1
|
Le Page L, Baldwin CL, Telfer JC. γδ T cells in artiodactyls: Focus on swine. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 128:104334. [PMID: 34919982 DOI: 10.1016/j.dci.2021.104334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 06/14/2023]
Abstract
Vaccination is the most effective medical strategy for disease prevention but there is a need to improve livestock vaccine efficacy. Understanding the structure of the immune system of swine, which are considered a γδ T cell "high" species, and thus, particularly how to engage their γδ T cells for immune responses, may allow for development of vaccine optimization strategies. The propensity of γδ T cells to home to specific tissues, secrete pro-inflammatory and regulatory cytokines, exhibit memory or recall responses and even function as antigen-presenting cells for αβ T cells supports the concept that they have enormous potential for priming by next generation vaccine constructs to contribute to protective immunity. γδ T cells exhibit several innate-like antigen recognition properties including the ability to recognize antigen in the absence of presentation via major histocompatibility complex (MHC) molecules enabling γδ T cells to recognize an array of peptides but also non-peptide antigens in a T cell receptor-dependent manner. γδ T cell subpopulations in ruminants and swine can be distinguished based on differential expression of the hybrid co-receptor and pattern recognition receptors (PRR) known as workshop cluster 1 (WC1). Expression of various PRR and other innate-like immune receptors diversifies the antigen recognition potential of γδ T cells. Finally, γδ T cells in livestock are potent producers of critical master regulator cytokines such as interferon (IFN)-γ and interleukin (IL)-17, whose production orchestrates downstream cytokine and chemokine production by other cells, thereby shaping the immune response as a whole. Our knowledge of the biology, receptor expression and response to infectious diseases by swine γδ T cells is reviewed here.
Collapse
Affiliation(s)
- Lauren Le Page
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Cynthia L Baldwin
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Janice C Telfer
- Department of Veterinary & Animal Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
2
|
Charpentier E, Ménard S, Marques C, Berry A, Iriart X. Immune Response in Pneumocystis Infections According to the Host Immune System Status. J Fungi (Basel) 2021; 7:jof7080625. [PMID: 34436164 PMCID: PMC8399367 DOI: 10.3390/jof7080625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
The host immune response is critical in Pneumocystis pneumonia (PCP). Immunocompetent hosts can eliminate the fungus without symptoms, while immunodeficient hosts develop PCP with an unsuitable excessive inflammatory response leading to lung damage. From studies based on rodent models or clinical studies, this review aimed to better understand the pathophysiology of Pneumocystis infection by analysing the role of immune cells, mostly lymphocytes, according to the immune status of the infected host. Hence, this review first describes the immune physiological response in infected immunocompetent hosts that are able to eliminate the fungus. The objective of the second part is to identify the immune elements required for the control of the fungus, focusing on specific immune deficiencies. Finally, the third part concentrates on the effect of the different immune elements in immunocompromised subjects during PCP, to better understand which cells are detrimental, and which, on the contrary, are beneficial once the disease has started. This work highlights that the immune response associated with a favourable outcome of the infection may differ according to the immune status of the host. In the case of immunocompetency, a close communication between B cells and TCD4 within tertiary lymphocyte structures appears critical to activate M2 macrophages without much inflammation. Conversely, in the case of immunodeficiency, a pro-inflammatory response including Th1 CD4, cytotoxic CD8, NK cells, and IFNγ release seems beneficial for M1 macrophage activation, despite the impact of inflammation on lung tissue.
Collapse
Affiliation(s)
- Eléna Charpentier
- Department of Parasitology-Mycology, Toulouse University Hospital, 31059 Toulouse, France;
- Infinity, Inserm, CNRS, University of Toulouse III, 31024 Toulouse, France; (S.M.); (C.M.)
- Correspondence: (E.C.); (X.I.)
| | - Sandie Ménard
- Infinity, Inserm, CNRS, University of Toulouse III, 31024 Toulouse, France; (S.M.); (C.M.)
| | - Catherine Marques
- Infinity, Inserm, CNRS, University of Toulouse III, 31024 Toulouse, France; (S.M.); (C.M.)
| | - Antoine Berry
- Department of Parasitology-Mycology, Toulouse University Hospital, 31059 Toulouse, France;
- Infinity, Inserm, CNRS, University of Toulouse III, 31024 Toulouse, France; (S.M.); (C.M.)
| | - Xavier Iriart
- Department of Parasitology-Mycology, Toulouse University Hospital, 31059 Toulouse, France;
- Infinity, Inserm, CNRS, University of Toulouse III, 31024 Toulouse, France; (S.M.); (C.M.)
- Correspondence: (E.C.); (X.I.)
| |
Collapse
|
3
|
Dunne MR, Wagener J, Loeffler J, Doherty DG, Rogers TR. Unconventional T cells - New players in antifungal immunity. Clin Immunol 2021; 227:108734. [PMID: 33895356 DOI: 10.1016/j.clim.2021.108734] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/06/2021] [Accepted: 04/20/2021] [Indexed: 12/29/2022]
Abstract
Life-threatening invasive fungal diseases (IFD) are increasing in incidence, especially in immunocompromised patients and successful resolution of IFD requires a variety of different immune cells. With the limited repertoire of available antifungal drugs there is a need for more effective therapeutic strategies. This review interrogates the evidence on the human immune response to the main pathogens driving IFD, with a focus on the role of unconventional lymphocytes e.g. natural killer (NK) cells, gamma/delta (γδ) T cells, mucosal associated invariant T (MAIT) cells, invariant natural killer T (iNKT) cells and innate lymphoid cells (ILC). Recent discoveries and new insights into the roles of these novel lymphocyte groups in antifungal immunity will be discussed, and we will explore how an improved understanding of antifungal action by lymphocytes can inform efforts to improve antifungal treatment options.
Collapse
Affiliation(s)
- Margaret R Dunne
- Department of Clinical Microbiology, Trinity College Dublin, Sir Patrick Dun Research Laboratory, St James's Hospital, Dublin 8, Ireland; Department of Immunology, School of Medicine, Trinity College Dublin, Dublin 8, Ireland.
| | - Johannes Wagener
- Department of Clinical Microbiology, Trinity College Dublin, Sir Patrick Dun Research Laboratory, St James's Hospital, Dublin 8, Ireland
| | - Juergen Loeffler
- Department of Internal Medicine II, WÜ4i, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Derek G Doherty
- Department of Immunology, School of Medicine, Trinity College Dublin, Dublin 8, Ireland
| | - Thomas R Rogers
- Department of Clinical Microbiology, Trinity College Dublin, Sir Patrick Dun Research Laboratory, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
4
|
Galeas-Pena M, McLaughlin N, Pociask D. The role of the innate immune system on pulmonary infections. Biol Chem 2019; 400:443-456. [PMID: 29604208 DOI: 10.1515/hsz-2018-0304] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/19/2018] [Indexed: 12/15/2022]
Abstract
Inhalation is required for respiration and life in all vertebrates. This process is not without risk, as it potentially exposes the host to environmental pathogens with every breath. This makes the upper respiratory tract one of the most common routes of infection and one of the leading causes of morbidity and mortality in the world. To combat this, the lung relies on the innate immune defenses. In contrast to the adaptive immune system, the innate immune system does not require sensitization, previous exposure or priming to attack foreign particles. In the lung, the innate immune response starts with the epithelial barrier and mucus production and is reinforced by phagocytic cells and T cells. These cells are vital for the production of cytokines, chemokines and anti-microbial peptides that are critical for clearance of infectious agents. In this review, we discuss all aspects of the innate immune response, with a special emphasis on ways to target aspects of the immune response to combat antibiotic resistant bacteria.
Collapse
Affiliation(s)
- Michelle Galeas-Pena
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, 333 S. Liberty St., New Orleans, LA 70112, USA
| | - Nathaniel McLaughlin
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, 333 S. Liberty St., New Orleans, LA 70112, USA
| | - Derek Pociask
- Department of Pulmonary Critical Care and Environmental Medicine, Tulane University School of Medicine, 333 S. Liberty St., New Orleans, LA 70112, USA
| |
Collapse
|
5
|
Lionakis MS, Levitz SM. Host Control of Fungal Infections: Lessons from Basic Studies and Human Cohorts. Annu Rev Immunol 2017; 36:157-191. [PMID: 29237128 DOI: 10.1146/annurev-immunol-042617-053318] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In the last few decades, the AIDS pandemic and the significant advances in the medical management of individuals with neoplastic and inflammatory conditions have resulted in a dramatic increase in the population of immunosuppressed patients with opportunistic, life-threatening fungal infections. The parallel development of clinically relevant mouse models of fungal disease and the discovery and characterization of several inborn errors of immune-related genes that underlie inherited human susceptibility to opportunistic mycoses have significantly expanded our understanding of the innate and adaptive immune mechanisms that protect against ubiquitous fungal exposures. This review synthesizes immunological knowledge derived from basic mouse studies and from human cohorts and provides an overview of mammalian antifungal host defenses that show promise for informing therapeutic and vaccination strategies for vulnerable patients.
