Center, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
10
|
Torres-Juarez F, Trejo-Martínez LA, Layseca-Espinosa E, Leon-Contreras JC, Enciso-Moreno JA, Hernandez-Pando R, Rivas-Santiago B. Platelets immune response against Mycobacterium tuberculosis infection. Microb Pathog 2021; 153:104768. [PMID: 33524564 DOI: 10.1016/j.micpath.2021.104768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 12/23/2022]
Abstract
Tuberculosis (TB) is the first cause of death by a single infectious agent. Previous reports have highlighted the presence of platelets within Tb granulomas, albeit the immune-associated platelet response to Mycobacterium tuberculosis (Mtb) has not been deeply studied. Our results showed that platelets are recruited into the granuloma in the late stages of tuberculosis. Furthermore, electron-microscopy studies showed that platelets can internalize Mtb and produce host defense peptides (HDPs), such as RNase 7, HBD2 and hPF-4 that bind to the internalized Mtb. Mtb-infected platelets exhibited higher transcription and secretion of IL-1β and TNF-α, whereas IL-10 and IL-6 protein levels decreased. These results suggest that platelets participate in the immune response against Mtb through HDPs and cytokines production.
Collapse
Affiliation(s)
- Flor Torres-Juarez
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico; Laboratory of Immunology, Autonomous University of San Luis Potosí, San Luis Potosi, Mexico
| | - Luis A Trejo-Martínez
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico
| | | | - Juan C Leon-Contreras
- Laboratory of Experimental Pathology, Nacional Institute of Medical Sciences and Nutrition "Salvador Zubiran", CDMX, Mexico
| | - Jose A Enciso-Moreno
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico
| | - Rogelio Hernandez-Pando
- Laboratory of Experimental Pathology, Nacional Institute of Medical Sciences and Nutrition "Salvador Zubiran", CDMX, Mexico
| | - Bruno Rivas-Santiago
- Biomedical Research Unit of Zacatecas-Mexican Institute of Social Security, Zacatecas, Mexico.
| |
Collapse
|
11
|
Scheuermann L, Pei G, Domaszewska T, Zyla J, Oberbeck-Müller D, Bandermann S, Feng Y, Ruiz Moreno JS, Opitz B, Mollenkopf HJ, Kaufmann SHE, Dorhoi A. Platelets Restrict the Oxidative Burst in Phagocytes and Facilitate Primary Progressive Tuberculosis. Am J Respir Crit Care Med 2020; 202:730-744. [PMID: 32421376 DOI: 10.1164/rccm.201910-2063oc] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Rationale: Platelets are generated in the capillaries of the lung, control hemostasis, and display immunological functions. Tuberculosis primarily affects the lung, and patients show platelet changes and hemoptysis. A role of platelets in immunopathology of pulmonary tuberculosis requires careful assessment.Objectives: To identify the dynamics and interaction partners of platelets in the respiratory tissue and establish their impact on the outcome of pulmonary tuberculosis.Methods: Investigations were primarily performed in murine models of primary progressive pulmonary tuberculosis, by analysis of mouse strains with variable susceptibility to Mycobacterium tuberculosis infection using platelet depletion and delivery of antiplatelet drugs.Measurements and Main Results: Platelets were present at the site of infection and formed aggregates with different myeloid subsets during experimental tuberculosis. Such aggregates were also detected in patients with tuberculosis. Platelets were detrimental during the early phase of infection, and this effect was uncoupled from their canonical activation. Platelets left lung cell dynamics and patterns of antimycobacterial T-cell responses unchanged but hampered antimicrobial defense by restricting production of reactive oxygen species in lung-residing myeloid cells.Conclusions: Platelets are detrimental in primary progressive pulmonary tuberculosis, orchestrate lung immunity by modulating innate immune responsiveness, and may be amenable to new interventions for this deadly disease.
Collapse
Affiliation(s)
| | - Gang Pei
- Immunology Department and.,Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany
| | | | - Joanna Zyla
- Immunology Department and.,Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | | | | | - Yonghong Feng
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juan Sebastian Ruiz Moreno
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Bastian Opitz
- Department of Internal Medicine/Infectious Diseases and Pulmonary Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Stefan H E Kaufmann
- Immunology Department and.,Hagler Institute for Advanced Study at Texas A&M University, College Station, Texas; and
| | - Anca Dorhoi
- Immunology Department and.,Institute of Immunology, Friedrich-Loeffler-Institut, Greifswald - Insel Riems, Germany.,Faculty of Mathematics and Natural Sciences, University of Greifswald, Greifswald, Germany
| |
Collapse
|
12
|
Teklu T, Wondale B, Taye B, Hailemariam M, Bekele S, Tamirat M, Zewude A, Mohamed T, Medhin G, Legesse M, Yu Y, Ameni G, Pieper R. Differences in plasma proteomes for active tuberculosis, latent tuberculosis and non-tuberculosis mycobacterial lung disease patients with and without ESAT-6/CFP10 stimulation. Proteome Sci 2020; 18:10. [PMID: 33292280 PMCID: PMC7603755 DOI: 10.1186/s12953-020-00165-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 09/24/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Tuberculosis (TB) is one of the world's most problematic infectious diseases. The pathogen Mycobacterium tuberculosis (Mtb) is contained by the immune system in people with latent TB infection (LTBI). No overt disease symptoms occur. The environmental and internal triggers leading to reactivation of TB are not well understood. Non-tuberculosis Mycobacteria (NTM) can also cause TB-like lung disease. Comparative analysis of blood plasma proteomes from subjects afflicted by these pathologies in an endemic setting may yield new differentiating biomarkers and insights into inflammatory and immunological responses to Mtb and NTM. METHODS Blood samples from 40 human subjects in a pastoral region of Ethiopia were treated with the ESAT-6/CFP-10 antigen cocktail to stimulate anti-Mtb and anti-NTM immune responses. In addition to those of active TB, LTBI, and NTM cohorts, samples from matched healthy control (HC) subjects were available. Following the generation of sample pools, proteomes were analyzed via LC-MS/MS. These experiments were also performed without antigen stimulation steps. Statistically significant differences using the Z-score method were determined and interpreted in the context of the proteins' functions and their contributions to biological pathways. RESULTS More than 200 proteins were identified from unstimulated and stimulated plasma samples (UPSs and SPSs, respectively). Thirty-four and 64 proteins were differentially abundant with statistical significance (P < 0.05; Benjamini-Hochberg correction with an FDR < 0.05) comparing UPS and SPS proteomic data of four groups, respectively. Bioinformatics analysis of such proteins via the Gene Ontology Resource was indicative of changes in cellular and metabolic processes, responses to stimuli, and biological regulations. The m7GpppN-mRNA hydrolase was increased in abundance in the LTBI group compared to HC subjects. Charged multivesicular body protein 4a and platelet factor-4 were increased in abundance in NTM as compared to HC and decreased in abundance in NTM as compared to active TB. C-reactive protein, α-1-acid glycoprotein 1, sialic acid-binding Ig-like lectin 16, and vitamin K-dependent protein S were also increased (P < 0.05; fold changes≥2) in SPSs and UPSs comparing active TB with LTBI and NTM cases. These three proteins, connected in a STRING functional network, contribute to the acute phase response and influence blood coagulation. CONCLUSION Plasma proteomes are different comparing LTBI, TB, NTM and HC cohorts. The changes are augmented following prior blood immune cell stimulation with the ESAT-6/CFP-10 antigen cocktail. The results encourage larger-cohort studies to identify specific biomarkers to diagnose NTM infection, LTBI, and to predict the risk of TB reactivation.
