1
|
Abstract
Pneumonia is a type of acute lower respiratory infection that is common and severe. The outcome of lower respiratory infection is determined by the degrees to which immunity is protective and inflammation is damaging. Intercellular and interorgan signaling networks coordinate these actions to fight infection and protect the tissue. Cells residing in the lung initiate and steer these responses, with additional immunity effectors recruited from the bloodstream. Responses of extrapulmonary tissues, including the liver, bone marrow, and others, are essential to resistance and resilience. Responses in the lung and extrapulmonary organs can also be counterproductive and drive acute and chronic comorbidities after respiratory infection. This review discusses cell-specific and organ-specific roles in the integrated physiological response to acute lung infection, and the mechanisms by which intercellular and interorgan signaling contribute to host defense and healthy respiratory physiology or to acute lung injury, chronic pulmonary disease, and adverse extrapulmonary sequelae. Pneumonia should no longer be perceived as simply an acute infection of the lung. Pneumonia susceptibility reflects ongoing and poorly understood chronic conditions, and pneumonia results in diverse and often persistent deleterious consequences for multiple physiological systems.
Collapse
Affiliation(s)
- Lee J Quinton
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Allan J Walkey
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
2
|
Oliveira da Silva C, Monte-Alto-Costa A, Renovato-Martins M, Viana Nascimento FJ, Dos Santos Valença S, Lagente V, Pôrto LC, Victoni T. Time Course of the Phenotype of Blood and Bone Marrow Monocytes and Macrophages in the Lung after Cigarette Smoke Exposure In Vivo. Int J Mol Sci 2017; 18:ijms18091940. [PMID: 28891938 PMCID: PMC5618589 DOI: 10.3390/ijms18091940] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/30/2017] [Accepted: 09/04/2017] [Indexed: 12/20/2022] Open
Abstract
Alveolar macrophages play a central role in the pathogenesis of chronic obstructive pulmonary disease (COPD). Monocytes are recruited from blood during inflammation and then mature into alveolar macrophages. The aim of this study was to investigate the effect of cigarette smoke (CS) at different times in lung macrophages and monocytes from blood and bone marrow in mice. Male mice (C57BL/6, n = 45) were divided into groups: control, CS 5 days, CS 14 days and CS 30 days. Five days’ CS exposure induced a pronounced influx of neutrophils and macrophages in the lung associated with increased levels of keratinocyte chemoattractant (KC), tumor necrosis factor-α (TNF-α), nitric oxide (NO) and matrix metalloproteinase (MMP)-12. After 14 days of CS exposure, neutrophil recruitment and cytokine production were greatly reduced. Moreover, chronic CS exposure led to increased recruitment of macrophages (with high expression of CD206), transforming growth factor-β (TGF-β) production as well as no detection of TNF-α, interleukin (IL)-6 and KC. CS can also change the monocyte phenotype in the blood and bone marrow, with an increase in Ly6Clow cells. These results show for the first time that CS can change not only macrophage polarization but also monocyte. These results suggest that continued recruitment of Ly6Clow monocytes may help the distinct renewing macrophage M2 population required for COPD progression.
Collapse
Affiliation(s)
- Camila Oliveira da Silva
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
| | - Andréa Monte-Alto-Costa
- Laboratório de Reparo Tecidual, DHE/IBRAG/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-003, Brazil.
| | - Mariana Renovato-Martins
- Instituto de Biologia Roberto Alcântara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20551-030, Brazil.
| | - Filipe Jorge Viana Nascimento
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
| | - Samuel Dos Santos Valença
- Laboratório de Biologia Redox, ICB/Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ 21941-902, Brazil.
| | - Vincent Lagente
- Nutrition, Métabolismes et Cancer, NUMECAN Unité, Institut national de la santé et de la recherche médicale, INSERM 1241/Institut national de la recherche agronomique, INRA 1341/Université de Rennes 1, 35000 Rennes, France.
| | - Luís Cristóvão Pôrto
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
| | - Tatiana Victoni
- Laboratório e Histocompatibilidade e Criopreservação, HLA/Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ 20950-000, Brazil.
- Nutrition, Métabolismes et Cancer, NUMECAN Unité, Institut national de la santé et de la recherche médicale, INSERM 1241/Institut national de la recherche agronomique, INRA 1341/Université de Rennes 1, 35000 Rennes, France.
| |
Collapse
|
3
|
Brazee PL, Soni PN, Tokhtaeva E, Magnani N, Yemelyanov A, Perlman HR, Ridge KM, Sznajder JI, Vagin O, Dada LA. FXYD5 Is an Essential Mediator of the Inflammatory Response during Lung Injury. Front Immunol 2017; 8:623. [PMID: 28620381 PMCID: PMC5451504 DOI: 10.3389/fimmu.2017.00623] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 05/10/2017] [Indexed: 12/28/2022] Open
Abstract
The alveolar epithelium secretes cytokines and chemokines that recruit immune cells to the lungs, which is essential for fighting infections but in excess can promote lung injury. Overexpression of FXYD5, a tissue-specific regulator of the Na,K-ATPase, in mice, impairs the alveolo-epithelial barrier, and FXYD5 overexpression in renal cells increases C-C chemokine ligand-2 (CCL2) secretion in response to lipopolysaccharide (LPS). The aim of this study was to determine whether FXYD5 contributes to the lung inflammation and injury. Exposure of alveolar epithelial cells (AEC) to LPS increased FXYD5 levels at the plasma membrane, and FXYD5 silencing prevented both the activation of NF-κB and the secretion of cytokines in response to LPS. Intratracheal instillation of LPS into mice increased FXYD5 levels in the lung. FXYD5 overexpression increased the recruitment of interstitial macrophages and classical monocytes to the lung in response to LPS. FXYD5 silencing decreased CCL2 levels, number of cells, and protein concentration in bronchoalveolar lavage fluid (BALF) after LPS treatment, indicating that FXYD5 is required for the NF-κB-stimulated epithelial production of CCL2, the influx of immune cells, and the increase in alveolo-epithelial permeability in response to LPS. Silencing of FXYD5 also prevented the activation of NF-κB and cytokine secretion in response to interferon α and TNF-α, suggesting that pro-inflammatory effects of FXYD5 are not limited to the LPS-induced pathway. Furthermore, in the absence of other stimuli, FXYD5 overexpression in AEC activated NF-κB and increased cytokine production, while FXYD5 overexpression in mice increased cytokine levels in BALF, indicating that FXYD5 is sufficient to induce the NF-κB-stimulated cytokine secretion by the alveolar epithelium. The FXYD5 overexpression also increased cell counts in BALF, which was prevented by silencing the CCL2 receptor (CCR2), or by treating mice with a CCR2-blocking antibody, confirming that FXYD5-induced CCL2 production leads to the recruitment of monocytes to the lung. Taken together, the data demonstrate that FXYD5 is a key contributor to inflammatory lung injury.