Collapse
Affiliation(s)
- Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892;
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01655;
| |
Collapse
|
6
|
Ripamonti C, Bishop LR, Kovacs JA. Pulmonary Interleukin-17-Positive Lymphocytes Increase during Pneumocystis murina Infection but Are Not Required for Clearance of Pneumocystis. Infect Immun 2017; 85:e00434-16. [PMID: 28438973 PMCID: PMC5478948 DOI: 10.1128/iai.00434-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 04/14/2017] [Indexed: 12/22/2022] Open
Abstract
Pneumocystis remains an important pathogen of immunosuppressed patients, causing a potentially life-threatening pneumonia. Despite its medical importance, the immune responses required to control infection, including the role of interleukin-17 (IL-17), which is important in controlling other fungal infections, have not been clearly defined. Using flow cytometry and intracellular cytokine staining after stimulation with phorbol myristate acetate and ionomycin, we examined gamma interferon (IFN-γ), IL-4, IL-5, and IL-17 production by lung lymphocytes in immunocompetent C57BL/6 mice over time following infection with Pneumocystismurina We also examined the clearance of Pneumocystis infection in IL-17A-deficient mice. The production of both IFN-γ and IL-17 by pulmonary lymphocytes increased during infection, with maximum production at approximately days 35 to 40, coinciding with peak Pneumocystis levels in the lungs, while minimal changes were seen in IL-4- and IL-5-positive cells. The proportion of cells producing IFN-γ was consistently higher than for cells producing IL-17, with peak levels of ∼25 to 30% of CD3+ T cells for the former compared to ∼15% for the latter. Both CD4+ T cells and γδ T cells produced IL-17. Administration of anti-IFN-γ antibody led to a decrease in IFN-γ-positive cells, and an increase in IL-5-positive cells, but did not impact clearance of Pneumocystis infection. Despite the increases in IL-17 production during infection, IL-17A-deficient mice cleared Pneumocystis infection with kinetics similar to C57BL/6 mice. Thus, while IL-17 production in the lungs is increased during Pneumocystis infection in immunocompetent mice, IL-17A is not required for control of Pneumocystis infection.
Collapse
Affiliation(s)
- Chiara Ripamonti
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Lisa R Bishop
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph A Kovacs
- Critical Care Medicine Department, NIH Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
DeWitt JC, Germolec DR, Luebke RW, Johnson VJ. Associating Changes in the Immune System with Clinical Diseases for Interpretation in Risk Assessment. CURRENT PROTOCOLS IN TOXICOLOGY 2016; 67:18.1.1-18.1.22. [PMID: 26828330 PMCID: PMC4780336 DOI: 10.1002/0471140856.tx1801s67] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This overview is an update of the unit originally published in 2004. While the basic tenets of immunotoxicity have not changed in the past 10 years, several publications have explored the application of immunotoxicological data to the risk assessment process. Therefore, the goal of this unit is still to highlight relationships between xenobiotic-induced immunosuppression and risk of clinical diseases progression. In immunotoxicology, this may require development of models to equate moderate changes in markers of immune functions to potential changes in incidence or severity of infectious diseases. For most xenobiotics, exposure levels and disease incidence data are rarely available, and safe exposure levels must be estimated based on observations from experimental models or human biomarker studies. Thus, it is important to establish a scientifically sound framework that allows accurate and quantitative interpretation of experimental or biomarker data in the risk assessment process.
Collapse
Affiliation(s)
- Jamie C DeWitt
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | - Dori R Germolec
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, North Carolina
| | - Robert W Luebke
- Cardiopulmonary and Immunotoxicology Branch, United States Environmental Protection Agency, Research Triangle Park, North Carolina
| | | |
Collapse
|
8
|
Treatment with Interleukin-7 Restores Host Defense against Pneumocystis in CD4+ T-Lymphocyte-Depleted Mice. Infect Immun 2015; 84:108-19. [PMID: 26483405 DOI: 10.1128/iai.01189-15] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 10/10/2015] [Indexed: 12/16/2022] Open
Abstract
Pneumocystis pneumonia (PCP) is a major cause of morbidity and mortality in patients with HIV infection. CD4(+) T lymphocytes are critical for host defense against this infection, but in the absence of CD4(+) T lymphocytes, CD8(+) T lymphocytes may provide limited host defense. The cytokine interleukin-7 (IL-7) functions to enhance lymphocyte proliferation, survival, and recruitment of immune cells to sites of infection. However, there is little known about the role of IL-7 in PCP or its potential use as an immunotherapeutic agent. We hypothesized that treatment with recombinant human IL-7 (rhIL-7) would augment host defense against Pneumocystis and accelerate pathogen clearance in CD4-depleted mice. Control and CD4-depleted mice were infected with Pneumocystis, and rhIL-7 was administered via intraperitoneal injection. Our studies indicate that endogenous murine IL-7 is part of the normal host response to Pneumocystis murina and that administration of rhIL-7 markedly enhanced clearance of Pneumocystis in CD4-depleted mice. Additionally, we observed increased recruitment of CD8(+) T lymphocytes to the lungs and decreased apoptosis of pulmonary CD8(+) T lymphocytes in rhIL-7-treated animals compared to those in untreated mice. The antiapoptotic effect of rhIL-7 was associated with increased levels of Bcl-2 protein in T lymphocytes. rhIL-7 immunotherapy in CD4-depleted mice also increased the number of gamma interferon (IFN-γ)-positive CD8(+) central memory T lymphocytes in the lungs. We conclude that rhIL-7 has a potent therapeutic effect in the treatment of murine Pneumocystis pneumonia in CD4-depleted mice. This therapeutic effect is mediated through enhanced recruitment of CD8(+) T cells and decreased apoptosis of lung T lymphocytes, with a preferential action on central memory CD8(+) T lymphocytes.
Collapse
|
9
|
Abstract
Evidence has increasingly shown that the lungs are a major site of immune regulation. A robust and highly regulated immune response in the lung protects the host from pathogen infection, whereas an inefficient or deleterious response can lead to various pulmonary diseases. Many cell types, such as epithelial cells, dendritic cells, macrophages, neutrophils, eosinophils, and B and T lymphocytes, contribute to lung immunity. This review focuses on the recent advances in understanding how T lymphocytes mediate pulmonary host defenses against bacterial, viral, and fungal pathogens.
Collapse
Affiliation(s)
- Kong Chen
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, Pennsylvania 15201, USA
| | | |
Collapse
|
10
|
Abstract
Although the incidence of Pneumocystis pneumonia (PCP) has decreased since the introduction of combination antiretroviral therapy, it remains an important cause of disease in both HIV-infected and non-HIV-infected immunosuppressed populations. The epidemiology of PCP has shifted over the course of the HIV epidemic both from changes in HIV and PCP treatment and prevention and from changes in critical care medicine. Although less common in non-HIV-infected immunosuppressed patients, PCP is now more frequently seen due to the increasing numbers of organ transplants and development of novel immunotherapies. New diagnostic and treatment modalities are under investigation. The immune response is critical in preventing this disease but also results in lung damage, and future work may offer potential areas for vaccine development or immunomodulatory therapy. Colonization with Pneumocystis is an area of increasing clinical and research interest and may be important in development of lung diseases such as chronic obstructive pulmonary disease. In this review, we discuss current clinical and research topics in the study of Pneumocystis and highlight areas for future research.
Collapse
|
11
|
Nelson MP, Christmann BS, Dunaway CW, Morris A, Steele C. Experimental Pneumocystis lung infection promotes M2a alveolar macrophage-derived MMP12 production. Am J Physiol Lung Cell Mol Physiol 2012; 303:L469-75. [PMID: 22773692 DOI: 10.1152/ajplung.00158.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Among several bacterial and viral pathogens, the atypical fungal organism Pneumocystis jirovecii has been implicated as a contributor to the pathogenesis of chronic obstructive pulmonary disease (COPD). In a previous study, we reported that Pneumocystis-colonized HIV-positive subjects had worse obstruction of airways and higher sputum levels of macrophage elastase/matrix metalloproteinase 12 (MMP12), a protease strongly associated with the development of COPD. Here, we examined parameters of Pneumocystis-induced MMP12 in the lungs of mice and its role in the lung immune response to murine Pneumocystis. Initial studies demonstrated that P. murina exposure induced Mmp12 mRNA expression in whole lungs and alveolar macrophages (AMs), which was dependent on the presence of CD4+ T cells as well as signal transducer and activator of transcription 6. Mmp12 mRNA expression was upregulated in AMs by interleukin (IL)-4 treatment, but downregulated by interferon (IFN)-γ, indicating preferential expression in alternatively activated (M2a) macrophages. IL-4 treatment induced the 54-kDa proenzyme form of MMP12 and the 22-kDa fully processed and active form, whereas IFN-γ failed to induce either. Despite a reported antimicrobial role in macrophage phagolysosomes, mice deficient in MMP12 were not found to be more susceptible to lung infection with P. murina. Collectively, our data indicate that MMP12 induction is a component of the P. murina-induced M2 response and thus provides insight into the link between Pneumocystis colonization/infection and exacerbations in COPD.
Collapse
Affiliation(s)
- Michael P Nelson
- Dept. of Medicine, Univ. of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | |
Collapse
|
12
|
Pneumocystis infection in an immunocompetent host can promote collateral sensitization to respiratory antigens. Infect Immun 2011; 79:1905-14. [PMID: 21343358 DOI: 10.1128/iai.01273-10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Infection with the opportunistic fungal pathogen Pneumocystis is assumed to pass without persistent pathology in immunocompetent hosts. However, when immunocompetent BALB/c mice were inoculated with Pneumocystis, a vigorous Th2-like pulmonary inflammation ensued and peaked at 14 days postinfection. This coincided with a 10-fold increase in the number of antigen-presenting cells (APCs) in the lung, and these cells were capable of presenting antigen in vitro, as well as greater uptake of antigen in vivo. When mice were presented with exogenous antigen at the 14-day time point of the infection, they developed respiratory sensitization to that antigen, in the form of increased airway hyperresponsiveness upon a later challenge, whereas mice not infected but presented with antigen did not. Like other forms of collateral sensitization, this response was dependent on interleukin-4 receptor signaling. This ability to facilitate sensitization to exogenous antigen has been previously reported for other infectious disease agents; however, Pneumocystis appears to be uniquely capable in this respect, as a single intranasal dose without added adjuvant, when it was administered at the appropriate time, was sufficient to initiate sensitization. Pneumocystis infection probably occurs in most humans during the first few years of life, and in the vast majority of cases, it fails to cause any overt direct pathology. However, as we show here, Pneumocystis can be an agent of comorbidity at this time by facilitating respiratory sensitization that may relate to the later development or exacerbation of obstructive airway disease.