Collapse
Affiliation(s)
- Takele Teklu
- Department of Immunology and Molecular Biology, University of Gondar, Gondar, Ethiopia.
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia.
| | - Biniam Wondale
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Biology, Arba Minch University, Arba Minch, Ethiopia
| | - Biruhalem Taye
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- European Molecular Biology Laboratory, Notkestraβe 85, 22607, Hamburg, Germany
| | | | | | - Mesfin Tamirat
- Laboratory department, Jinka General Hospital, Jinka, Ethiopia
| | - Aboma Zewude
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- Ethiopian Public health Institute, P.O box 1242, Addis Ababa, Ethiopia
| | - Temesgen Mohamed
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Girmay Medhin
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Mengistu Legesse
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Yanbao Yu
- J. Craig Venter Institute, Rockville, MD, USA
| | - Gobena Ameni
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- Department of Veterinary Medicine, College of Food and Agriculture, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | | |
Collapse
|
13
|
Barbosa-Lima G, Hottz ED, de Assis EF, Liechocki S, Souza TML, Zimmerman GA, Bozza FA, Bozza PT. Dengue virus-activated platelets modulate monocyte immunometabolic response through lipid droplet biogenesis and cytokine signaling. J Leukoc Biol 2020; 108:1293-1306. [PMID: 32663907 DOI: 10.1002/jlb.4ma0620-658r] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 01/09/2023] Open
Abstract
Dengue is characterized as one of the most important arthropod-borne human viral diseases, representing a public health problem. Increased activation of immune cells is involved in the progression of infection to severe forms. Recently, our group demonstrated the contribution of platelet-monocyte interaction to inflammatory responses in dengue, adding to evolving evidence that platelets have inflammatory functions and can regulate different aspects of innate immune responses. Furthermore, stimuli-specific-activated platelets can promote phenotypic changes and metabolic reprogramming in monocytes. Thus, this study aimed to evaluate the roles of dengue virus (DENV)-activated platelets on immunometabolic reprogramming of monocytes in vitro, focusing on lipid droplet (LD) biogenesis. We demonstrated that platelets exposed to DENV in vitro form aggregates with monocytes and signal to LD formation and CXCL8/IL-8, IL-10, CCL2, and PGE2 secretion. Pharmacologic inhibition of LD biogenesis prevents PGE2 secretion, but not CXCL8/IL-8 release, by platelet-monocyte complexes. In exploring the mechanisms involved, we demonstrated that LD formation in monocytes exposed to DENV-activated platelets is partially dependent on platelet-produced MIF. Additionally, LD formation is higher in monocytes, which have platelets adhered on their surface, suggesting that beyond paracrine signaling, platelet adhesion is an important event in platelet-mediated modulation of lipid metabolism in monocytes. Together, our results demonstrate that activated platelets aggregate with monocytes during DENV infection and signal to LD biogenesis and the secretion of inflammatory mediators, which may contribute to dengue immunopathogenesis.
Collapse
Affiliation(s)
- Giselle Barbosa-Lima
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Eugenio D Hottz
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil.,Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora (UFJF), Juiz de Fora, MG, Brazil
| | - Edson F de Assis
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Sally Liechocki
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Thiago Moreno L Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil.,National Institute for Science and Technology on Innovation on Neglected Diseases (INCT/IDN), Center for Technological Development in Health (CDTS), Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| | - Guy A Zimmerman
- Molecular Medicine Program and Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Fernando A Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil.,Intensive Medicine Laboratory, National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil.,D'Or Institute of Research, Rio de Janeiro, RJ, Brazil
| | - Patricia T Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
14
|
Coakley G, Volpe B, Bouchery T, Shah K, Butler A, Geldhof P, Hatherill M, Horsnell WGC, Esser-von Bieren J, Harris NL. Immune serum-activated human macrophages coordinate with eosinophils to immobilize Ascaris suum larvae. Parasite Immunol 2020; 42:e12728. [PMID: 32394439 DOI: 10.1111/pim.12728] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/19/2022]
Abstract
Helminth infection represents a major health problem causing approximately 5 million disability-adjusted life years worldwide. Concerns that repeated anti-helminthic treatment may lead to drug resistance render it important that vaccines are developed but will require increased understanding of the immune-mediated cellular and antibody responses to helminth infection. IL-4 or antibody-activated murine macrophages are known to immobilize parasitic nematode larvae, but few studies have addressed whether this is translatable to human macrophages. In the current study, we investigated the capacity of human macrophages to recognize and attack larval stages of Ascaris suum, a natural porcine parasite that is genetically similar to the human helminth Ascaris lumbricoides. Human macrophages were able to adhere to and trap A suum larvae in the presence of either human or pig serum containing Ascaris-specific antibodies and other factors. Gene expression analysis of serum-activated macrophages revealed that CCL24, a potent eosinophil attractant, was the most upregulated gene following culture with A suum larvae in vitro, and human eosinophils displayed even greater ability to adhere to, and trap, A suum larvae. These data suggest that immune serum-activated macrophages can recruit eosinophils to the site of infection, where they act in concert to immobilize tissue-migrating Ascaris larvae.