Collapse
Affiliation(s)
- Patricia L Brazee
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Pritin N Soni
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Elmira Tokhtaeva
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Natalia Magnani
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Alex Yemelyanov
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Harris R Perlman
- Division of Rheumatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Karen M Ridge
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jacob I Sznajder
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Olga Vagin
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States.,Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Laura A Dada
- Pulmonary and Critical Care Division, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
4
|
Prohl A, Wolf K, Weber C, Müller KE, Menge C, Sachse K, Rödel J, Reinhold P, Berndt A. Kinetics of Local and Systemic Leucocyte and Cytokine Reaction of Calves to Intrabronchial Infection with Chlamydia psittaci. PLoS One 2015; 10:e0135161. [PMID: 26252769 PMCID: PMC4529195 DOI: 10.1371/journal.pone.0135161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 07/18/2015] [Indexed: 11/19/2022] Open
Abstract
Infection of cattle with chlamydiae is ubiquitous and, even in the absence of clinical sequeleae, has a quantifiable negative impact on livestock productivity. Despite recent progress, our knowledge about immune response mechanisms capable of counteracting the infection and preventing its detrimental effects is still limited. A well-established model of bovine acute respiratory Chlamydia (C.) psittaci infection was used here to characterize the kinetics of the local and systemic immune reactions in calves. In the course of two weeks following inoculation, leukocyte surface marker expression was monitored by flow cytometry in blood and bronchoalveolar lavage fluid (BALF). Immune-related protein and receptor transcription were determined by quantitative real-time reverse transcription PCR in blood, BALF and lung tissue. An early increase of IL2RA, IL10 and HSPA1A mRNA expressions was followed by a rise of lymphocytes, monocytes, and granulocytes exhibiting activated phenotypes in blood. Monocytes showed elevated expression rates of CD11b, CD14 and MHC class II. The rates of CD62L expression on CD8hi T cells in blood and on CD4+ T cells in BALF were also augmented and peaked between 2 and 4 dpi. Notably, CD25 antigen expression was significantly elevated, not only on CD8dim/CD62L+ and CD8-/CD62L+ cells in blood, but also on granulocytes in blood and BALF between 2–3 dpi. From 4 dpi onwards, changes declined and the calves recovered from the infection until 10 dpi. The findings highlight the effectiveness of rapid local and systemic immune reaction and indicate activated T cells, monocytes and granulocytes being essential for rapid eradication of the C. psittaci infection.
Collapse
Affiliation(s)
- Annette Prohl
- Institute of Molecular Pathogenesis at ‘Friedrich-Loeffler-Institut’ (Federal Research Institute for Animal Health), Jena, Germany
| | - Katharina Wolf
- Institute of Medical Microbiology, Friedrich Schiller University of Jena, Jena, Germany
| | - Corinna Weber
- Ruminant and Swine Clinic at Freie Universität Berlin, Berlin, Germany
| | - Kerstin E. Müller
- Ruminant and Swine Clinic at Freie Universität Berlin, Berlin, Germany
| | - Christian Menge
- Institute of Molecular Pathogenesis at ‘Friedrich-Loeffler-Institut’ (Federal Research Institute for Animal Health), Jena, Germany
| | - Konrad Sachse
- Institute of Molecular Pathogenesis at ‘Friedrich-Loeffler-Institut’ (Federal Research Institute for Animal Health), Jena, Germany
| | - Jürgen Rödel
- Institute of Medical Microbiology, Friedrich Schiller University of Jena, Jena, Germany
| | - Petra Reinhold
- Institute of Molecular Pathogenesis at ‘Friedrich-Loeffler-Institut’ (Federal Research Institute for Animal Health), Jena, Germany
- * E-mail:
| | - Angela Berndt
- Institute of Molecular Pathogenesis at ‘Friedrich-Loeffler-Institut’ (Federal Research Institute for Animal Health), Jena, Germany
| |
Collapse
|
5
|
Kaya H, Zorlu A, Yucel H, Dogan OT, Sarikaya S, Aydin G, Kivrak T, Yilmaz MB. Cancer antigen-125 levels predict long-term mortality in chronic obstructive pulmonary disease. Biomarkers 2015; 20:162-7. [DOI: 10.3109/1354750x.2015.1045033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
| | | | | | - Omer Tamer Dogan
- Department of Chest Disease, Cumhuriyet University Medical School, Sivas, Turkey,
| | - Savas Sarikaya
- Department of Cardiology, Bozok University Medical School, Yozgat, Turkey,
| | - Gulay Aydin
- Department of Cardiology, Unye State Hospital, Ordu, Turkey, and
| | - Tarik Kivrak
- Department of Cardiology, Sivas State Hospital, Sivas, Turkey
| | | |
Collapse
|
6
|