Collapse
|
13
|
Nelson MP, Christmann BS, Werner JL, Metz AE, Trevor JL, Lowell CA, Steele C. IL-33 and M2a alveolar macrophages promote lung defense against the atypical fungal pathogen Pneumocystis murina. THE JOURNAL OF IMMUNOLOGY 2011; 186:2372-81. [PMID: 21220696 DOI: 10.4049/jimmunol.1002558] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We have recently reported that mice deficient in the myeloid Src-family tyrosine kinases Hck, Fgr, and Lyn (Src triple knockout [TKO]) had augmented innate lung clearance of Pneumocystis murina that correlated with a higher ability of alveolar macrophages (AMs) from these mice to kill P. murina. In this article, we show that despite possessing enhanced killing, AMs from naive Src TKO mice did not demonstrate enhanced inflammatory responses to P. murina. We subsequently discovered that both AMs and lungs from P. murina-infected Src TKO mice expressed significantly greater levels of the M2a markers RELM-α and Arg1, and the M2a-associated chemokines CCL17 and CCL22 than did wild-type mice. IL-4 and IL-13, the primary cytokines that promote M2a polarization, were not differentially produced in the lungs between wild-type and Src TKO mice. P. murina infection in Src TKO mice resulted in enhanced lung production of the novel IL-1 family cytokine IL-33. Immunohistochemical analysis of IL-33 in lung tissue revealed localization predominantly in the nucleus of alveolar epithelial cells. We further demonstrate that experimental polarization of naive AMs to M2a resulted in more efficient killing of P. murina compared with untreated AMs, which was further enhanced by the addition of IL-33. Administration of IL-33 to C57BL/6 mice increased lung RELM-α and CCL17 levels, and enhanced clearance of P. murina, despite having no effect on the cellular composition of the lungs. Collectively, these results indicate that M2a AMs are potent effector cells against P. murina. Furthermore, enhancing M2a polarization may be an adjunctive therapy for the treatment of Pneumocystis.
Collapse
Affiliation(s)
- Michael P Nelson
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Meissner N, Swain S, McInnerney K, Han S, Harmsen AG. Type-I IFN signaling suppresses an excessive IFN-gamma response and thus prevents lung damage and chronic inflammation during Pneumocystis (PC) clearance in CD4 T cell-competent mice. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:2806-18. [PMID: 20395428 DOI: 10.2353/ajpath.2010.091158] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Immune-reconstitution after highly active antiretroviral therapy (HAART) is often incomplete, and some HIV-infected individuals fail to regenerate type-I interferon (IFN)-producing pDCs. We recently demonstrated that during Pneumocystis (PC) infection in CD4 T cell-competent mice the absence of type-I IFN signaling results in chronic pulmonary inflammation and fibrosis despite clearance. Because the mechanisms involved are poorly understood, we further characterized the role of type-I IFN signaling in immune responses to PC. We show that type-I IFN signaling around day 7 postinfection is critical to the outcome of inflammation. Microarray analysis of pulmonary CD11c(+) cells revealed that at day 7 post infection, wild-type cells up-regulated type-I IFN-responsive genes as well as SOCS1, which is a critical negative-regulator of type-I IFN and IFN-gamma signaling. This was associated with an eosinophilic lung inflammation, PC clearance, and complete restitution. However, pulmonary CD11c(+) cells from IFNAR(-/-) mice demonstrated increased tumor necrosis factor (TNF)-alpha production and lacked SOCS1-induction at day 7. This was followed by a transient lymphocytic and IFN-gamma response before switching to a chronic eosinophilic inflammation of the lung. Early neutralization of TNF-alpha did not prevent chronic inflammation in IFNAR(-/-) mice, but treatment with an anti-IFN-gamma antibody did. We propose that during PC lung infection type-I IFNs induce SOCS1-associated regulatory mechanisms, which prevent excessive IFN-gamma-mediated responses that cause chronic lung damage. Therefore, partial immune-reconstitution in AIDS, attributable to reduced type-I IFN actions, might disrupt regulatory aspects of inflammation, causing unexplained chronic pulmonary complications as seen in some patients during HAART.
Collapse
Affiliation(s)
- Nicole Meissner
- Veterinary Molecular Biology Department, Montana State University, 960 Technology Blvd., Bozeman, MT 59718, USA.
| | | | | | | | | |
Collapse
|
15
|
Simonian PL, Roark CL, Born WK, O'Brien RL, Fontenot AP. Gammadelta T cells and Th17 cytokines in hypersensitivity pneumonitis and lung fibrosis. Transl Res 2009; 154:222-7. [PMID: 19840763 PMCID: PMC2766087 DOI: 10.1016/j.trsl.2009.08.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 07/29/2009] [Indexed: 02/06/2023]
Abstract
Hypersensitivity pneumonitis (HP) is an inflammatory lung disease caused by the repeated inhalation of aerosolized antigens. With chronic exposure to an inhaled antigen, patients are at risk of developing irreversible pulmonary fibrosis as well as an increased morbidity and mortality. Although alphabeta T cells have been shown to be important in the pathogenesis of HP, gammadelta T cells also accumulate in the bronchoalveolar lavage of patients with HP. gammadelta T cells represent a distinct lymphocyte subset, whose primary function is not well understood. In contrast to alphabeta T cells, gammadelta T cells recognize unprocessed antigens, such as those upregulated on injured or stressed epithelial cells. In a murine model of HP induced by exposure to the ubiquitous microorganism Bacillus subtilis, gammadelta T cells expressing the canonical Vgamma6/Vdelta1 T cell receptor were dramatically expanded in the lung. The predominant cytokines expressed by this gammadelta T-cell subset were T-helper 17 (Th17) cytokines that were critical for bacterial clearance and the resolution of lung inflammation. Th17-expressing gammadelta T cells are also expanded in other murine models of lung infection and inflammation, which suggests that these cells play a sentinel role in mucosal immunity. Thus, an increased understanding of gammadelta T cells that express Th17 cytokines in HP and other inflammatory lung diseases may lead to the development of novel therapeutic and clinical strategies that prevent the development of fibrotic lung disease.
Collapse
Affiliation(s)
- Philip L Simonian
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.
| | | | | | | | | |
Collapse
|
16
|
Simonian PL, Roark CL, Wehrmann F, Lanham AM, Born WK, O'Brien RL, Fontenot AP. IL-17A-expressing T cells are essential for bacterial clearance in a murine model of hypersensitivity pneumonitis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:6540-9. [PMID: 19414809 PMCID: PMC2766088 DOI: 10.4049/jimmunol.0900013] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hypersensitivity pneumonitis (HP) is an inflammatory lung disease characterized by a diffuse mononuclear cell infiltrate in the lung that can progress to pulmonary fibrosis with chronic exposure to an inhaled Ag. We previously reported that C57BL/6 mice repeatedly exposed to the ubiquitous microorganism Bacillus subtilis develop mononuclear infiltrates in the lung that contain Vgamma6/Vdelta1(+) gammadelta T cells. In the absence of this T cell subset, mice treated with B. subtilis had significantly increased collagen deposition in the lung, suggesting a regulatory role for Vgamma6/Vdelta1(+) gammadelta T cells. To further investigate the role of Vgamma6/Vdelta1(+) gammadelta T cells in B. subtilis-induced lung fibrosis, we exposed transgenic Vgamma6/Vdelta1 mice to this microorganism and found decreased collagen content in the lung compared with wild-type C57BL/6 mice. Cytokine analysis of lung homogenates from wild-type C57BL/6 mice demonstrated increased IL-17A concentrations with repeated exposure to B. subtilis. In the absence of IL-17 receptor signaling, IL-17ra(-/-) mice had delayed clearance of B. subtilis with increased lung inflammation and fibrosis. Although IL-17A was predominantly expressed by Vgamma6/Vdelta1(+) T cells, a compensatory increase in IL-17A expression by CD4(+) T cells was seen in the absence of gammadelta T cells that resulted in similar levels of IL-17A in the lungs of TCRdelta(-/-) and wild-type C57BL/6 mice. In combination, our data suggest an important role for IL-17A-expressing T lymphocytes, both gammadelta and alphabeta T cells, in eliminating this microorganism that prevents excessive inflammation and eventual lung fibrosis in this murine model of B. subtilis-induced hypersensitivity pneumonitis.
Collapse
Affiliation(s)
- Philip L Simonian
- Department of Medicine, University of Colorado Denver, Aurora, CO 80045, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
The absence of Hck, Fgr, and Lyn tyrosine kinases augments lung innate immune responses to Pneumocystis murina. Infect Immun 2009; 77:1790-7. [PMID: 19255189 DOI: 10.1128/iai.01441-08] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Src family tyrosine kinases (SFKs) phosphorylate immunotyrosine activation motifs in the cytoplasmic tail of multiple immunoreceptors, leading to the initiation of cellular effector functions, such as phagocytosis, reactive oxygen species production, and cytokine production. SFKs also play important roles in regulating these responses through the activation of immunotyrosine inhibitory motif-containing inhibitory receptors. As myeloid cells preferentially express the SFKs Hck, Fgr, and Lyn, we questioned the role of these kinases in innate immune responses to Pneumocystis murina. Increased phosphorylation of Hck was readily detectable in alveolar macrophages after stimulation with P. murina. We further observed decreased phosphorylation of Lyn on its C-terminal inhibitory tyrosine in P. murina-stimulated alveolar macrophages, indicating that SFKs were activated in alveolar macrophages in response to P. murina. Mice deficient in Hck, Fgr, and Lyn exhibited augmented clearance 3 and 7 days after intratracheal administration of P. murina, which correlated with elevated levels of interleukin 1beta (IL-1beta), IL-6, CXCL1/KC, CCL2/monocyte chemoattractant protein 1, and granulocyte colony-stimulating factor in lung homogenates and a dramatic increase in macrophage and neutrophil recruitment. Augmented P. murina clearance was also observed in Lyn(-/-) mice 3 days postchallenge, although the level was less than that observed in Hck(-/-) Fgr(-/-) Lyn(-/-) mice. A correlate to augmented clearance of P. murina in Hck(-/-) Fgr(-/-) Lyn(-/-) mice was a greater ability of alveolar macrophages from these mice to kill P. murina in vitro, suggesting that SFKs regulate the alveolar macrophage effector function against P. murina. Mice deficient in paired immunoglobulin receptor B (PIR-B), an inhibitory receptor activated by SFKs, did not exhibit enhanced inflammatory responsiveness to or clearance of P. murina. Our results suggest that SFKs regulate innate lung responses to P. murina in a PIR-B-independent manner.