Collapse
Affiliation(s)
- Gillian Coakley
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Beatrice Volpe
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Tiffany Bouchery
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Kathleen Shah
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| | - Alana Butler
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Peter Geldhof
- Department of Virology, Parasitology and Immunology, Laboratory of Parasitology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Mark Hatherill
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - William G C Horsnell
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Julia Esser-von Bieren
- Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland.,Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Nicola Laraine Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Global Health Institute, Swiss Federal Institute of Technology, Lausanne, Switzerland
| |
Collapse
|
15
|
Hortle E, Oehlers SH. Host-directed therapies targeting the tuberculosis granuloma stroma. Pathog Dis 2020; 78:5800987. [DOI: 10.1093/femspd/ftaa015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
ABSTRACT
Mycobacteria have co-evolved with their hosts resulting in pathogens adept at intracellular survival. Pathogenic mycobacteria actively manipulate infected macrophages to drive granuloma formation while subverting host cell processes to create a permissive niche. Granuloma residency confers phenotypic antimicrobial resistance by physically excluding or neutralising antibiotics. Host-directed therapies (HDTs) combat infection by restoring protective immunity and reducing immunopathology independent of pathogen antimicrobial resistance status. This review covers innovative research that has discovered ‘secondary’ symptoms of infection in the granuloma stroma are actually primary drivers of infection and that relieving these stromal pathologies with HDTs benefits the host. Advances in our understanding of the relationship between tuberculosis and the host vasculature, haemostatic system and extracellular matrix reorganisation are discussed. Preclinical and clinical use of HDTs against these stromal targets are summarised.
Collapse
Affiliation(s)
- Elinor Hortle
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
- The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW 2050, Australia
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney, Camperdown, NSW 2050, Australia
- The University of Sydney, Faculty of Medicine and Health & Marie Bashir Institute, Camperdown, NSW 2050, Australia
| |
Collapse
|
16
|
Fox KA, Kirwan DE, Whittington AM, Krishnan N, Robertson BD, Gilman RH, López JW, Singh S, Porter JC, Friedland JS. Platelets Regulate Pulmonary Inflammation and Tissue Destruction in Tuberculosis. Am J Respir Crit Care Med 2019; 198:245-255. [PMID: 29420060 DOI: 10.1164/rccm.201710-2102oc] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
RATIONALE Platelets may interact with the immune system in tuberculosis (TB) to regulate human inflammatory responses that lead to morbidity and spread of infection. OBJECTIVES To identify a functional role of platelets in the innate inflammatory and matrix-degrading response in TB. METHODS Markers of platelet activation were examined in plasma from 50 patients with TB before treatment and 50 control subjects. Twenty-five patients were followed longitudinally. Platelet-monocyte interactions were studied in a coculture model infected with live, virulent Mycobacterium tuberculosis (M.tb) and dissected using qRT-PCR, Luminex multiplex arrays, matrix degradation assays, and colony counts. Immunohistochemistry detected CD41 (cluster of differentiation 41) expression in a pulmonary TB murine model, and secreted platelet factors were measured in BAL fluid from 15 patients with TB and matched control subjects. MEASUREMENTS AND MAIN RESULTS Five of six platelet-associated mediators were upregulated in plasma of patients with TB compared with control subjects, with concentrations returning to baseline by Day 60 of treatment. Gene expression of the monocyte collagenase MMP-1 (matrix metalloproteinase-1) was upregulated by platelets in M.tb infection. Platelets also enhanced M.tb-induced MMP-1 and -10 secretion, which drove type I collagen degradation. Platelets increased monocyte IL-1 and IL-10 and decreased IL-12 and MDC (monocyte-derived chemokine; also known as CCL-22) secretion, as consistent with an M2 monocyte phenotype. Monocyte killing of intracellular M.tb was decreased. In the lung, platelets were detected in a TB mouse model, and secreted platelet mediators were upregulated in human BAL fluid and correlated with MMP and IL-1β concentrations. CONCLUSIONS Platelets drive a proinflammatory, tissue-degrading phenotype in TB.
Collapse
Affiliation(s)
- Katharine A Fox
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Daniela E Kirwan
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Ashley M Whittington
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Nitya Krishnan
- 2 Medical Research Council Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, United Kingdom
| | - Brian D Robertson
- 2 Medical Research Council Centre for Molecular Bacteriology and Infection, Department of Medicine, Imperial College London, United Kingdom
| | - Robert H Gilman
- 3 Department of International Health, Johns Hopkins University, Baltimore Maryland.,4 Laboratorio de Investigación en Enfermedades Infecciosas, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - José W López
- 5 Laboratorio de Bioinformática y Biología Molecular, Universidad Peruana Cayetano Heredia, Lima, Peru.,6 Instituto Nacional de Salud del Niño, Lima, Peru; and
| | - Shivani Singh
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| | - Joanna C Porter
- 7 Centre for Inflammation and Tissue Repair, Respiratory Medicine, University College London, United Kingdom
| | - Jon S Friedland
- 1 Infectious Diseases and Immunity, Wellcome Trust Centre for Global Health Research, and
| |
Collapse
|
17
|
Cox DJ, Keane J. Platelets and Tuberculosis: Small Cells, Not So Innocent Bystanders. Am J Respir Crit Care Med 2019; 198:153-154. [PMID: 29509503 DOI: 10.1164/rccm.201802-0279ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Donal J Cox
- 1 Department of Clinical Medicine Trinity College Dublin Dublin, Ireland and
| | - Joseph Keane
- 1 Department of Clinical Medicine Trinity College Dublin Dublin, Ireland and.,2 Respiratory Medicine St. James's Hospital Dublin, Ireland
| |
Collapse
|
18
|
Hortle E, Johnson KE, Johansen MD, Nguyen T, Shavit JA, Britton WJ, Tobin DM, Oehlers SH. Thrombocyte Inhibition Restores Protective Immunity to Mycobacterial Infection in Zebrafish. J Infect Dis 2019; 220:524-534. [PMID: 30877311 PMCID: PMC6603966 DOI: 10.1093/infdis/jiz110] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/07/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Infection-induced thrombocytosis is a clinically important complication of tuberculosis infection. Recent studies have highlighted the utility of aspirin as a host-directed therapy modulating the inflammatory response to infection but have not investigated the possibility that the effect of aspirin is related to an antiplatelet mode of action. METHODS In this study, we utilize the zebrafish-Mycobacterium marinum model to show mycobacteria drive host hemostasis through the formation of granulomas. Treatment of infected zebrafish with aspirin markedly reduced mycobacterial burden. This effect is reproduced by treatment with platelet-specific glycoprotein IIb/IIIa inhibitors demonstrating a detrimental role for infection-induced thrombocyte activation. RESULTS We find that the reduction in mycobacterial burden is dependent on macrophages and granuloma formation, providing the first in vivo experimental evidence that infection-induced platelet activation compromises protective host immunity to mycobacterial infection. CONCLUSIONS Our study illuminates platelet activation as an efficacious target of aspirin, a widely available and affordable host-directed therapy candidate for tuberculosis.