Xiang Y, Yan H, Zhou J, Zhang Q, Hanley G, Caudle Y, LeSage G, Zhang X, Yin D. The role of toll-like receptor 9 in chronic stress-induced apoptosis in macrophage. PLoS One 2015; 10:e0123447. [PMID: 25885582 PMCID: PMC4401452 DOI: 10.1371/journal.pone.0123447] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 03/03/2015] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence implied that chronic stress has been exerting detrimental impact on immune system functions in both humans and animals. Toll-like receptors (TLRs) have been shown to play an essential role in modulating immune responses and cell survival. We have recently shown that TLR9 deficiency protects against lymphocyte apoptosis induced by chronic stress. However, the exact role of TLR9 in stress-mediated change of macrophage function remains unclear. The results of the current study showed that when BALB/c mice were treated with restraint stress (12 h daily for 2 days), the number of macrophages recruited to the peritoneal cavity was obviously increased. Results also demonstrated that the sustained effects of stress elevated cytokine IL-1β, TNF-α and IL-10 production yet diminished IFN-γ production from macrophage, which led to apoptotic cell death. However, TLR9 deficiency prevented the chronic stress-mediated accumulation of macrophages. In addition, knocking out TLR9 significantly abolished the chronic stress-induced imbalance of cytokine levels and apoptosis in macrophage. TLR9 deficiency was also found to reverse elevation of plasma IL-1β, IL-10 and IL-17 levels and decrease of plasma IFN-γ level under the condition of chronic stress. These results indicated that TLR9-mediated macrophage responses were required for chronic stress-induced immunosuppression. Further exploration showed that TLR9 deficiency prevented the increment of p38 MAPK phosphorylation and reduction of Akt/Gsk-3β phosphorylation; TLR9 deficiency also attenuated the release of mitochondrial cytochrome c into cytoplasm, caused upregulation of Bcl-2/Bax protein ratio, downregulation of cleavage of caspase-3 and PARP, as well as decreased TUNEL-positive cells in macrophage of stressed mice. Collectively, our studies demonstrated that deficiency of TLR9 maintained macrophage function by modulating macrophage accumulation and attenuating macrophage apoptosis, thus preventing immunosuppression in restraint-stressed mice.
Collapse
Affiliation(s)
- Yanxiao Xiang
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, United States of America
- Department of Pharmacology, Shandong University School of Medicine, Jinan, People's Republic of China
| | - Hui Yan
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, United States of America
| | - Jun Zhou
- Department of Radiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Qi Zhang
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, United States of America
| | - Gregory Hanley
- Laboratory Animal Resources, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, United States of America
| | - Yi Caudle
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, United States of America
| | - Gene LeSage
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, United States of America
| | - Xiumei Zhang
- Department of Pharmacology, Shandong University School of Medicine, Jinan, People's Republic of China
- * E-mail: (XZ); (DY)
| | - Deling Yin
- Department of Internal Medicine, College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614, United States of America
- * E-mail: (XZ); (DY)
| |
Collapse
|
7
|
Lilja A, Weeden CE, McArthur K, Nguyen T, Donald A, Wong ZX, Dousha L, Bozinovski S, Vlahos R, Burns CJ, Asselin-Labat ML, Anderson GP. HSP90 inhibition suppresses lipopolysaccharide-induced lung inflammation in vivo. PLoS One 2015; 10:e0114975. [PMID: 25615645 PMCID: PMC4304786 DOI: 10.1371/journal.pone.0114975] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 11/15/2014] [Indexed: 11/18/2022] Open
Abstract
Inflammation is an important component of cancer diathesis and treatment-refractory inflammation is a feature of many chronic degenerative lung diseases. HSP90 is a 90kDa protein which functions as an ATP-dependent molecular chaperone that regulates the signalling conformation and expression of multiple protein client proteins especially oncogenic mediators. HSP90 inhibitors are in clinical development as cancer therapies but the myeleosuppressive and neutropenic effect of first generation geldanamycin-class inhibitors has confounded studies on the effects on HSP90 inhibitors on inflammation. To address this we assessed the ability of Ganetespib, a non-geldanamycin HSP90 blocker, to suppress lipopolysaccharide (LPS)-induced cellular infiltrates, proteases and inflammatory mediator and transcriptional profiles. Ganetespib (10-100 mg/kg, i.v.) did not directly cause myelosuppression, as assessed by video micrography and basal blood cell count, but it strongly and dose-dependently suppressed LPS-induced neutrophil mobilization into blood and neutrophil- and mononuclear cell-rich steroid-refractory lung inflammation. Ganetespib also suppressed B cell and NK cell accumulation, inflammatory cytokine and chemokine induction and MMP9 levels. These data identify non-myelosuppresssive HSP90 inhibitors as potential therapies for inflammatory diseases refractory to conventional therapy, in particular those of the lung.