Collapse
|
18
|
Kruse R, Essén-Gustavsson B, Fossum C, Jensen-Waern M. Blood concentrations of the cytokines IL-1beta, IL-6, IL-10, TNF-alpha and IFN-gamma during experimentally induced swine dysentery. Acta Vet Scand 2008; 50:32. [PMID: 18700003 PMCID: PMC2527004 DOI: 10.1186/1751-0147-50-32] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Accepted: 08/12/2008] [Indexed: 12/31/2022] Open
Abstract
Background Knowledge of the cytokine response at infection with Brachyspira hyodysenteriae can help understanding disease mechanisme involved during swine dysentery. Since this knowledge is still limited the aim of the present study was to induce dysentery experimentally in pigs and to monitor the development of important immunoregulatory cytokines in blood collected at various stages of the disease. Methods Ten conventional pigs (~23 kg) were orally inoculated with Brachyspira hyodysenteriae B204T. Eight animals developed muco-haemorrhagic diarrhoea with impaired general body condition. Blood was sampled before inoculation and repeatedly during acute dysentery and recovery periods and cytokine levels of IL-1β, IL-6, Il-10, TNF-α and IFN-γ were measured by ELISA. Results IL-1β was increased at the beginning of the dysentery period and coincided with the appearance of Serum amyloid A and clinical signs of disease. TNF-α increased in all animals after inoculation, with a peak during dysentery, and IL-6 was found in 3 animals during dysentery and in the 2 animals that did not develop clinical signs of disease. IL-10 was found in all sick animals during the recovery period. IFN-γ was not detected on any occasion. Conclusion B. hyodysenteriae inoculation induced production of systemic levels of IL-1β during the dysentery period and increased levels of IL-10 coincided with recovery from dysentery.
Collapse
|
19
|
Hernandez-Novoa B, Bishop L, Logun C, Munson PJ, Elnekave E, Rangel ZG, Barb J, Danner RL, Kovacs JA. Immune responses to Pneumocystis murina are robust in healthy mice but largely absent in CD40 ligand-deficient mice. J Leukoc Biol 2008; 84:420-30. [PMID: 18467653 DOI: 10.1189/jlb.1207816] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pneumocystis is a pathogen of immunocompromised hosts but can also infect healthy hosts, in whom infection is rapidly controlled and cleared. Microarray methods were used to examine differential gene expression in the lungs of C57BL/6 and CD40 ligand knockout (CD40L-KO) mice over time following exposure to Pneumocystis murina. Immunocompetent C57BL/6 mice, which control and clear infection efficiently, showed a robust response to infection characterized by the up-regulation of 349 primarily immune response-associated genes. Temporal changes in the expression of these genes identified an early (Week 2), primarily innate response, which waned before the infection was controlled; this was followed by primarily adaptive immune responses that peaked at Week 5, which coincided with clearance of the infection. In conjunction with the latter, there was an increased expression of B cell-associated (Ig) genes at Week 6 that persisted through 11 weeks. In contrast, CD40L-KO mice, which are highly susceptible to developing severe Pneumocystis pneumonia, showed essentially no up-regulation of immune response-associated genes at Days 35-75. Immunohistochemical staining supported these observations by demonstrating an increase in CD4+, CD68+, and CD19+ cells in C57BL/6 but not CD40L-KO mice. Thus, the healthy host demonstrates a robust, biphasic response to infection by Pneumocystis; CD40L is an essential upstream regulator of the adaptive immune responses that efficiently control infection and prevent development of progressive pneumonia.
Collapse
Affiliation(s)
- Beatriz Hernandez-Novoa
- Critical Care Medicine Department, National Institutes of Health Clinical Center, NIH, Building 10, Room 2C145, MSC 1662, Bethesda, MD 20892-1662, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Brown DA, Sawchenko PE. Time course and distribution of inflammatory and neurodegenerative events suggest structural bases for the pathogenesis of experimental autoimmune encephalomyelitis. J Comp Neurol 2007; 502:236-60. [PMID: 17348011 DOI: 10.1002/cne.21307] [Citation(s) in RCA: 165] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Murine models of experimental autoimmune encephalomyelitis (EAE) are important vehicles for studying the effects of genetic manipulation on disease processes related to multiple sclerosis (MS). Currently, a comprehensive assessment of EAE pathogenesis with respect to inflammatory and degenerating neuronal elements is lacking. By using Fluoro-jade histochemistry to mark neurodegeneration and dual immunostaining to follow T-cell, microglial, and vascular responses, the time course and distribution of pathological events in EAE was surveyed. C57BL/6J mice were killed at 7, 10, 14, 21 or 35 days after vaccination with the myelin oligodendrocyte glycoprotein peptide MOG(35-55). Disease onset occurred at day 14 and peaked at day 21. Early T-cell infiltration and microglial activation in periventricular and superficial white matter structures adjacent to meninges suggested initial recruitment of effector T cells via the cerebrospinal fluid and choroid plexus. This was associated with microglial activation at distal sites along the same white matter tracts, with subsequent vascular recruitment of T cells associated with further injury. Systematic examination of the entire CNS supported this two-step model of EAE pathogenesis, with inflammation and neurodegeneration commencing at similar times and affecting multiple levels of predominantly sensory central pathways, including their terminal fields. This included aspects of the visual, auditory/vestibular, somatosensory (lemniscal), and proprioceptive (spinocerebellar) systems. The early targeting of visual and periventricular structures followed by more widespread CNS involvement is consistent with common presenting signs in human MS patients and suggestive of a similar basis in neuropathology.
Collapse
Affiliation(s)
- David A Brown
- Laboratory of Neuronal Structure and Function, The Salk Institute for Biological Studies, and The Foundation for Medical Research, La Jolla, CA 92037, USA
| | | |
Collapse
|
21
|
Rapaka RR, Goetzman ES, Zheng M, Vockley J, McKinley L, Kolls JK, Steele C. Enhanced defense against Pneumocystis carinii mediated by a novel dectin-1 receptor Fc fusion protein. THE JOURNAL OF IMMUNOLOGY 2007; 178:3702-12. [PMID: 17339468 DOI: 10.4049/jimmunol.178.6.3702] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pneumocystis carinii (PC) pneumonia is a leading opportunistic infection found among HIV-infected individuals worldwide. Although CD4(+) T cell deficiency clearly correlates with susceptibility to PC pneumonia, murine models of disease indicate that PC-directed Abs may prevent infection and/or inhibit growth of existing PC within the lungs. Recognition of PC by alveolar macrophages involves the beta-glucan receptor Dectin-1 and macrophage effector function against PC is enhanced by Abs derived from PC-vaccinated hosts. We developed a fusion protein consisting of the extracellular domain of Dectin-1 linked to the Fc portion of murine IgG1, which we hypothesized would enhance host recognition and opsonic phagocytosis of PC. The recombinant protein, Dectin-Fc, is dimeric and the Ag recognition site identifies beta-1,3 glucan linkages specifically and with high affinity (K(D) = 2.03 x 10(-7) M). Dectin-Fc enhances RAW264.7 macrophage recognition of the beta-glucan containing particulate zymosan in an FcgammaRII- and FcgammaRIII-dependent manner and preopsonization of PC organisms with Dectin-Fc increased alveolar and peritoneal macrophage-dependent killing of PC. SCID mice treated with a replication incompetent adenoviral vector expressing Dectin-Fc had attenuated growth of PC within the lungs, overall decreased PC lung burden, and diminished correlates of PC-related lung damage relative to SCID mice receiving a control vector. These findings demonstrate that targeting PC beta-glucan with Dectin-Fc enhances host recognition and clearance of PC in the absence of B and T cells, and suggest that FcgammaR-based targeting of PC, via cell wall carbohydrate recognition, may promote resistance against PC pneumonia in the immunodeficient host.
Collapse
Affiliation(s)
- Rekha R Rapaka
- Department of Pediatrics, Division of Pulmonary Medicine, Allergy, and Immunology, Children's Hospital of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Empey KM, Hollifield M, Garvy BA. Exogenous heat-killed Escherichia coli improves alveolar macrophage activity and reduces Pneumocystis carinii lung burden in infant mice. Infect Immun 2007; 75:3382-93. [PMID: 17485459 PMCID: PMC1932967 DOI: 10.1128/iai.00174-07] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Pneumocystis carinii is an opportunistic fungal pathogen that causes life-threatening pneumonia in immunocompromised individuals. Infants appear to be particularly susceptible to Pneumocystis pulmonary infections. We have previously demonstrated that there is approximately a 3-week delay in the clearance of Pneumocystis organisms from pup mouse lungs compared to that in adults. We have further shown that there is approximately a 1-week delay in alveolar macrophage activation in pups versus adult mice. Alveolar macrophages are the primary effector cells responsible for the killing and clearance of Pneumocystis, suggesting that pup alveolar macrophages may be involved in the delayed clearance of this organism. Alveolar macrophages cultured in vitro with Pneumocystis alone demonstrate little to no activation, as indicated by a lack of cytokine production. However, when cultured with lipopolysaccharide (LPS) or zymosan, cytokine production was markedly increased, suggesting that pup alveolar macrophages are specifically unresponsive to Pneumocystis organisms rather than being intrinsically unable to become activated. Furthermore, pup mice treated with aerosolized, heat-killed Escherichia coli in vivo were able to clear Pneumocystis more efficiently than were control mice. Together, these data suggest that while pup alveolar macrophages are unresponsive to P. carinii f. sp. muris organisms, they are capable of activation by heat-killed E. coli in vivo, as well as LPS and zymosan in vitro. The lack of response of pup mice to P. carinii f. sp. muris may reflect protective mechanisms specific to the developing pup lung, but ultimately it results in insufficient clearance of Pneumocystis organisms.