Collapse
Affiliation(s)
- Elinor Hortle
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, Australia
- The University of Sydney, Central Clinical School and Marie Bashir Institute, Camperdown, Australia
| | - Khelsey E Johnson
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina
| | - Matt D Johansen
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, Australia
| | - Tuong Nguyen
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, Australia
| | - Jordan A Shavit
- Department of Pediatrics and Cellular and Molecular Biology Program, University of Michigan, Ann Arbor
| | - Warwick J Britton
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, Australia
- The University of Sydney, Central Clinical School and Marie Bashir Institute, Camperdown, Australia
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, North Carolina
| | - Stefan H Oehlers
- Tuberculosis Research Program Centenary Institute, The University of Sydney, Camperdown, Australia
- The University of Sydney, Central Clinical School and Marie Bashir Institute, Camperdown, Australia
| |
Collapse
|
19
|
Use of Antiplatelet Agents and Survival of Tuberculosis Patients: A Population-Based Cohort Study. J Clin Med 2019; 8:jcm8070923. [PMID: 31252593 PMCID: PMC6678265 DOI: 10.3390/jcm8070923] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/19/2019] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
While evidence is accumulating that platelets contribute to tissue destruction in tuberculosis (TB) disease, it is still not known whether antiplatelet agents are beneficial to TB patients. We performed this retrospective cohort study and identified incident TB cases in the Taiwan National Tuberculosis Registry from 2008 to 2014. These cases were further classified into antiplatelet users and non-users according to the use of antiplatelet agents prior to the TB diagnosis, and the cohorts were matched using propensity scores (PSs). The primary outcome was survival after a TB diagnosis. In total, 74,753 incident TB cases were recruited; 9497 (12.7%) were antiplatelet users, and 7764 (10.4%) were aspirin (ASA) users. A 1:1 PS-matched cohort with 8864 antiplatelet agent users and 8864 non-users was created. After PS matching, antiplatelet use remained associated with a longer survival (adjusted hazard ratio (HR): 0.91, 95% confidence interval (CI): 0.88–0.95, p < 0.0001). The risk of major bleeding was not elevated in antiplatelet users compared to non-users (p = 0.604). This study shows that use of antiplatelet agents has been associated with improved survival in TB patients. The immunomodulatory and anti-inflammatory effects of antiplatelet agents in TB disease warrant further investigation. Antiplatelets are promising as an adjunct anti-TB therapy.
Collapse
|
20
|
Thirunavukkarasu S, Khader SA. Advances in Cardiovascular Disease Lipid Research Can Provide Novel Insights Into Mycobacterial Pathogenesis. Front Cell Infect Microbiol 2019; 9:116. [PMID: 31058102 PMCID: PMC6482252 DOI: 10.3389/fcimb.2019.00116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/02/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in industrialized nations and an emerging health problem in the developing world. Systemic inflammatory processes associated with alterations in lipid metabolism are a major contributing factor that mediates the development of CVDs, especially atherosclerosis. Therefore, the pathways promoting alterations in lipid metabolism and the interplay between varying cellular types, signaling agents, and effector molecules have been well-studied. Mycobacterial species are the causative agents of various infectious diseases in both humans and animals. Modulation of host lipid metabolism by mycobacteria plays a prominent role in its survival strategy within the host as well as in disease pathogenesis. However, there are still several knowledge gaps in the mechanistic understanding of how mycobacteria can alter host lipid metabolism. Considering the in-depth research available in the area of cardiovascular research, this review presents an overview of the parallel areas of research in host lipid-mediated immunological changes that might be extrapolated and explored to understand the underlying basis of mycobacterial pathogenesis.
Collapse
Affiliation(s)
- Shyamala Thirunavukkarasu
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| | - Shabaana A Khader
- Department of Molecular Microbiology, Washington University in St. Louis School of Medicine, St. Louis, MO, United States
| |
Collapse
|
21
|
Qi H, Zhang YB, Sun L, Chen C, Xu B, Xu F, Liu JW, Liu JC, Chen C, Jiao WW, Shen C, Xiao J, Li JQ, Guo YJ, Wang YH, Li QJ, Yin QQ, Li YJ, Wang T, Wang XY, Gu ML, Yu J, Shen AD. Discovery of susceptibility loci associated with tuberculosis in Han Chinese. Hum Mol Genet 2018; 26:4752-4763. [PMID: 29036319 DOI: 10.1093/hmg/ddx365] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 09/19/2017] [Indexed: 12/22/2022] Open
Abstract
Genome-wide association studies (GWASs) have revealed the worldwide heterogeneity of genetic factors in tuberculosis (TB) susceptibility. Despite having the third highest global TB burden, no TB-related GWAS has been performed in China. Here, we performed the first three-stage GWAS on TB in the Han Chinese population. In the stage 1 (discovery stage), after quality control, 691 388 SNPs present in 972 TB patients and 1537 controls were retained. After replication on an additional 3460 TB patients and 4862 controls (stages 2 and 3), we identified three significant loci associated with TB, the most significant of which was rs4240897 (logistic regression P = 1.41 × 10-11, odds ratio = 0.79). The aforementioned three SNPs were harbored by MFN2, RGS12 and human leukocyte antigen class II beta chain paralogue encoding genes, all of which are candidate immune genes associated with TB. Our findings provide new insight into the genetic background of TB in the Han Chinese population.