Collapse
Affiliation(s)
- Andrew Lilja
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Clare E. Weeden
- Division of ACRF Stem Cells and Cancer, the Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia
| | - Kate McArthur
- Division of Chemical Biology, the Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; The Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Thao Nguyen
- Division of Chemical Biology, the Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; The Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Alastair Donald
- Division of Chemical Biology, the Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; The Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Zi Xin Wong
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Lovisa Dousha
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Steve Bozinovski
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Ross Vlahos
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Christopher J. Burns
- Division of Chemical Biology, the Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; The Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Marie-Liesse Asselin-Labat
- Division of ACRF Stem Cells and Cancer, the Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia
- * E-mail: (GPA); (MLAL)
| | - Gary P. Anderson
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, 3010 VIC, Australia
- * E-mail: (GPA); (MLAL)
| |
Collapse
|
8
|
Pérez-Rial S, del Puerto-Nevado L, Terrón-Expósito R, Girón-Martínez Á, González-Mangado N, Peces-Barba G. Role of recently migrated monocytes in cigarette smoke-induced lung inflammation in different strain of mice. PLoS One 2013; 8:e72975. [PMID: 24058452 PMCID: PMC3772796 DOI: 10.1371/journal.pone.0072975] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 07/15/2013] [Indexed: 11/19/2022] Open
Abstract
This study investigates the role of proinflammatory monocytes recruited from blood circulation and recovered in bronchoalveolar lavage (BAL) fluid in mediating the lung damage in a model of acute cigarette smoke (CS)-induced lung inflammation in two strains of mice with different susceptibility to develop emphysema (susceptible -C57BL/6J and non susceptible -129S2/SvHsd). Exposure to whole-body CS for 3 consecutive research cigarettes in one single day induced acute inflammation in the lung of mice. Analysis of BAL fluid showed more influx of recently migrated monocytes at 72 h after CS-exposition in susceptible compared to non susceptible mice. It correlated with an increase in MMP-12 and TNF-α protein levels in the lung tissue, and with an increment of NF-κB translocation to the nucleus measured by electrophoretic mobility shift assay in C57BL/6J mice. To determine the functional role of these proinflammatory monocytes in mediating CS-induced airway inflammation, alveolar macrophages and blood monocytes were transiently removed by pretreatment with intratracheal and intravenous liposome-encapsulated CL2MDP, given 2 and 4 days prior to CS exposure and their repopulation was studied. Monocytes/macrophages were maximally depleted 48 h after last liposome application and subsequently recently migrated monocytes reappeared in BAL fluid of susceptible mice at 72 h after CS exposure. Recently migrated monocytes influx to the lung correlated with an increase in the MMP-12 protein level in the lung tissue, indicating that the increase in proinflammatory monocytes is associated with a major tissue damaging. Therefore our data confirm that the recruitment of proinflammatory recently migrated monocytes from the blood are responsible for the increase in MMP-12 and has an important role in the pathogenesis of lung disease induced by acute lung inflammation. These results could contribute to understanding the different susceptibility to CS of these strains of mice.
Collapse
Affiliation(s)
- Sandra Pérez-Rial
- Respiratory Research Group, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES (IIS-FJD-CIBERES), Madrid, Spain
| | - Laura del Puerto-Nevado
- Respiratory Research Group, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES (IIS-FJD-CIBERES), Madrid, Spain
| | - Raúl Terrón-Expósito
- Respiratory Research Group, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES (IIS-FJD-CIBERES), Madrid, Spain
| | - Álvaro Girón-Martínez
- Respiratory Research Group, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES (IIS-FJD-CIBERES), Madrid, Spain
| | - Nicolás González-Mangado
- Respiratory Research Group, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES (IIS-FJD-CIBERES), Madrid, Spain
| | - Germán Peces-Barba
- Respiratory Research Group, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-CIBERES (IIS-FJD-CIBERES), Madrid, Spain
| |
Collapse
|
9
|
Duan M, Li WC, Vlahos R, Maxwell MJ, Anderson GP, Hibbs ML. Distinct macrophage subpopulations characterize acute infection and chronic inflammatory lung disease. THE JOURNAL OF IMMUNOLOGY 2012; 189:946-55. [PMID: 22689883 DOI: 10.4049/jimmunol.1200660] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Although great progress has been made in delineating lung dendritic cell and lymphocyte subpopulations, similar advances in lung macrophages (MΦs) have been hampered by their intrinsic autofluorescence, cell plasticity, and the complexities of monocyte-MΦ compartmentalization. Using spectral scanning, we define alveolar MΦ autofluorescence characteristics, which has allowed us to develop an alternative flow cytometry method. Using this methodology, we show that mouse lung MΦs form distinct subpopulations during acute inflammation after challenge with LPS or influenza virus, and in chronic inflammatory lung disease consequent to SHIP-1 deletion. These subpopulations are distinguished by differential Mac-1 and CD11c integrin expression rather than classical M1 or M2 markers, and display differential gene signatures ex vivo. Whereas the resolution of acute inflammation is characterized by restoration to a homogenous population of CD11c(high)Mac-1(neg/low) MΦs reflective of lung homeostasis, chronic inflammatory lung disease associated with SHIP-1 deficiency is accompanied by an additional subpopulation of CD11c(high)Mac-1(pos) MΦs that tracks with lung disease in susceptible genetic background SHIP-1(-/-) animals and disease induction in chimeric mice. These findings may help better understand the roles of MΦ subpopulations in lung homeostasis and disease.
Collapse
Affiliation(s)
- Mubing Duan
- Ludwig Institute for Cancer Research, Melbourne, Victoria, Australia
| | | | | | | | | | | |
Collapse
|
10
|
Understanding delayed T-cell priming, lung recruitment, and airway luminal T-cell responses in host defense against pulmonary tuberculosis. Clin Dev Immunol 2012; 2012:628293. [PMID: 22545059 PMCID: PMC3321538 DOI: 10.1155/2012/628293] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 01/18/2012] [Indexed: 11/18/2022]
Abstract
Mycobacterium tuberculosis (M.tb), the causative bacterium of pulmonary tuberculosis (TB), is a serious global health concern. Central to M.tb effective immune avoidance is its ability to modulate the early innate inflammatory response and prevent the establishment of adaptive T-cell immunity for nearly three weeks. When compared with other intracellular bacterial lung pathogens, such as Legionella pneumophila, or even closely related mycobacterial species such as M. smegmatis, this delay is astonishing. Customarily, the alveolar macrophage (AM) acts as a sentinel, detecting and alerting surrounding cells to the presence of an invader. However, in the case of M.tb, this may be impaired, thus delaying the recruitment of antigen-presenting cells (APCs) to the lung. Upon uptake by APC populations, M.tb is able to subvert and delay the processing of antigen, MHC class II loading, and the priming of effector T cell populations. This delay ultimately results in the deferred recruitment of effector T cells to not only the lung interstitium but also the airway lumen. Therefore, it is of upmost importance to dissect the mechanisms that contribute to the delayed onset of immune responses following M.tb infection. Such knowledge will help design the most effective vaccination strategies against pulmonary TB.