Collapse
Affiliation(s)
- Kerry M Empey
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky Medical Center, 800 Rose Street, Lexington, KY 40536-0298, USA
| | | | | |
Collapse
|
23
|
Nanno M, Shiohara T, Yamamoto H, Kawakami K, Ishikawa H. gammadelta T cells: firefighters or fire boosters in the front lines of inflammatory responses. Immunol Rev 2007; 215:103-13. [PMID: 17291282 DOI: 10.1111/j.1600-065x.2006.00474.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Intradermal inoculation of cloned self-reactive alphabeta T cells into the footpads of mice induced cutaneous graft-versus-host disease (GVHD), and after recovery from GVHD, the epidermis became resistant to subsequent attempts to induce GVHD. Resistance to GVHD was not induced in the epidermis of T-cell receptor delta-deficient (TCRdelta(-/-)) mice that lacked gammadelta T cells bearing canonical Vgamma5/Vdelta1(+)gammadeltaTCRs, known as dendritic epidermal T cells (DETCs), and resistance was restored by reconstitution of these mutant mice with precursors of Vgamma5(+) DETCs. Pulmonary infection by Cryptococcus neoformans induced an increase of gammadelta T cells in the lung, and in comparison with wildtype mice, TCRdelta(-/-) mice eliminated C. neoformans more rapidly and synthesized more interferon-gamma in the lung. In the mouse small intestine, the absence of gammadelta T cells is associated with a reduction in epithelial cell turnover and downregulation of the expression of major histocompatibility complex class II molecules. The protective role of gammadelta T cells was verified in a dextran sodium sulfate-induced inflammatory bowel disease (IBD) model, whereas in a spontaneous model of IBD, gammadelta T cells were involved in the exacerbation of colitis in TCRalpha(-/-) mice. Taken together, in addition to the homeostatic regulation of epithelial tissues, gammadelta T cells appear to play a pivotal role in the modification of inflammatory responses induced in many organs containing epithelia.
Collapse
Affiliation(s)
- Masanobu Nanno
- Yakult Central Institute for Microbiological Research, Kunitachi, Tokyo, Japan
| | | | | | | | | |
Collapse
|
24
|
McKinley L, Logar AJ, McAllister F, Zheng M, Steele C, Kolls JK. Regulatory T cells dampen pulmonary inflammation and lung injury in an animal model of pneumocystis pneumonia. THE JOURNAL OF IMMUNOLOGY 2006; 177:6215-26. [PMID: 17056551 PMCID: PMC3912571 DOI: 10.4049/jimmunol.177.9.6215] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
CD4+CD25+FoxP3+ regulatory T cells are decreased in patients infected with HIV and have been shown to be critical in mediating Ag tolerance in the lung. Because a subset of Pneumocystis-infected individuals develop substantial lung injury, which can be modeled in immune reconstituted scid mice, we used mouse models of Pneumocystis carinii to investigate the role of regulatory T cells in opportunistic infection and immune reconstitution. In this study, we show that CD4+CD25+FoxP3+ cells are part of the host response to Pneumocystis in CD4+ T cell-intact mice. Moreover, lung injury and proinflammatory Th1 and Th2 cytokine levels in the bronchoalveolar lavage fluid and lung homogenate were increased following CD4+CD25- immune reconstitution in Pneumocystis-infected SCID mice but not in CD4+CD25+ T cell-reconstituted animals. The ability of CD4+CD25+ T cells to control inflammation and injury during the course of Pneumocystis was confirmed by treatment of wild-type C57BL/6 mice with anti-CD25 mAb. These data show that CD4+CD25+ T cells control pulmonary inflammation and lung injury associated with Pneumocystis infection both in the setting of immune reconstitution as well as new acquisition of infection.
Collapse
Affiliation(s)
| | | | | | | | | | - Jay K. Kolls
- Address correspondence and reprint requests to Dr. Jay K. Kolls, Children’s Hospital of Pittsburgh, Suite 3765, 3705 Fifth Avenue, Pittsburgh, PA 15213.
| |
Collapse
|
25
|
Gigliotti F, Crow EL, Bhagwat SP, Wright TW. Sensitized CD8+ T cells fail to control organism burden but accelerate the onset of lung injury during Pneumocystis carinii pneumonia. Infect Immun 2006; 74:6310-6. [PMID: 16940142 PMCID: PMC1695514 DOI: 10.1128/iai.00668-06] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 06/21/2006] [Accepted: 08/15/2006] [Indexed: 11/20/2022] Open
Abstract
While CD8+ cells have been shown to contribute to lung injury during Pneumocystis carinii pneumonia (PCP), there are conflicting reports concerning the ability of CD8+ cells to kill P. carinii. To address these two issues, we studied the effect of the presence of CD8+ cells in two mouse models of PCP. In the reconstituted SCID mouse model, depletion of CD8+ cells in addition to CD4+ cells after reconstitution did not result in increased numbers of P. carinii cysts compared to the numbers of cysts in mice with only CD4+ cells depleted. This result was observed regardless of whether the mice were reconstituted with naïve or P. carinii-sensitized lymphocytes. In contrast, reconstitution with sensitized lymphocytes resulted in more rapid onset of lung injury that was dependent on the presence of CD8+ cells. The course of organism replication over a 6-week period was also examined in the CD4+-T-cell-depleted and CD4+- and CD8+-T-cell-depleted mouse model of PCP. Again, the organism burdens were identical at all times regardless of whether CD8+ cells were present. Thus, in the absence of CD4+ T cells, CD8+ T cells are a key contributor to the inflammatory lung injury associated with PCP. However, we were unable to demonstrate an in vivo effect of these cells on the course of P. carinii infection.
Collapse
Affiliation(s)
- Francis Gigliotti
- Department of Pediatrics, University of Rochester School of Medicine & Dentistry, 601 Elmwood Avenue, Box 690, Rochester, NY 14642, USA.
| | | | | | | |
Collapse
|
26
|
Bocchino M, Marruchella A, Saltini C. Immunogenetics of severe respiratory infections: models for the development of new therapeutic strategies. Respiration 2006; 72:449-57. [PMID: 16210881 DOI: 10.1159/000087666] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Innate and adaptive immunity plays a critical role in the defence of the lung and other mucosal surfaces exposed to micro-organisms. Anti-microbial peptides and proteins, cytokines and chemokines are important immune weapons as they build up the protective front for the respiratory tract. The notion that susceptibility to infectious diseases may be inherited is widely accepted and, as it is the failure to activate adaptive immunity that may allow infection to become established and progress toward invasion and dissemination, the recognition of specific gene defects affecting the ability of the immune system to overcome invading pathogens may shed light upon those mechanisms of immune regulation that are playing the most critical roles. The aim of the present review is to discuss some of the advances in infection immunogenetics that may lead to identify new strategies in the development of new anti-infectious and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Marialuisa Bocchino
- Specialization School of Respiration Medicine, University of Rome Tor Vergata, Rome, Italy
| | | | | |
Collapse
|
27
|
McAllister F, Ruan S, Steele C, Zheng M, McKinley L, Ulrich L, Marrero L, Shellito JE, Kolls JK. CXCR3 and IFN protein-10 in Pneumocystis pneumonia. THE JOURNAL OF IMMUNOLOGY 2006; 177:1846-54. [PMID: 16849496 PMCID: PMC3912555 DOI: 10.4049/jimmunol.177.3.1846] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have previously shown that Tc1 CD8(+) T cells have in vitro and in vivo effector activity against Pneumocystis (PC) infection in mice. Because these cells have preferential expression of CXCR3, we investigated whether CXCR3 was required for host defense activity against PC. Mice deficient in CXCR3 but CD4(+) T cell intact, showed an initial delay but were able to clear the infectious challenge, indicating that CXCR3 signaling is not essential for clearance of PC. CD4-depleted mice had lower levels of monokine induced by IFN-gamma, IFN protein-10 (IP-10), and IFN-inducible T cell alpha-chemoattractant at day 7 of infection and are permissive to PC infection. Overexpression of IP-10 in the lungs by adenoviral gene transfer did not accelerate clearance of infection in control mice but accelerated clearance by day 28 in mice depleted of CD4(+) T cells. This effect was associated with increased recruitment of CD8(+) T to the lungs with higher CXCR3(+) expression levels and enhanced IFN-gamma secretion upon in vitro activation compared with control mice. These results indicate that the CXCR3 chemokines are part of the host defense response to PC, and that IP-10 can direct Tc1 CD8(+) T cell recruitment to the lungs and contribute to host defense against PC even in the absence of CD4(+) T cells.