Collapse
Affiliation(s)
- Hui Qi
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yong-Biao Zhang
- Chinese Academy of Sciences and Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Lin Sun
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Cheng Chen
- Department of Chronic Communicable Disease, Center for Disease Control and Prevention, Jiangsu 210009, China
| | - Biao Xu
- School of Public Health, Fudan University, Shanghai 200433, China.,Department of Public Health Sciences (Global Health/IHCAR), Karolinska Institute, S-17177 Stockholm, Sweden
| | - Fang Xu
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Jia-Wen Liu
- Beijing Geriatric Hospital, Beijing 100095, China
| | - Jin-Cheng Liu
- Tuberculosis Hospital of Shaanxi Province 710100, Shaanxi Province, China
| | - Chen Chen
- Tuberculosis Hospital of Shaanxi Province 710100, Shaanxi Province, China
| | - Wei-Wei Jiao
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Chen Shen
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Jing Xiao
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Jie-Qiong Li
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Ya-Jie Guo
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Yong-Hong Wang
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Qin-Jing Li
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Qing-Qin Yin
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Ying-Jia Li
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Ting Wang
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xing-Yun Wang
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Ming-Liang Gu
- Chinese Academy of Sciences and Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Yu
- Chinese Academy of Sciences and Key Laboratory of Genome Science and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China
| | - A-Dong Shen
- Beijing Key Laboratory of Pediatric Respiratory Infection Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, National Key Discipline of Pediatrics (Capital Medical University), Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| |
Collapse
|
22
|
Singhal R, Chawla S, Batra H, Gupta S, Ojha A, Rathore DK, Seth T, Guchhait P. Engulfment of Hb-activated platelets differentiates monocytes into pro-inflammatory macrophages in PNH patients. Eur J Immunol 2018; 48:1285-1294. [PMID: 29677388 DOI: 10.1002/eji.201747449] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 04/04/2018] [Accepted: 04/17/2018] [Indexed: 12/31/2022]
Abstract
The distinct response shown by different phenotypes of macrophages and monocytes under various clinical conditions has put the heterogeneity of these cells into focus of investigation for several diseases. Recently, we have described that after engulfing hemoglobin (Hb)-activated platelets, classical monocytes differentiated into pro-inflammatory phenotypes, which were abundant in the circulation of paroxysmal nocturnal hemoglobinuria (PNH) and sickle cell disease patients. Our current study shows that upon engulfment of Hb-activated platelets, monocytes differentiate into M1-macrophages under M1-polarization stimulus (GM-CSF, IFN-γ + LPS). When grown under M2-polarization stimulus (M-CSF, IL-4 + IL13), the cells exhibited an M1-like phenotype, secreted elevated levels of pro-inflammatory cytokines including TNF-α and IL-1β, and displayed loss of the secretion of cytokine such as IL-10 and also phagocytic ability unlike the conventional M2 macrophages. Interestingly, when differentiated under the above polarization stimulus, monocytes from PNH patients expressed high levels of CD80 and phospho-STAT1, like M1 macrophages. Hemolytic mice also exhibited a gradual increase in monocyte-platelet aggregates in circulation and accumulation of CD80high macrophages in thioglycollate-induced inflamed peritoneum. The spleen of the mice was also populated by CD80high macrophages with compromised phagocytic capacity. Our findings suggest that the hemolytic environment and specifically the Hb-activated platelets, which are abundant in circulation during intravascular hemolysis, closely regulate monocyte differentiation.
Collapse
Affiliation(s)
- Rashi Singhal
- Disease Biology Laboratory, Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Sheetal Chawla
- Disease Biology Laboratory, Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Harish Batra
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sameer Gupta
- Disease Biology Laboratory, Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Amrita Ojha
- Disease Biology Laboratory, Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Deepak K Rathore
- Translational Health Science and Technology Institute, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| | - Tulika Seth
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Prasenjit Guchhait
- Disease Biology Laboratory, Regional Centre for Biotechnology, National Capital Region Biotech Science Cluster, Faridabad, Haryana, India
| |
Collapse
|
23
|
Kullaya V, van der Ven A, Mpagama S, Mmbaga BT, de Groot P, Kibiki G, de Mast Q. Platelet-monocyte interaction in Mycobacterium tuberculosis infection. Tuberculosis (Edinb) 2018; 111:86-93. [PMID: 30029921 DOI: 10.1016/j.tube.2018.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 04/23/2018] [Accepted: 05/06/2018] [Indexed: 02/05/2023]
Abstract
The immune effects of platelets and platelet-leukocyte aggregation are increasingly recognized. We studied the occurrence of platelet-monocyte aggregation (PMA) in patients with pulmonary tuberculosis (TB), the processes underlying PMA and consequences for cytokine responses. In a cross-sectional study involving 65 Tanzanian TB patients in different phases of treatment and 29 healthy controls, TB patients had a significantly higher PMA. This increased PMA in TB patients was associated with increased monocyte CCR5, CD16 expression and PF4, but not with increased membrane-expressed or soluble P-selectin expression. These findings were confirmed in vitro: whereas incubation of whole blood with Mycobacterium tuberculosis (Mtb) did not activate platelets, monocytes became activated with higher CD11b, CD16 and CCR5 expression, but this was independent of platelet-monocyte interaction. Still, platelets had an anti-inflammatory effect on cytokine responses as peripheral blood mononuclear cells (PBMC) incubated with Mtb in the presence of platelets produced less interleukin (IL)-1β, tumor necrosis factor-α, IL-6 and interferon-γ and more IL-10. In conclusion, increased PMA during TB infection is caused by monocyte and not platelet activation. By counteracting the Mtb-induced pro-inflammatory leukocyte response, platelets may protect against excessive tissue damage, but may also compromise the production of protective cytokines, such as IFNƴ and TNFα.
Collapse
Affiliation(s)
- Vesla Kullaya
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania.
| | - Andre van der Ven
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stellah Mpagama
- Kibong'oto Infectious Diseases Hospital, Kilimanjaro Clinical Research Institute, Sanya Juu, Tanzania
| | - Blandina T Mmbaga
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Philip de Groot
- Department of Clinical Chemistry and Haematology, University of Utrecht, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Gibson Kibiki
- Kilimanjaro Clinical Research Institute, Kilimanjaro Christian Medical Center, Moshi, Tanzania
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
24
|
Experimental infection of cattle with Mycobacterium tuberculosis isolates shows the attenuation of the human tubercle bacillus for cattle. Sci Rep 2018; 8:894. [PMID: 29343690 PMCID: PMC5772528 DOI: 10.1038/s41598-017-18575-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 12/13/2017] [Indexed: 12/27/2022] Open
Abstract
The Mycobacterium tuberculosis complex (MTBC) is the collective term given to the group of bacteria that cause tuberculosis (TB) in mammals. It has been reported that M. tuberculosis H37Rv, a standard reference MTBC strain, is attenuated in cattle compared to Mycobacterium bovis. However, as M. tuberculosis H37Rv was isolated in the early 1930s, and genetic variants are known to exist, we sought to revisit this question of attenuation of M. tuberculosis for cattle by performing a bovine experimental infection with a recent M. tuberculosis isolate. Here we report infection of cattle using M. bovis AF2122/97, M. tuberculosis H37Rv, and M. tuberculosis BTB1558, the latter isolated in 2008 during a TB surveillance project in Ethiopian cattle. We show that both M. tuberculosis strains caused reduced gross pathology and histopathology in cattle compared to M. bovis. Using M. tuberculosis H37Rv and M. bovis AF2122/97 as the extremes in terms of infection outcome, we used RNA-Seq analysis to explore differences in the peripheral response to infection as a route to identify biomarkers of progressive disease in contrast to a more quiescent, latent infection. Our work shows the attenuation of M. tuberculosis strains for cattle, and emphasizes the potential of the bovine model as a ‘One Health’ approach to inform human TB biomarker development and post-exposure vaccine development.