Collapse
|
11
|
Kallapur SG, Kramer BW, Nitsos I, Pillow JJ, Collins JJP, Polglase GR, Newnham JP, Jobe AH. Pulmonary and systemic inflammatory responses to intra-amniotic IL-1α in fetal sheep. Am J Physiol Lung Cell Mol Physiol 2011; 301:L285-95. [PMID: 21665964 DOI: 10.1152/ajplung.00446.2010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Clinical and epidemiological studies implicate IL-1 as an important mediator of perinatal inflammation. We tested the hypothesis that intra-amniotic IL-1α would induce pulmonary and systemic fetal inflammatory responses. Sheep with singleton fetuses were given an intra-amniotic injection of recombinant sheep IL-1α (100 μg) and were delivered 1, 3, or 7 days later, at 124 ± 1 days gestation (n=5-8/group). A separate group of sheep were given two intra-amniotic IL-1α injections (100 μg dose each): 7 days and again 1 day prior to delivery. IL-1α induced a robust increase in monocytes, neutrophils, lymphocytes, and IL-8 protein in bronchoalveolar lavage fluid. H(2)O(2) secretion was increased in inflammatory cells isolated from lungs of IL-1α-exposed lambs upon LPS challenge in vitro compared with control monocytes. T lymphocytes were recruited to the lung. IL-1β, cyclooxygenase-1, and cyclooxygenase-2 mRNA expression increased in the lung 1 day after intra-amniotic IL-1α exposure. Lung volumes increased 7 days after intra-amniotic IL-1α exposure, with minimal anatomic changes in air space morphology. The weight of the posterior mediastinal lymph node draining the lung and the gastrointestinal tract doubled, inducible nitric oxide synthase (NOSII)-positive cells increased, and Foxp3-positive T-regulatory lymphocytes decreased in the lymph node after IL-1α exposure. In the blood, neutrophil counts and plasma haptoglobin increased after IL-1α exposure. Compared with a single exposure, exposure to intra-amniotic IL-1α 7 days and again 1 day before delivery had a variable effect (increases in some inflammatory markers, but not pulmonary cytokines). IL-1α is a potent mediator of the fetal inflammatory response syndrome.
Collapse
Affiliation(s)
- Suhas G Kallapur
- Cincinnati Children's Hospital Medical Center, Univ. of Cincinnati, Division of Pulmonary Biology, 3333 Burnet Ave., Cincinnati, OH 45229-3039, USA.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Immunolocalization of Pulmonary Intravascular Macrophages, TLR4, TLR9 and IL-8 in Normal and Pasteurella multocida-infected Lungs of Water Buffalo (Bubalus bubalis). J Comp Pathol 2011; 144:135-44. [DOI: 10.1016/j.jcpa.2010.08.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 05/31/2010] [Accepted: 08/08/2010] [Indexed: 01/20/2023]
|
13
|
Azevedo RB, Valois CRA, Chaves SB, Silva JR, Garcia MP. Leukocyte transepithelial migration in lung induced by DMSA functionalized magnetic nanoparticles. Cell Adh Migr 2011; 5:29-33. [PMID: 20935452 DOI: 10.4161/cam.5.1.13558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Magnetic nanoparticles surface-covered with meso-2,3-dimercaptosuccinic acid (MNPs-DMSA) constitute a promising approach for tissue- and cell-targeted delivery of therapeutic drugs in the lung. However, they can also induce a transient transendothelial migration of leukocytes in the organ as a side effect after endovenous administration of MNPs-DMSA. We demonstrated that monocytes/macrophages constitute the main subpopulation of leukocytes involved in this process. Our recent research found that MNPs-DMSA up-regulated the mRNA expression of E-, L- and P-selectin and macrophage-1 antigen, and increased concentration of tumor necrosis factor-α in lung, in a time dependent manner. The critical relevance of the β2 integrin-dependent pathway in leukocyte transmigration elicited by MNPs-DMSA was demonstrated by use of knockout mice. Our work characterizes mechanisms of the pro-inflammatory effects of MNPs-DMSA in the lung, and identifies β2 integrin-targeted interventions as promising strategies to reduce pulmonary side effects of MNPs-DMSA during biomedical applications. In addition, MNPs-DMSA could be used as modulators of lung immune response.
Collapse
Affiliation(s)
- Ricardo Bentes Azevedo
- Department of Genetic and Morphology, Institute of Biology Science, University of Brasília, Brasília-DF, Brazil.
| | | | | | | | | |
Collapse
|
14
|
Bansal S, Chhibber S. Curcumin alone and in combination with augmentin protects against pulmonaryinflammation and acute lung injury generated during Klebsiella pneumoniae B5055-induced lung infection in BALB/c mice. J Med Microbiol 2010; 59:429-437. [DOI: 10.1099/jmm.0.016873-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Acute lung injuries due to acute lung infections remain a major cause ofmortality. Thus a combination of an antibiotic and a compound with immunomodulatoryand anti-inflammatory activities can help to overcome acute lung infection-inducedinjuries. Curcumin derived from the rhizome of turmeric has been used fordecades and it exhibits anti-inflammatory, anti-carcinogenic, immunomodulatoryproperties by downregulation of various inflammatory mediators. Keeping theseproperties in mind, we investigated the anti-inflammatory properties of curcuminin a mouse model of acute inflammation by introducing Klebsiella pneumoniae B5055 into BALB/c mice via the intranasal route. Intranasal instillationof bacteria in this mouse model of acute pneumonia-induced inflammation resultedin a significant increase in neutrophil infiltration in the lungs along withincreased production of various inflammatory mediators [i.e. malondialdehyde (MDA),myeloperoxidase (MPO), nitric oxide (NO), tumour necrosisfactor (TNF)-α] in the lung tissue. The animalsthat received curcumin alone orally or in combination with augmentin, 15 daysprior to bacterial instillation into the lungs via the intranasal route, showeda significant (P <0.05) decrease in neutrophil influxinto the lungs and a significant (P <0.05) decreasein the production of MDA, NO, MPO activity and TNF-α levels.Augmentin treatment alone did not decrease the MDA, MPO, NO and TNF-α levels significantly (P >0.05) as compared tothe control group. We therefore conclude that curcumin ameliorates lung inflammationinduced by K. pneumoniae B5055 without significantly (P <0.05) decreasing the bacterial load in the lung tissue whereasaugmentin takes care of bacterial proliferation. Hence, curcumin can be usedas an adjunct therapy along with antibiotics as an anti-inflammatory or animmunomodulatory agent in the case of acute lung infection.