Collapse
MESH Headings
- Adenoviridae/genetics
- Adenoviridae/immunology
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/microbiology
- CD4-Positive T-Lymphocytes/pathology
- Cell Movement/genetics
- Cell Movement/immunology
- Chemokine CXCL10
- Chemokines, CXC/administration & dosage
- Chemokines, CXC/genetics
- Chemokines, CXC/pharmacokinetics
- Chemokines, CXC/physiology
- Gene Transfer Techniques
- Inflammation/genetics
- Inflammation/immunology
- Inflammation/microbiology
- Interferon-gamma/biosynthesis
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Ligands
- Lung/immunology
- Lung/metabolism
- Lung/microbiology
- Lung/pathology
- Lymphocyte Depletion
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Pneumonia, Pneumocystis/genetics
- Pneumonia, Pneumocystis/immunology
- Pneumonia, Pneumocystis/microbiology
- Pneumonia, Pneumocystis/pathology
- Receptors, CXCR3
- Receptors, Chemokine/deficiency
- Receptors, Chemokine/genetics
- Receptors, Chemokine/metabolism
- Receptors, Chemokine/physiology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/microbiology
- T-Lymphocyte Subsets/pathology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/microbiology
Collapse
Affiliation(s)
- Florencia McAllister
- Children’s Hospital of Pittsburgh, University of Pittsburgh, Department of Pediatrics, Pittsburgh, PA 15213
| | - Sanbao Ruan
- Louisiana State University Health Sciences Center Gene Therapy Program, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112
| | - Chad Steele
- Children’s Hospital of Pittsburgh, University of Pittsburgh, Department of Pediatrics, Pittsburgh, PA 15213
| | - Mingquan Zheng
- Children’s Hospital of Pittsburgh, University of Pittsburgh, Department of Pediatrics, Pittsburgh, PA 15213
| | - Laura McKinley
- Children’s Hospital of Pittsburgh, University of Pittsburgh, Department of Pediatrics, Pittsburgh, PA 15213
| | - Lauren Ulrich
- Children’s Hospital of Pittsburgh, University of Pittsburgh, Department of Pediatrics, Pittsburgh, PA 15213
| | - Luis Marrero
- Louisiana State University Health Sciences Center Gene Therapy Program, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112
| | - Judd E. Shellito
- Louisiana State University Health Sciences Center Gene Therapy Program, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA 70112
| | - Jay K. Kolls
- Children’s Hospital of Pittsburgh, University of Pittsburgh, Department of Pediatrics, Pittsburgh, PA 15213
- Address correspondence and reprint requests to Dr. Jay K. Kolls, Children’s Hospital of Pittsburgh, Suite 3765, 3705 Fifth Avenue, Pittsburgh, PA 15213.
| |
Collapse
|
28
|
Miguez-Burbano MJ, Ashkin D, Rodriguez A, Duncan R, Flores M, Acosta B, Quintero N, Pitchenik A. Cellular immune response to pulmonary infections in HIV-infected individuals hospitalized with diverse grades of immunosuppression. Epidemiol Infect 2006; 134:271-8. [PMID: 16490130 PMCID: PMC2870395 DOI: 10.1017/s0950268805005030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2005] [Indexed: 01/15/2023] Open
Abstract
The lymphocyte profile of 521 HIV-infected subjects hospitalized at Jackson Memorial (2001-2002) was compared across main respiratory diseases. Study data included medical history and all laboratory evaluations performed during hospitalization. Community-acquired pneumonias (CAP, 52%), Pneumocystis jiroveci pneumonia (PCP, 24%), tuberculosis (TB, 9%) and non-tuberculous mycobacterial diseases (NTM, 12%) were the most frequent causes of admission. Patients hospitalized with PCP and NTM exhibited the lowest CD4 counts (P=0.003). PCP patients had the highest B-cell percentages (P=0.04). CAP patients had the highest CD8 and CD4 percentages and the lowest percentage of Natural Killer (NK) cells and viral burdens. TB patients exhibited the lowest NK-cell (11.4+/-6.3) and B-cell percentages (13.6+/-12) and the highest CD8 (59+/-14) percentage. NTM patients, in contrast, had the highest NK-cell percentages of the groups (19.1+/-11.6, P=0.01). Additionally, immune responses associated with respiratory pathogens differed in HIV-infected patients with CD4(+) cells above and below 200 counts.
Collapse
Affiliation(s)
- M J Miguez-Burbano
- Division of Disease Prevention, Department of Psychiatry and Behavioral Sciences, University of Miami School of Medicine, Miami, FL 33136, USA.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Meissner NN, Lund FE, Han S, Harmsen A. CD8 T cell-mediated lung damage in response to the extracellular pathogen pneumocystis is dependent on MHC class I expression by radiation-resistant lung cells. THE JOURNAL OF IMMUNOLOGY 2006; 175:8271-9. [PMID: 16339567 DOI: 10.4049/jimmunol.175.12.8271] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pneumocystis, a fungal, extracellular pathogen causes a life-threatening pneumonia in patients with severe immunodeficiencies. In the absence of CD4 T cells, Pneumocystis infection results in vigorous CD8 T cell influx into the alveolar and interstitial spaces of the lung. This response results in lung damage characterized by low pO2 and albumin leakage into the bronchoalveolar lavage fluid similar to other CD8 T cell-mediated interstitial lung diseases. How this extracellular pathogen elicits a CD8 T cell response is not clear, and it was the aim of our study to determine the Ag specificity of the recruited CD8 T cells and to determine whether MHC class I (MHC I) expression was necessary to initiate lung damage. Using an adoptive T cell-transfer model with either polyclonal wild-type CD8 T cells or transgenic influenza virus-specific CD8 T cells we found that CD8 T cell recruitment is Ag-specific and requires the continuous presence of the Pneumocystis pathogen. Bone marrow chimera experiments using Rag-1 and beta2-microglobulin-deficient mice as hosts demonstrated a requirement for MHC I expression on nonbone marrow-derived cells of the lung. This suggests either direct processing of Pneumocystis Ags by nonbone marrow-derived cells of the lung or the induction of lung damage triggered by a lung-specific autoantigen. Using perforin-, Fas-, and IFN-gamma-deficient animals, we showed that these molecules are not directly involved in the CD8-mediated lung damage. However, CD8 T cell-mediated lung damage is Ag-specific is induced by a MHC I-expressing nonbone marrow-derived cell in the lung and is dependent on the continued presence of live Pneumocystis.
Collapse
Affiliation(s)
- Nicole N Meissner
- Department of Veterinary Molecular Biology, Montana State University, Bozeman, MT 59717, USA.
| | | | | | | |
Collapse
|
30
|
Linke M, Ashbaugh A, Demland J, Koch J, Tanaka R, Walzer P. Resolution of Pneumocystis murina infection following withdrawal of corticosteroid induced immunosuppression. Microb Pathog 2006; 40:15-22. [PMID: 16321497 DOI: 10.1016/j.micpath.2005.10.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2005] [Revised: 10/09/2005] [Accepted: 10/09/2005] [Indexed: 11/24/2022]
Abstract
The host response during resolution of Pneumocystis murina infection following withdrawal of Dexamethasone (Dex) induced immunosuppression was analyzed. Mice were inoculated with P. murina and treated with Dex for 4 weeks. Treatment was stopped and mice were sacrificed at d1, d7, and d14. Control mice were treated in the same manner, but were inoculated with nonviable P. murina. P. murina was actively cleared from the lungs following withdrawal of Dex treatment. No P. murina was detected in control mice. Significantly more neutrophils, lymphocytes, macrophages, and eosinophils were recovered from the lungs of mice that had been infected with P. murina than from control mice at d7, but only neutrophils remained significantly elevated at d14. Significantly more CD4+ and CD8+ T cells were purified from the lungs of mice that had been infected with P. murina mice at d7 and d14. Cytokine levels were measured in lung lavage fluid by ELISA. TNF-alpha, IFN-gamma, IL-1, and IL-6 levels were higher in mice that had been inoculated with P. murina at all three time points. TNF-alpha and IL-1 levels did not change significantly following withdrawal of Dex treatment. Low levels of IL 6 were detected at d1, but increased significantly by d7 and d14. IFN-gamma levels peaked at d14. Chemokine message levels were measured in lung tissue by ribonuclease protection assay. MIP-1beta and IP-10 message increased between d1 and d7 and then decreased by d14. RANTES message levels increased from d1 to d7 and remained elevated at d14. Withdrawal of Dex induced immunosuppression from P. murina infected mice resulted in activation of many arms of the host response that lead to resolution of the infection.
Collapse
Affiliation(s)
- Michael Linke
- Department of Veterans Affairs Medical Center, Research Service, Cincinnati, OH 45220, USA.
| | | | | | | | | | | |
Collapse
|
31
|
McAllister F, Steele C, Zheng M, Shellito JE, Kolls JK. In vitro effector activity of Pneumocystis murina-specific T-cytotoxic-1 CD8+ T cells: role of granulocyte-macrophage colony-stimulating factor. Infect Immun 2005; 73:7450-7. [PMID: 16239546 PMCID: PMC1273863 DOI: 10.1128/iai.73.11.7450-7457.2005] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human immunodeficiency virus (HIV)-related opportunistic infections continue to occur in patients who are newly diagnosed with HIV infection, those in the early course of highly active antiretroviral therapy or nonadherent to HIV care, and other immunosuppressed individuals. One of the most common opportunistic infections in these patients is Pneumocystis pneumonia. CD8+ T cells are recruited to the lung after P. carinii infection and have been associated with both lung injury and host defense. This variability may be due to subpopulations of CD8+ T cells recruited to the lung. We have previously shown using adoptive transfer studies that in vivo-generated T-cytotoxic-1 (Tc1) CD8+ T cells, defined by the secretion of gamma interferon (IFN-gamma), have effector activity against Pneumocystis spp. in vitro as well as in vivo. To better understand the mechanisms of these effects, we generated, expanded, and tested Tc1 and Tc2 CD8+ T cells specific for P. murina ex vivo. Tc1-polarized CD8+ T cells secreted higher levels of IFN-gamma and granulocyte-macrophage colony-stimulating factor (GM-CSF) and lower levels of interleukin-4 (IL-4), IL-5, IL-10, and IL-13 than Tc2 CD8+ T cells when stimulated with P. murina antigen. Moreover, Tc1 CD8+ T cells demonstrated enhanced effector activity in a macrophage-mediated killing assay which was independent of cell contact. The augmentation in macrophage-mediated P. murina killing was significantly abrogated when GM-CSF was neutralized in the Tc1 CD8+ T cells. These data support the possibility that antigen-specific GM-CSF secretion is critical for effector activity of P. murina-specific Tc1 CD8+ T cells in vitro.