Collapse
|
25
|
Liu CH, Liu H, Ge B. Innate immunity in tuberculosis: host defense vs pathogen evasion. Cell Mol Immunol 2017; 14:963-975. [PMID: 28890547 PMCID: PMC5719146 DOI: 10.1038/cmi.2017.88] [Citation(s) in RCA: 326] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/16/2022] Open
Abstract
The major innate immune cell types involved in tuberculosis (TB) infection are macrophages, dendritic cells (DCs), neutrophils and natural killer (NK) cells. These immune cells recognize the TB-causing pathogen Mycobacterium tuberculosis (Mtb) through various pattern recognition receptors (PRRs), including but not limited to Toll-like receptors (TLRs), Nod-like receptors (NLRs) and C-type lectin receptors (CLRs). Upon infection by Mtb, the host orchestrates multiple signaling cascades via the PRRs to launch a variety of innate immune defense functions such as phagocytosis, autophagy, apoptosis and inflammasome activation. In contrast, Mtb utilizes numerous exquisite strategies to evade or circumvent host innate immunity. Here we discuss recent research on major host innate immune cells, PRR signaling, and the cellular functions involved in Mtb infection, with a specific focus on the host's innate immune defense and Mtb immune evasion. A better understanding of the molecular mechanisms underlying host-pathogen interactions could provide a rational basis for the development of effective anti-TB therapeutics.
Collapse
Affiliation(s)
- Cui Hua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Haiying Liu
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, and Center for Tuberculosis Research, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100176, China
| | - Baoxue Ge
- Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
26
|
Abstract
Tuberculosis (TB) affects the production and life span of all hematologic cellular components. In addition, plasma coagulation factors may be affected, resulting in sometimes life-threatening complications. Iron, folate, and vitamin B12 metabolism is derailed. The pharmacological agents used for TB therapy may also cause hematologic changes. There are some uncommon manifestations of TB in nontuberculous hematologic patients. There have been some exciting developments in the field of imaging to screen for TB, TB pathophysiology at the cellular level, and our understanding of immune response in TB. Advances have been made in pharmacologic therapeutic options, including discovery of new drugs in the fight against drug-resistant TB, bearing in mind their hematologic effects. This chapter reviews and updates known hematologic effects of TB and its therapy and some lesser known effects of TB in patients with nontuberculous hematologic conditions.
Collapse
|
27
|
Abstract
Cystic and alveolar echinococcosis are severe chronic helminthic diseases caused by the cystic growth or the intrahepatic tumour-like growth of the metacestode of Echinococcus granulosus or Echinococcus multilocularis, respectively. Both parasites have evolved sophisticated strategies to escape host immune responses, mainly by manipulating and directing this immune response towards anergy and/or tolerance. Recent research studies have revealed a number of respective immunoregulatory mechanisms related to macrophages and dendritic cell as well as T cell activities (regulatory T cells, Tregs). A better understanding of this complex parasite-host relationship, and the elucidation of specific crucial events that lead to disease, represents targets towards the development of novel treatment strategies and options.
Collapse
|
28
|
Santucci P, Bouzid F, Smichi N, Poncin I, Kremer L, De Chastellier C, Drancourt M, Canaan S. Experimental Models of Foamy Macrophages and Approaches for Dissecting the Mechanisms of Lipid Accumulation and Consumption during Dormancy and Reactivation of Tuberculosis. Front Cell Infect Microbiol 2016; 6:122. [PMID: 27774438 PMCID: PMC5054039 DOI: 10.3389/fcimb.2016.00122] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/22/2016] [Indexed: 12/11/2022] Open
Abstract
Despite a slight decline since 2014, tuberculosis (TB) remains the major deadly infectious disease worldwide with about 1.5 million deaths each year and with about one-third of the population being latently infected with Mycobacterium tuberculosis, the etiologic agent of TB. During primo-infection, the recruitment of immune cells leads to the formation of highly organized granulomas. Among the different cells, one outstanding subpopulation is the foamy macrophage (FM), characterized by the abundance of triacylglycerol-rich lipid bodies (LB). M. tuberculosis can reside in FM, where it acquires, from host LB, the neutral lipids which are subsequently processed and stored by the bacilli in the form of intracytosolic lipid inclusions (ILI). Although host LB can be viewed as a reservoir of nutrients for the pathogen during latency, the molecular mechanisms whereby intraphagosomal mycobacteria interact with LB and assimilate the LB-derived lipids are only beginning to be understood. Past studies have emphasized that these physiological processes are critical to the M. tuberculosis infectious-life cycle, for propagation of the infection, establishment of the dormancy state and reactivation of the disease. In recent years, several animal and cellular models have been developed with the aim of dissecting these complex processes and of determining the nature and contribution of their key players. Herein, we review some of the in vitro and in vivo models which allowed to gain significant insight into lipid accumulation and consumption in M. tuberculosis, two important events that are directly linked to pathogenicity, granuloma formation/maintenance and survival of the tubercle bacillus under non-replicative conditions. We also discuss the advantages and limitations of each model, hoping that this will serve as a guide for future investigations dedicated to persistence and innovative therapeutic approaches against TB.
Collapse
Affiliation(s)
- Pierre Santucci
- Aix-Marseille Université, Centre National de la Recherche Scientifique, EIPL Marseille, France
| | - Feriel Bouzid
- Aix-Marseille Université, Centre National de la Recherche Scientifique, EIPLMarseille, France; Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, URMITEMarseille, France
| | - Nabil Smichi
- Aix-Marseille Université, Centre National de la Recherche Scientifique, EIPLMarseille, France; Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, Centre National de la Recherche Scientifique FRE3689, Université de MontpellierMontpellier, France
| | - Isabelle Poncin
- Aix-Marseille Université, Centre National de la Recherche Scientifique, EIPL Marseille, France
| | - Laurent Kremer
- Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, Centre National de la Recherche Scientifique FRE3689, Université de MontpellierMontpellier, France; Centre d'études d'agents Pathogènes et Biotechnologies pour la Santé, Institut National de la Santé et de la Recherche MédicaleMontpellier, France
| | - Chantal De Chastellier
- Aix-Marseille Université, Centre National de la Recherche Scientifique, EIPL Marseille, France
| | - Michel Drancourt
- Aix-Marseille Université, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de Recherche pour le Développement, URMITE Marseille, France
| | - Stéphane Canaan
- Aix-Marseille Université, Centre National de la Recherche Scientifique, EIPL Marseille, France
| |
Collapse
|
29
|
Malkwitz I, Berndt A, Zhang R, Daugschies A, Bangoura B. Replication of Toxoplasma gondii in chicken erythrocytes and thrombocytes compared to macrophages. Parasitol Res 2016; 116:123-131. [PMID: 27696227 DOI: 10.1007/s00436-016-5268-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/19/2016] [Indexed: 10/20/2022]
Abstract
Toxoplasma (T.) gondii is able to infect various cell types in different hosts. The replication of this parasite within different peripheral mononuclear blood cell populations in chicken has not yet been fully understood. Aim of the present study was to investigate the impact of chicken erythrocytes and thrombocytes as potential host cells for T. gondii. Cultures of primary avian erythrocytes and thrombocytes were inoculated with tachyzoites of T. gondii type II strain ME49. Parasite replication was detected by a quantitative real-time PCR at different times postinoculation until 24 or 48 h, respectively, displaying long-term investigations for the chosen cultures. The parasite replication curve showed a continuous decrease of parasite stages in erythrocytes and thrombocytes. Observations by light microscopy showed massive destruction for both cell populations. Few macrophages in between the infected thrombocytes were viable during the investigation period and showed internalised tachyzoites by confocal laser scanning microscopy. These findings show that T. gondii is not capable of replication in chicken erythrocytes and thrombocytes; therefore, both cannot be considered as potential host cells. In further consequence, monocyte-derived macrophages seem to be the key to the dissemination mechanisms for T. gondii in chicken.