Collapse
Affiliation(s)
- Shruti Bansal
- Department of Microbiology, Panjab University, Chandigarh 160014, India
| | - Sanjay Chhibber
- Department of Microbiology, Panjab University, Chandigarh 160014, India
| |
Collapse
|
15
|
Ebner B, Behm P, Jacoby C, Burghoff S, French BA, Schrader J, Flögel U. Early assessment of pulmonary inflammation by 19F MRI in vivo. Circ Cardiovasc Imaging 2010; 3:202-10. [PMID: 20061515 DOI: 10.1161/circimaging.109.902312] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Emulsified perfluorocarbons (PFCs) are preferentially phagocytized by monocytes/macrophages and are readily detected by (19)F MRI. This study tests the hypothesis that (19)F MRI can be used to quantitate pulmonary inflammation by tracking of infiltrating PFC-loaded monocytes. METHODS AND RESULTS Pneumonia was induced in mice by intratracheal instillation of lipopolysaccharides (LPS) followed by intravenous injection of PFCs. Whereas regular (1)H MRI provided no evidence of lung injury 24 hours after LPS, the concurrent (19)F images clearly show PFC accumulation in both pulmonary lobes. Imaging at 48 hours after LPS revealed signals in (1)H images at the same location as the 24-hour (19)F signals. Thus, progressive pneumonia was first predicted by (19)F MRI early after PFC administration. Without LPS, at no time were (19)F signals observed within the lung. Histology and fluorescence-activated cell sorting (FACS) combined with (19)F MRI confirmed the presence of infiltrating PFC-loaded monocytes/macrophages after LPS challenge. Additional experiments with graded doses of LPS demonstrated that (19)F signal intensity strongly correlated with both LPS dose and pathological markers of lung inflammation. In separate studies, dexamethasone and CGS21680 (adenosine 2A receptor agonist) were used to demonstrate the ability of (19)F MRI to monitor anti-inflammatory therapies. CONCLUSIONS PFCs serve as a contrast agent for the prognostic and quantitative assessment of pulmonary inflammation by in vivo (19)F MRI, which is characterized by a high degree of specificity due to the lack of any (19)F background. Because PFCs are biochemically inert, this approach may also be suitable for human applications.
Collapse
Affiliation(s)
- Bernd Ebner
- Institut für Herz- und Kreislaufphysiologie, Heinrich-Heine-Universität, Düsseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
16
|
Wilson AA, Murphy GJ, Hamakawa H, Kwok LW, Srinivasan S, Hovav AH, Mulligan RC, Amar S, Suki B, Kotton DN. Amelioration of emphysema in mice through lentiviral transduction of long-lived pulmonary alveolar macrophages. J Clin Invest 2009; 120:379-89. [PMID: 20038801 DOI: 10.1172/jci36666] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2008] [Accepted: 10/28/2009] [Indexed: 11/17/2022] Open
Abstract
Directed gene transfer into specific cell lineages in vivo is an attractive approach for both modulating gene expression and correcting inherited mutations such as emphysema caused by human alpha1 antitrypsin (hAAT) deficiency. However, somatic tissues are mainly comprised of heterogeneous, differentiated cell lineages that can be short lived and difficult to specifically transfect. Here, we describe an intratracheally instilled lentiviral system able to deliver genes selectively to as many as 70% of alveolar macrophages (AMs) in the mouse lung. Following a single in vivo lentiviral transduction, genetically tagged AMs persisted in lung alveoli and expressed transferred genes for the lifetime of the adult mouse. A prolonged macrophage lifespan, rather than precursor cell proliferation, accounted for the surprisingly sustained presence of transduced AMs. We utilized this long-lived population to achieve localized secretion of therapeutic levels of hAAT protein in lung epithelial lining fluid. In an established mouse model of emphysema, lentivirally delivered hAAT ameliorated the progression of emphysema, as evidenced by attenuation of increased lung compliance and alveolar size. After 24 weeks of sustained gene expression, no humoral or cellular immune responses to hAAT protein were detected. Our results challenge the dogma that AMs are short lived and suggest that these differentiated cells may be a possible target cell population for in vivo gene therapy applications, including the sustained correction of hAAT deficiency.
Collapse
Affiliation(s)
- Andrew A Wilson
- Pulmonary Center and Department of Medicine, Boston University School of Medicine, 715 Albany Street, Boston, Massachusetts 02118, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Valois CRA, Braz JM, Nunes ES, Vinolo MAR, Lima ECD, Curi R, Kuebler WM, Azevedo RB. The effect of DMSA-functionalized magnetic nanoparticles on transendothelial migration of monocytes in the murine lung via a beta2 integrin-dependent pathway. Biomaterials 2009; 31:366-74. [PMID: 19822361 DOI: 10.1016/j.biomaterials.2009.09.053] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2009] [Accepted: 09/15/2009] [Indexed: 10/20/2022]
Abstract
Magnetic nanoparticles surface-functionalized with meso-2,3-dimercaptosuccinic acid (MNPs-DMSA) constitute an innovative and promising approach for tissue- and cell-targeted delivery of therapeutic drugs in the lung. Transendothelial migration of leukocytes in the lung is a side effect of endovenous administration of MNPs-DMSA. Using cytologic and phenotypic analysis of murine bronchoalveolar lavage cells, we identified monocytes/macrophages as the main subpopulation of leukocytes involved in this process. Moreover, ultrastructural analysis revealed the presence of nanoparticles inside of numerous macrophages from bronchoalveolar lavage. MNPs-DMSA at concentrations as high as 1 x 10(15) nanoparticles/mL had no toxic effects on macrophages, as evidenced by 3-(4, 5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) assay. Notably, MNPs-DMSA up-regulated the mRNA expression of E-, L- and P-selectin and macrophage-1 antigen in the murine lung. Upregulation of these cell adhesion molecules was associated with an increased concentration of tumor necrosis factor-alpha in lung. Finally, the critical relevance of the beta(2) integrin-dependent pathway in leukocyte transmigration elicited by MNPs-DMSA was demonstrated by use of knockout mice. Our results characterize mechanisms of the pro-inflammatory effects of MNPs-DMSA in the lung, and identify beta(2) integrin-targeted interventions as promising strategies to reduce pulmonary side effects of MNPs-DMSA during biomedical applications.