Collapse
MESH Headings
- Animals
- Cell Polarity
- Cells, Cultured
- Culture Media, Conditioned/pharmacology
- Dose-Response Relationship, Drug
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- Granulocyte-Macrophage Colony-Stimulating Factor/physiology
- Interferon-gamma/metabolism
- Macrophages, Alveolar/drug effects
- Macrophages, Alveolar/immunology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Pneumocystis/immunology
- Spleen/cytology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Florencia McAllister
- Children's Hospital of Pittsburgh, Department of Pediatrics, Suite 3765, 3705 Fifth Ave., Pittsburgh, PA 15213, USA
| | | | | | | | | |
Collapse
|
32
|
Qureshi MH, Garvy BA, Pomeroy C, Inayat MS, Oakley OR. A murine model of dual infection with cytomegalovirus and Pneumocystis carinii: Effects of virus-induced immunomodulation on disease progression. Virus Res 2005; 114:35-44. [PMID: 16002171 DOI: 10.1016/j.virusres.2005.05.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2005] [Revised: 05/19/2005] [Accepted: 05/19/2005] [Indexed: 01/16/2023]
Abstract
Despite the use of antimicrobial prophylaxis, cytomegalovirus (CMV) and Pneumocystis carinii (PC) pneumonia (PCP) are both leading causes of morbidity and mortality in immunocompromised patients. It has previously been reported that CMV infection modulates host immune responses with a variety of mechanisms which include the suppression of helper T cell functions and antigen presenting cell (APC) functions, both of which are critical for PCP resolution. However, the mechanisms of these interactions and other possible immune regulatory effects are not clearly understood. In this study, we investigated the impact of murine CMV (MCMV) induced immunomodulation on the progression of PCP in a co-infection model. Initial results show that dually infected mice had evidence of more severe PC disease, which include a greater loss of body weight, an excess lung PC burden and delayed clearance of PC from lungs, compared to mice with PC infection alone. At day 7 post-infection, dually infected mice had reduced numbers of MHC-II expressing cells in the lung interstitium and lymph nodes and reduced migration of CD11c+ cells to both the tracheobronchial lymph nodes and alveolar spaces. Dual infected mice showed elevated numbers of specific CD8 responses concomitant with a decrease in activated CD4+ T cells in both the lymph nodes and in alveolar spaces when compared to mice infected with MCMV alone. These data suggest that MCMV infection inhibits the immune responses generated against PC which contribute to the delayed clearance of the organism.
Collapse
Affiliation(s)
- Mahboob H Qureshi
- Department of Basic Sciences, College of Osteopathic Medicine, Touro University Nevada, Henderson, NV, USA
| | | | | | | | | |
Collapse
|
33
|
Andrew EM, Carding SR. Murine γδ T cells in infections: beneficial or deleterious? Microbes Infect 2005; 7:529-36. [PMID: 15777712 DOI: 10.1016/j.micinf.2004.12.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2004] [Accepted: 12/14/2004] [Indexed: 11/19/2022]
Abstract
Although the importance of gammadelta T cells in pathogen-induced immune responses is becoming increasingly apparent, it is not clear that their involvement is always of benefit to the host. Here we review evidence for the protective and damaging roles of gammadelta T cells in infection and discuss how these disparate findings might be resolved by considering the nature and properties of the pathogen, the sites of infection and conditions under which gammadelta T cell responses are initiated, and the involvement of different subsets of gammadelta T cells.
Collapse
Affiliation(s)
- Elizabeth M Andrew
- School of Biochemistry and Microbiology, The University of Leeds, Leeds LS2 9JT, UK
| | | |
Collapse
|
34
|
Atochina EN, Beck JM, Preston AM, Haczku A, Tomer Y, Scanlon ST, Fusaro T, Casey J, Hawgood S, Gow AJ, Beers MF. Enhanced lung injury and delayed clearance of Pneumocystis carinii in surfactant protein A-deficient mice: attenuation of cytokine responses and reactive oxygen-nitrogen species. Infect Immun 2004; 72:6002-11. [PMID: 15385504 PMCID: PMC517574 DOI: 10.1128/iai.72.10.6002-6011.2004] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Surfactant protein A (SP-A), a member of the collectin family, selectively binds to Pneumocystis carinii and mediates interactions between pathogen and host alveolar macrophages in vitro. To test the hypothesis that mice lacking SP-A have delayed clearance of Pneumocystis organisms and enhanced lung injury, wild-type C57BL/6 (WT) and SP-A-deficient mice (SP-A(-/-)) with or without selective CD4(+)-T-cell depletion were intratracheally inoculated with Pneumocystis organisms. Four weeks later, CD4-depleted SP-A-deficient mice had developed a more severe Pneumocystis infection than CD4-depleted WT (P. carinii pneumonia [PCP] scores of 3 versus 2, respectively). Whereas all non-CD4-depleted WT mice were free of PCP, intact SP-A(-/-) mice also had evidence of increased organism burden. Pneumocystis infection in SP-A-deficient mice was associated histologically with enhanced peribronchial and/or perivascular cellularity (score of 4 versus 2, SP-A(-/-) versus C57BL/6 mice, respectively) and a corresponding increase in bronchoalveolar lavage (BAL) cell counts. Increases in SP-D content, gamma interferon, interleukin-4, interleukin-5, and tumor necrosis factor alpha in BAL fluid occurred but were attenuated in PCP-infected SP-A(-/-) mice compared to WT mice. There were increases in total BAL NO levels in both infected groups, but nitrite levels were higher in SP-A(-/-) mice, indicating a reduction in production of higher oxides of nitrogen that was also reflected in lower levels of 3-nitrotyrosine staining in the SP-A(-/-) group. We conclude that despite increases in inflammatory cells, SP-A-deficient mice infected with P. carinii exhibit an enhanced susceptibility to the organism and attenuated production of proinflammatory cytokines and reactive oxygen-nitrogen species. These data support the concept that SP-A is a local effector molecule in the lung host defense against P. carinii in vivo.
Collapse
Affiliation(s)
- Elena N. Atochina
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - James M. Beck
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Angela M. Preston
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Angela Haczku
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Yaniv Tomer
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Seth T. Scanlon
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Trevor Fusaro
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - John Casey
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Samuel Hawgood
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Andrew J. Gow
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
| | - Michael F. Beers
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School and Veterans Affairs Medical Center, Ann Arbor, Michigan, Division of Neonatology, University of California at San Francisco, San Francisco, California
- Corresponding author. Mailing address: Pulmonary and Critical Care Division, University of Pennsylvania School of Medicine, 807 BRB II/III Bldg., 421 Curie Blvd., Philadelphia, PA 19104. Phone: (215) 898-9106. Fax: (215) 573-4469. E-mail:
| |
Collapse
|
35
|
Park F. Correction of bleeding diathesis without liver toxicity using arenaviral-pseudotyped HIV-1-based vectors in hemophilia A mice. Hum Gene Ther 2004; 14:1489-94. [PMID: 14577928 DOI: 10.1089/104303403769211691] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hemophilia A is an inheritable X-linked bleeding disorder most frequently occurring as a consequence of genetic alterations within the factor VIII (FVIII) gene. In the present study, pseudotyped human immunodeficiency virus type 1 (HIV-1)-derived lentivectors expressing hFVIII were assessed for the ability to correct the hemophilia A phenotype in FVIII knockout mice. Therapeutic levels of plasma hFVIII (1-7 ng/mL) were detected in C57B1/6 mice (4-5 weeks old) after portal vein administration of hFVIII-expressing lentivectors pseudotyped with the rhabdoviral vesicular stomatitis viral G protein (VSV-G). More importantly, transduction of hemophilia A mice with FVIII expressing lentivectors resulted in transient correction of the bleeding diathesis phenotype. Moreover, the use of alternate viral pseudotypes based on the lymphocytic choriomeningitis virus (LCMV) resulted in similar circulating levels of FVIII. Interestingly, similar doses of LCMV-pseudotyped lentiviral vectors resulted in minimal systemic or hepatic injury as measured by plasma alanine transferase (ALT), aspartate transferase (AST), and tumor necrosis factor (TNF)-alpha compared to the more commonly used envelope, VSV-G. In summary, these studies demonstrated both the potential merit of lentivectors in terms of correcting monogenic inherited disorders, and also the importance of using alternate pseudotypes, such as LCMV, to safely transfer therapeutic genes in vivo without producing adverse effects.
Collapse
Affiliation(s)
- Frank Park
- Louisiana State University Health Sciences Center, Department of Medicine, New Orleans, LA 70112, USA.