Collapse
Affiliation(s)
- Irene Malkwitz
- Institute of Parasitology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 35, 04103, Leipzig, Germany
| | - Angela Berndt
- Institute of Molecular Pathogenesis, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Naumburger Str. 96A, 07743, Jena, Germany
| | - Runhui Zhang
- Institute of Parasitology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 35, 04103, Leipzig, Germany
| | - Arwid Daugschies
- Institute of Parasitology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 35, 04103, Leipzig, Germany
| | - Berit Bangoura
- Institute of Parasitology, Faculty of Veterinary Medicine, Leipzig University, An den Tierkliniken 35, 04103, Leipzig, Germany.
| |
Collapse
|
30
|
Watanabe A, Araki K, Hirai K, Kubo N, Igarashi T, Tsukagoshi M, Ishii N, Hoshino K, Kuwano H, Shirabe K. A Novel Clinical Factor, D-Dimer Platelet Multiplication, May Predict Postoperative Recurrence and Prognosis for Patients with Cholangiocarcinoma. Ann Surg Oncol 2016; 23:886-891. [DOI: 10.1245/s10434-016-5422-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Indexed: 12/31/2022]
|
31
|
McClean CM, Tobin DM. Macrophage form, function, and phenotype in mycobacterial infection: lessons from tuberculosis and other diseases. Pathog Dis 2016; 74:ftw068. [PMID: 27402783 DOI: 10.1093/femspd/ftw068] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2016] [Indexed: 02/07/2023] Open
Abstract
Macrophages play a central role in mycobacterial pathogenesis. Recent work has highlighted the importance of diverse macrophage types and phenotypes that depend on local environment and developmental origins. In this review, we highlight how distinct macrophage phenotypes may influence disease progression in tuberculosis. In addition, we draw on work investigating specialized macrophage populations important in cancer biology and atherosclerosis in order to suggest new areas of investigation relevant to mycobacterial pathogenesis. Understanding the mechanisms controlling the repertoire of macrophage phenotypes and behaviors during infection may provide opportunities for novel control of disease through modulation of macrophage form and function.
Collapse
Affiliation(s)
- Colleen M McClean
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, DUMC 3020, Durham, NC 27710, USA Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA Medical Scientist Training Program, Duke University School of Medicine, Durham, NC 27710, USA
| | - David M Tobin
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, DUMC 3020, Durham, NC 27710, USA Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
32
|
Fletcher HA, Filali-Mouhim A, Nemes E, Hawkridge A, Keyser A, Njikan S, Hatherill M, Scriba TJ, Abel B, Kagina BM, Veldsman A, Agudelo NM, Kaplan G, Hussey GD, Sekaly RP, Hanekom WA. Human newborn bacille Calmette-Guérin vaccination and risk of tuberculosis disease: a case-control study. BMC Med 2016; 14:76. [PMID: 27183822 PMCID: PMC4869393 DOI: 10.1186/s12916-016-0617-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 04/23/2016] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND An incomplete understanding of the immunological mechanisms underlying protection against tuberculosis (TB) hampers the development of new vaccines against TB. We aimed to define host correlates of prospective risk of TB disease following bacille Calmette-Guérin (BCG) vaccination. METHODS In this study, 5,726 infants vaccinated with BCG at birth were enrolled. Host responses in blood collected at 10 weeks of age were compared between infants who developed pulmonary TB disease during 2 years of follow-up (cases) and those who remained healthy (controls). RESULTS Comprehensive gene expression and cellular and soluble marker analysis failed to identify a correlate of risk. We showed that distinct host responses after BCG vaccination may be the reason: two major clusters of gene expression, with different myeloid and lymphoid activation and inflammatory patterns, were evident when all infants were examined together. Cases from each cluster demonstrated distinct patterns of gene expression, which were confirmed by cellular assays. CONCLUSIONS Distinct patterns of host responses to Mycobacterium bovis BCG suggest that novel TB vaccines may also elicit distinct patterns of host responses. This diversity should be considered in future TB vaccine development.
Collapse
Affiliation(s)
| | - Ali Filali-Mouhim
- Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Elisa Nemes
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Anthony Hawkridge
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Alana Keyser
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Samuel Njikan
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Mark Hatherill
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Brian Abel
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Benjamin M Kagina
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Ashley Veldsman
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Nancy Marín Agudelo
- Grupo de Inmunología Celular e Inmunogenética, Sede de Investigación Universitaria, Universidad de Antioquia, Medellín, Colombia
| | - Gilla Kaplan
- Public Health Research Institute, Rutgers Biomedical and Health Sciences, Newark, NJ, USA
| | - Gregory D Hussey
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | | | - Willem A Hanekom
- South African Tuberculosis Vaccine Initiative (SATVI), Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa.
| | | |
Collapse
|
33
|
Robinson RT, Orme IM, Cooper AM. The onset of adaptive immunity in the mouse model of tuberculosis and the factors that compromise its expression. Immunol Rev 2015; 264:46-59. [PMID: 25703551 DOI: 10.1111/imr.12259] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mycobacterium tuberculosis (Mtb) has been evolving with its human host for over 50 000 years and is an exquisite manipulator of the human immune response. It induces both a strong inflammatory and a strong acquired immune response, and Mtb then actively regulates these responses to create an infectious lesion in the lung while maintaining a relatively ambulatory host. The CD4(+) T cell plays a critical yet contradictory role in this process by both controlling disseminated disease while promoting the development of the lesion in the lung that mediates transmission. In light of this manipulative relationship between Mtb and the human immune response, it is not surprising that our ability to vaccinate against tuberculosis (TB) has not been totally successful. To overcome the current impasse in vaccine development, we need to define the phenotype of CD4(+) T cells that mediate protection and to determine those bacterial and host factors that regulate the effective function of these cells. In this review, we describe the initiation and expression of T cells during TB as well as the fulminant inflammatory response that can compromise T-cell function and survival.