Collapse
Affiliation(s)
- Caroline R A Valois
- Department of Genetics and Morphology, Institute of Biological Sciences, University of Brasília, Brasília, DF 70910-900, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Cabanski M, Wilhelm J, Zaslona Z, Steinmüller M, Fink L, Seeger W, Lohmeyer J. Genome-wide transcriptional profiling of mononuclear phagocytes recruited to mouse lungs in response to alveolar challenge with the TLR2 agonist Pam3CSK4. Am J Physiol Lung Cell Mol Physiol 2009; 297:L608-18. [PMID: 19617307 DOI: 10.1152/ajplung.90433.2008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Compared with the Toll-like receptor 4 (TLR4) ligand LPS restricted to gram-negative bacteria, few studies have addressed induction of lung inflammation and concomitant leukocyte recruitment in response to TLR2 ligands. This study is the first report showing that selective TLR2 stimulation by its ligand Pam(3)-Cys-Ser-Lys-Lys-Lys-Lys-OH (Pam(3)CSK(4)) within the alveolar compartment promoted lung inflammation in mice and induced the migration of circulatory immune cells including mononuclear phagocytes into the inflamed alveolar space. By using the transgenic CX(3)CR1(+/GFP) mouse strain for high-purity sorting of circulating and alveolar recruited mononuclear phagocytes together with SMART preamplification and whole genome oligonucleotide microarray techniques, we found that alveolar trafficking of mononuclear phagocytes was associated with profound changes of their gene expression profiles (approximately 900 differentially regulated genes postrecruitment). In particular, alveolar recruited mononuclear phagocytes showed upregulated transcripts of genes encoding cytokines/chemokines and pattern recognition receptor (PRR)-associated molecules. Notably, we observed a dynamic change of the genetic program of recruited mononuclear phagocytes obtained from bronchoalveolar lavage fluid at different time points (24 vs. 48 h) post-Pam(3)CSK(4) challenge. In early alveolar recruited mononuclear phagocytes, mRNA levels of both proinflammatory (e.g., TNF-alpha, CCL2, and IL-6) and central anti-inflammatory/ proresolution [e.g., IL-1-receptor antagonist (IL-1RN), CD200 receptor (CD200R), IL-1 receptor-associated kinase (IRAK-M), IL-10, and Bcl-2-associated X protein (Bax)] mediators were found to be highly upregulated simultaneously. In corresponding cells recruited until later time points, transcript levels of anti-inflammatory/proresolution molecules persisted at the same level, whereas mRNA levels of proinflammatory mediators were found to decline. Collectively, our in vivo study identifies genetic programs by which alveolar recruited mononuclear phagocytes may contribute to the development and termination of pneumonia caused by gram-positive bacteria.
Collapse
Affiliation(s)
- Maciej Cabanski
- Dept. of Internal Medicine, Division of Pulmonary and Critical Care Medicine and Infectious Diseases, Hannover Medical School, Carl-Neuberg-Str.1, 30625 Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
19
|
Manicone AM, Birkland TP, Lin M, Betsuyaku T, van Rooijen N, Lohi J, Keski-Oja J, Wang Y, Skerrett SJ, Parks WC. Epilysin (MMP-28) restrains early macrophage recruitment in Pseudomonas aeruginosa pneumonia. THE JOURNAL OF IMMUNOLOGY 2009; 182:3866-76. [PMID: 19265166 DOI: 10.4049/jimmunol.0713949] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Several members of the matrix metalloproteinase (MMP) family function in various processes of innate immunity, particularly in controlling leukocyte influx. Epilysin (MMP-28) is expressed in numerous tissues and, in adult mice, it has the highest expression in lung, where it is detected in bronchial epithelial cells (Clara cells). Epilysin is also expressed by bone marrow-derived macrophages, but not by alveolar macrophages, suggesting that its expression by macrophages is dependent on localization and differentiation. To assess the role of this MMP, we generated epilysin-null (Mmp28(-/-)) mice. Although epilysin is constitutively expressed in normal tissues, Mmp28(-/-) mice have no overt phenotype. However, using a murine model of Pseudomonas aeruginosa pneumonia, we found that Mmp28(-/-) mice had an early increase in macrophage recruitment into the lungs, as well as enhanced bacterial clearance and reduced pulmonary neutrophilia, which we predicted were due to accelerated macrophage influx. Macrophage depletion in WT and Mmp28(-/-) mice confirmed a role for macrophages in clearing P. aeruginosa and regulating neutrophil recruitment. Furthermore, we observed that macrophages derived from Mmp28(-/-) mice migrated faster than did wild-type cells to bronchoalveolar lavage fluid from P. aeruginosa-treated mice of either genotype. These observations indicate that epilysin functions as an intrinsic negative regulator of macrophage recruitment by retarding the chemotaxis of these cells.