| |
Collapse
|
36
|
McAllister F, Mc Allister F, Steele C, Zheng M, Young E, Shellito JE, Marrero L, Kolls JK. T cytotoxic-1 CD8+ T cells are effector cells against pneumocystis in mice. THE JOURNAL OF IMMUNOLOGY 2004; 172:1132-8. [PMID: 14707088 DOI: 10.4049/jimmunol.172.2.1132] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Host defenses are profoundly compromised in HIV-infected hosts due to progressive depletion of CD4+ T lymphocytes. A hallmark of HIV infection is Pneumocystis carinii (PC) pneumonia. Recently, CD8+ T cells, which are recruited to the lung in large numbers in response to PC infection, have been associated with some level of host defense as well as contributing to lung injury in BALB/c mice. In this study, we show that CD8+ T cells that have a T cytotoxic-1 response to PC in BALB/c mice, as determined by secretion of IFN-gamma, have in vitro killing activity against PC and effect clearance of the organism in adoptive transfer studies. Moreover, non-T cytotoxic-1 CD8+ T cells lacked in vitro effector activity and contributed to lung injury upon adoptive transfer. This dichotomous response in CD8+ T cell response may in part explain the clinical heterogeneity in the severity of PC pneumonia.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- CD4-Positive T-Lymphocytes/cytology
- Chemokine CXCL10
- Chemokine CXCL9
- Chemokines, CXC/biosynthesis
- Cytotoxicity Tests, Immunologic
- Cytotoxicity, Immunologic
- Genetic Vectors
- Intercellular Signaling Peptides and Proteins/biosynthesis
- Interferon-gamma/administration & dosage
- Interferon-gamma/genetics
- Interferon-gamma/pharmacokinetics
- Lung/cytology
- Lung/immunology
- Lung/microbiology
- Lymphocyte Depletion
- Male
- Mice
- Mice, Inbred BALB C
- Mice, SCID
- Neutrophil Infiltration/genetics
- Neutrophil Infiltration/immunology
- Pneumocystis carinii/growth & development
- Pneumocystis carinii/immunology
- Pneumonia, Pneumocystis/immunology
- Pneumonia, Pneumocystis/microbiology
- Pneumonia, Pneumocystis/pathology
- Pneumonia, Pneumocystis/virology
- Receptors, CXCR3
- Receptors, Chemokine/biosynthesis
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/microbiology
- T-Lymphocyte Subsets/virology
- T-Lymphocytes, Regulatory/cytology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/microbiology
Collapse
Affiliation(s)
- Florencia McAllister
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh, 3705 Fifth Avenue, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Thullen TD, Ashbaugh AD, Daly KR, Linke MJ, Steele PE, Walzer PD. Sensitized splenocytes result in deleterious cytokine cascade and hyperinflammatory response in rats with Pneumocystis pneumonia despite the presence of corticosteroids. Infect Immun 2004; 72:757-65. [PMID: 14742518 PMCID: PMC321630 DOI: 10.1128/iai.72.2.757-765.2004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The immune response to the opportunistic pulmonary pathogen Pneumocystis can have beneficial and harmful effects on the host despite the presence of corticosteroids. We hypothesized that this deleterious hyperinflammatory response is associated with exaggerated cytokine production. The adoptive transfer of at least 10(7) immune splenocytes reduced the cyst count in rats with corticosteroid-induced pneumocystosis. About 18% of these rats developed clinical illness, an increased lung weight/body weight (LW/BW) ratio, and elevated levels of interleukin 1alpha (IL-1alpha), IL-1beta, IL-6, tumor necrosis factor alpha, IL-5, IL-10, and gamma interferon in the lungs. This hyperinflammatory reaction was not observed in rats that remained clinically well or in control rats. Thus, in this model, corticosteroids have little effect on the cytokine cascade or other adverse effects of the host immune response to Pneumocystis.
Collapse
|
38
|
Steele C, Marrero L, Swain S, Harmsen AG, Zheng M, Brown GD, Gordon S, Shellito JE, Kolls JK. Alveolar macrophage-mediated killing of Pneumocystis carinii f. sp. muris involves molecular recognition by the Dectin-1 beta-glucan receptor. ACTA ACUST UNITED AC 2004; 198:1677-88. [PMID: 14657220 PMCID: PMC2194130 DOI: 10.1084/jem.20030932] [Citation(s) in RCA: 226] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Innate immune mechanisms against Pneumocystis carinii, a frequent cause of pneumonia in immunocompromised individuals, are not well understood. Using both real time polymerase chain reaction as a measure of organism viability and fluorescent deconvolution microscopy, we show that nonopsonic phagocytosis of P. carinii by alveolar macrophages is mediated by the Dectin-1 β-glucan receptor and that the subsequent generation of hydrogen peroxide is involved in alveolar macrophage–mediated killing of P. carinii. The macrophage Dectin-1 β-glucan receptor colocalized with the P. carinii cyst wall. However, blockage of Dectin-1 with high concentrations of anti–Dectin-1 antibody inhibited binding and concomitant killing of P. carinii by alveolar macrophages. Furthermore, RAW 264.7 macrophages overexpressing Dectin-1 bound P. carinii at a higher level than control RAW cells. In the presence of Dectin-1 blockage, killing of opsonized P. carinii could be restored through FcγRII/III receptors. Opsonized P. carinii could also be efficiently killed in the presence of FcγRII/III receptor blockage through Dectin-1–mediated phagocytosis. We further show that Dectin-1 is required for P. carinii–induced macrophage inflammatory protein 2 production by alveolar macrophages. Taken together, these results show that nonopsonic phagocytosis and subsequent killing of P. carinii by alveolar macrophages is dependent upon recognition by the Dectin-1 β-glucan receptor.
Collapse
Affiliation(s)
- Chad Steele
- Department of Pediatrics, Division of Pulmonology, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Experimental models of pulmonary infection are being discussed, focused on various aspects of good experimental design, such as choice of animal species and infecting strain, and route of infection/inoculation techniques (intranasal inoculation, aerosol inoculation, and direct instillation into the lower respiratory tract). In addition, parameters to monitor pulmonary infection are being reviewed such as general clinical signs, pulmonary-associated signs, complication of the pulmonary infection, mortality rate, and parameters after dissection of animals. Examples of pulmonary infection models caused by bacteria, fungi, viruses or parasites in experimental animals with intact or impaired host defense mechanisms are shortly summarized including key-references.
Collapse
Affiliation(s)
- Irma A J M Bakker-Woudenberg
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC, P.O. Box 1738, 3000 DR Rotterdam, The Netherlands.
| |
Collapse
|
40
|
Happel KI, Zheng M, Young E, Quinton LJ, Lockhart E, Ramsay AJ, Shellito JE, Schurr JR, Bagby GJ, Nelson S, Kolls JK. Cutting edge: roles of Toll-like receptor 4 and IL-23 in IL-17 expression in response to Klebsiella pneumoniae infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4432-6. [PMID: 12707317 PMCID: PMC2841978 DOI: 10.4049/jimmunol.170.9.4432] [Citation(s) in RCA: 363] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Local production of IL-17 is a significant factor in effective host defense against Gram-negative bacteria. However, the proximal events mediating IL-17 elaboration by T cells remain unclear. In this study, we show in vivo that intact Toll-like receptor 4 signaling in the lung is required for induction of both the p19 transcript of IL-23 and IL-17 protein elaboration in response to Klebsiella pneumoniae. Although IL-17 is widely considered a CD4(+) T cell product, we also demonstrate significant in vitro IL-17 production by CD8(+) T cells after culture in medium from dendritic cells exposed to these bacteria. The dominant portion of this IL-17-inducing activity for both CD4(+) and CD8(+) T cells is IL-23. These data demonstrate the critical signaling pathway for IL-17 induction in the host response to Gram-negative pulmonary infection and suggest a direct role for IL-23 in CD8(+) T cell IL-17 production.
Collapse
Affiliation(s)
- Kyle I. Happel
- Section of Pulmonary and Critical Care Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112
- Alcohol Research Center, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Mingquan Zheng
- Gene Therapy Program, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Erana Young
- Gene Therapy Program, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Lee J. Quinton
- Alcohol Research Center, Louisiana State University Health Science Center, New Orleans, LA 70112
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Euan Lockhart
- Gene Therapy Program, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Alistair J. Ramsay
- Section of Pulmonary and Critical Care Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112
- Gene Therapy Program, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Judd E. Shellito
- Section of Pulmonary and Critical Care Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112
- Alcohol Research Center, Louisiana State University Health Science Center, New Orleans, LA 70112
- Gene Therapy Program, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Jill R. Schurr
- Gene Therapy Program, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Gregory J. Bagby
- Section of Pulmonary and Critical Care Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112
- Alcohol Research Center, Louisiana State University Health Science Center, New Orleans, LA 70112
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Steve Nelson
- Section of Pulmonary and Critical Care Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112
- Alcohol Research Center, Louisiana State University Health Science Center, New Orleans, LA 70112
- Department of Physiology, Louisiana State University Health Science Center, New Orleans, LA 70112
| | - Jay K. Kolls
- Section of Pulmonary and Critical Care Medicine, Louisiana State University Health Science Center, New Orleans, LA 70112
- Alcohol Research Center, Louisiana State University Health Science Center, New Orleans, LA 70112
- Gene Therapy Program, Louisiana State University Health Science Center, New Orleans, LA 70112
- Address correspondence and reprint requests to Dr. Jay Kolls, Louisiana State University Health Science Center, Clinical Sciences Research Building, Room 601, 533 Bolivar Street, New Orleans, LA 70112.
| |
Collapse
|
41
|
Hedges JF, Cockrell D, Jackiw L, Meissner N, Jutila MA. Differential mRNA expression in circulating gammadelta T lymphocyte subsets defines unique tissue-specific functions. J Leukoc Biol 2003; 73:306-14. [PMID: 12554808 DOI: 10.1189/jlb.0902453] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
To elucidate the functions of circulating gammadelta T cells, in the absence of antigen stimulation, the differential gene expression of two circulating gammadelta T cell subsets was analyzed. The two subsets, with distinct trafficking phenotypes in young calves, were GD3.5(+), CD8(-), WC1(+) or GD3.5(-), CD2(+), WC1(-), and 90-100% CD8(+) and were sorted based on GD3.5 and gammadelta T cell receptor expression. Results from two different human arrays probed with cDNA from these gammadelta T cell subsets indicated that they have markedly different tissue-specific functions. The genes preferentially expressed by GD3.5(+) (CD8(-)) gammadelta T cells demonstrated that they were highly activated, proliferative, and inflammatory, whereas those expressed by GD3.5(-) (primarily CD8(+)) gammadelta T cells were involved in promoting quiescence, consistent with a role for gammadelta T cells as sentinel mucosal cells, and several were interferon-regulated genes. Gene expression and phenotypic assays indicated that CD8(+) gammadelta T cells were apoptotic, whereas CD8(-) gammadelta T cells were apoptosis-resistant. Differential expression of multiple genes was confirmed in both arrays: That of 14 genes was confirmed by quantitative reverse transcriptase-polymerase chain reaction and that of seven proteins was confirmed by flow cytometry. This novel, genomic analysis of circulating gammadelta T cell subsets, without confounding effects of the tissue microenvironment, offers new insight into the biology and development of neonatal gammadelta T cells.
Collapse
Affiliation(s)
- Jodi F Hedges
- Department of Veterinary Molecular Biology, Marsh Laboratory, Montana State University, 19th and Lincoln, Bozeman, MT 59717, USA
| | | | | | | | | |
Collapse
|