Collapse
Affiliation(s)
- Richard T Robinson
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | |
Collapse
|
34
|
Dorhoi A, Kaufmann SHE. Versatile myeloid cell subsets contribute to tuberculosis-associated inflammation. Eur J Immunol 2015; 45:2191-202. [PMID: 26140356 DOI: 10.1002/eji.201545493] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/23/2015] [Accepted: 06/29/2015] [Indexed: 12/21/2022]
Abstract
Tuberculosis (TB), a chronic bacterial infectious disease caused by Mycobacterium tuberculosis (Mtb), typically affects the lung and causes profound morbidity and mortality rates worldwide. Recent advances in cellular immunology emphasize the complexity of myeloid cell subsets controlling TB inflammation. The specialization of myeloid cell subsets for particular immune processes has tailored their roles in protection and pathology. Among myeloid cells, dendritic cells (DCs) are essential for the induction of adaptive immunity, macrophages predominantly harbor Mtb within TB granulomas and polymorphonuclear neutrophils (PMNs) orchestrate lung damage. However, within each myeloid cell population, diverse phenotypes with unique functions are currently recognized, differentially influencing TB pneumonia and granuloma functionality. More recently, myeloid-derived suppressor cells (MDSCs) have been identified at the site of Mtb infection. Along with PMNs, MDSCs accumulate within the inflamed lung, interact with granuloma-residing cells and contribute to exuberant inflammation. In this review, we discuss the contribution of different myeloid cell subsets to inflammation in TB by highlighting their interactions with Mtb and their role in lung pathology. Uncovering the manifold nature of myeloid cells in TB pathogenesis will inform the development of future immune therapies aimed at tipping the inflammation balance to the benefit of the host.
Collapse
Affiliation(s)
- Anca Dorhoi
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
35
|
Pathology and immune reactivity: understanding multidimensionality in pulmonary tuberculosis. Semin Immunopathol 2015; 38:153-66. [PMID: 26438324 DOI: 10.1007/s00281-015-0531-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 09/13/2015] [Indexed: 12/19/2022]
Abstract
Heightened morbidity and mortality in pulmonary tuberculosis (TB) are consequences of complex disease processes triggered by the causative agent, Mycobacterium tuberculosis (Mtb). Mtb modulates inflammation at distinct stages of its intracellular life. Recognition and phagocytosis, replication in phagosomes and cytosol escape induce tightly regulated release of cytokines [including interleukin (IL)-1, tumor necrosis factor (TNF), IL-10], chemokines, lipid mediators, and type I interferons (IFN-I). Mtb occupies various lung lesions at sites of pathology. Bacteria are barely detectable at foci of lipid pneumonia or in perivascular/bronchiolar cuffs. However, abundant organisms are evident in caseating granulomas and at the cavity wall. Such lesions follow polar trajectories towards fibrosis, encapsulation and mineralization or liquefaction, extensive matrix destruction, and tissue injury. The outcome is determined by immune factors acting in concert. Gradients of cytokines and chemokines (CCR2, CXCR2, CXCR3/CXCR5 agonists; TNF/IL-10, IL-1/IFN-I), expression of activation/death markers on immune cells (TNF receptor 1, PD-1, IL-27 receptor) or abundance of enzymes [arginase-1, matrix metalloprotease (MMP)-1, MMP-8, MMP-9] drive genesis and progression of lesions. Distinct lesions coexist such that inflammation in TB encompasses a spectrum of tissue changes. A better understanding of the multidimensionality of immunopathology in TB will inform novel therapies against this pulmonary disease.
Collapse
|
36
|
Dorhoi A, Feng Y, Kaufmann SHE. Reply to Crawford. J Infect Dis 2015; 212:1173-4. [PMID: 25801653 DOI: 10.1093/infdis/jiv156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 03/04/2015] [Indexed: 11/14/2022] Open
Affiliation(s)
- Anca Dorhoi
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Yonghong Feng
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany Shanghai Key Lab of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
37
|
Crawford CL. The Epithelioid Cell in Tuberculosis is Secretory and Not a Macrophage. J Infect Dis 2015; 212:1172-3. [PMID: 25801654 DOI: 10.1093/infdis/jiv155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 03/04/2015] [Indexed: 12/24/2022] Open
|
38
|
Nagasawa T, Nakayasu C, Rieger AM, Barreda DR, Somamoto T, Nakao M. Phagocytosis by Thrombocytes is a Conserved Innate Immune Mechanism in Lower Vertebrates. Front Immunol 2014; 5:445. [PMID: 25278940 PMCID: PMC4165319 DOI: 10.3389/fimmu.2014.00445] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Accepted: 09/01/2014] [Indexed: 01/07/2023] Open
Abstract
Thrombocytes, nucleated hemostatic blood cells of non-mammalian vertebrates, are regarded as the functional equivalent of anucleated mammalian platelets. Additional immune functions, including phagocytosis, have also been suggested for thrombocytes, but no conclusive molecular or cellular experimental evidence for their potential ingestion and clearance of infiltrating microbes has been provided till date. In the present study, we demonstrate the active phagocytic ability of thrombocytes in lower vertebrates using teleost fishes and amphibian models. Ex vivo, common carp thrombocytes were able to ingest live bacteria as well as latex beads (0.5-3 μm in diameter) and kill the bacteria. In vivo, we found that thrombocytes represented nearly half of the phagocyte population in the common carp total peripheral blood leukocyte pool. Phagocytosis efficiency was further enhanced by serum opsonization. Particle internalization led to phagolysosome fusion and killing of internalized bacteria, pointing to a robust ability for microbe elimination. We find that this potent phagocytic activity is shared across teleost (Paralichthys olivaceus) and amphibian (Xenopus laevis) models examined, implying its conservation throughout the lower vertebrate lineage. Our results provide novel insights into the dual nature of thrombocytes in the immune and homeostatic response and further provide a deeper understanding of the potential immune function of mammalian platelets based on the conserved and vestigial functions.
Collapse
Affiliation(s)
- Takahiro Nagasawa
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Chihaya Nakayasu
- National Research Institute of Aquaculture, Fisheries Research Agency, Minami-Ise, Japan
| | - Aja M. Rieger
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Daniel R. Barreda
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Tomonori Somamoto
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| | - Miki Nakao
- Department of Bioscience and Biotechnology, Graduate School of Bioresource and Bioenvironmental Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
39
|
Lugo-Villarino G, Neyrolles O. Of clots and granulomas: platelets are new players in immunity to tuberculosis. J Infect Dis 2014; 210:1687-90. [PMID: 24987032 DOI: 10.1093/infdis/jiu356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Geanncarlo Lugo-Villarino
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Olivier Neyrolles
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| |
Collapse
|