Collapse
Affiliation(s)
- Anne M Manicone
- Division of Pulmonary and Critical Care Medicine, Center for Lung Biology, University of Washington, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zasłona Z, Wilhelm J, Cakarova L, Marsh LM, Seeger W, Lohmeyer J, von Wulffen W. Transcriptome profiling of primary murine monocytes, lung macrophages and lung dendritic cells reveals a distinct expression of genes involved in cell trafficking. Respir Res 2009; 10:2. [PMID: 19149869 PMCID: PMC2639356 DOI: 10.1186/1465-9921-10-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2008] [Accepted: 01/16/2009] [Indexed: 01/09/2023] Open
Abstract
Background Peripheral blood monocytes (PBMo) originate from the bone marrow, circulate in the blood and emigrate into various organs where they differentiate into tissue resident cellular phenotypes of the mononuclear phagocyte system, including macrophages (Mϕ) and dendritic cells (DC). Like in other organs, this emigration and differentiation process is essential to replenish the mononuclear phagocyte pool in the lung under both inflammatory and non-inflammatory steady-state conditions. While many studies have addressed inflammation-driven monocyte trafficking to the lung, the emigration and pulmonary differentiation of PBMo under non-inflammatory conditions is much less understood. Methods In order to assess the transcriptional profile of circulating and lung resident mononuclear phagocyte phenotypes, PBMo, lung Mϕ and lung DC from naïve mice were flow-sorted to high purity, and their gene expression was compared by DNA microarrays on a genome-wide scale. Differential regulation of selected genes was validated by quantitative PCR and on protein level by flow cytometry. Results Differentially-expressed genes related to cell traffic were selected and grouped into the clusters (i) matrix metallopeptidases, (ii) chemokines/chemokine receptors, and (iii) integrins. Expression profiles of clustered genes were further assessed at the mRNA and protein levels in subsets of circulating PBMo (GR1- vs GR1+) and lung resident macrophages (alveolar vs interstitial Mϕ). Our data identify differentially activated genetic programs in circulating monocytes and their lung descendents. Lung DC activate an extremely diverse set of gene families but largely preserve a mobile cell profile with high expression levels of integrin and chemokine/chemokine receptors. In contrast, interstitial and even more pronounced alveolar Mϕ, stepwise downregulate gene expression of these traffic relevant communication molecules, but strongly upregulate a distinct set of matrix metallopetidases potentially involved in tissue invasion and remodeling. Conclusion Our data provide new insight in the changes of the genetic profiles of PBMo and their lung descendents, namely DC and Mϕ under non-inflammatory, steady-state conditions. These findings will help to better understand the complex relations within the mononuclear phagocyte pool of the lung.
Collapse
Affiliation(s)
- Zbigniew Zasłona
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Giessen Lung Center, Giessen, Germany.
| | | | | | | | | | | | | |
Collapse
|
21
|
Modified vaccinia virus ankara triggers chemotaxis of monocytes and early respiratory immigration of leukocytes by induction of CCL2 expression. J Virol 2009; 83:2540-52. [PMID: 19129447 DOI: 10.1128/jvi.01884-08] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Orthopoxviruses commonly enter into humans and animals via the respiratory tract. Herein, we show that immigration of leukocytes into the lung is triggered via intranasal infection of mice with modified vaccinia virus Ankara (MVA) and not with the vaccinia virus (VACV) Elstree, Wyeth, or Western Reserve (WR) strain. Immigrating cells were identified as monocytes, neutrophils, and CD4(+) lymphocytes by flow cytometry and could be detected 24 h and 48 h postinfection. Using an in vitro chemotaxis assay, we confirmed that infection with MVA induces the expression of a soluble chemotactic factor for monocytes, identified as CCL2 (monocyte chemotactic protein-1 [MCP-1]). In contrast to infection with several other VACV strains, MVA induced the expression of CCL2, CCL3, CCL4, and CXCL10 in the human monocytic cell line THP-1 as well as in primary human monocytes. Thus, MVA, and not the VACV Elstree, Wyeth, or WR strain, consistently triggered the expression of a panel of chemokines, including CCL2, in the murine lung, correlating considerably with the immigration of leukocytes. Using CCL2-deficient mice, we demonstrate that CCL2 plays a key role in MVA-triggered respiratory immigration of leukocytes. Moreover, UV irradiation of MVA prevented CCL2 expression in vitro and in vivo as well as respiratory immigration of leukocytes, demonstrating the requirement for an activated molecular viral life cycle. We propose that MVA-triggered chemokine expression causes early immigration of leukocytes to the site of infection, a feature that is important for rapid immunization and its safety and efficiency as a viral vector.
Collapse
|
22
|
Philippakis GE, Lazaris AC, Papathomas TG, Zissis C, Agrogiannis G, Thomopoulou G, Nonni A, Xiromeritis K, Nikolopoulou-Stamati P, Bramis J, Patsouris E, Perrea D, Bellenis I. Adrenaline Attenuates the Acute Lung Injury After Intratracheal Lipopolysaccharide Instillation: an Experimental Study. Inhal Toxicol 2008; 20:445-53. [DOI: 10.1080/08958370801903891] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
23
|
von Wulffen W, Steinmueller M, Herold S, Marsh LM, Bulau P, Seeger W, Welte T, Lohmeyer J, Maus UA. Lung dendritic cells elicited by Fms-like tyrosine 3-kinase ligand amplify the lung inflammatory response to lipopolysaccharide. Am J Respir Crit Care Med 2007; 176:892-901. [PMID: 17690334 DOI: 10.1164/rccm.200608-1068oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Strategically located beneath the alveolar epithelial barrier, dendritic cells (DCs) of the lung are centrally involved in the sampling and processing of inhaled antigens. However, the contribution of DCs to acute lung inflammation induced by inhaled bacterial toxins is largely unknown. OBJECTIVES To determine the effect of increased lung DC numbers elicited by Fms-like tyrosine kinase-3 ligand (Flt3L) on the acute lung inflammatory response to Escherichia coli lipopolysaccharide (LPS) and Klebsiella pneumoniae infection. METHODS Mice were pretreated with Flt3L either in the absence or presence of anti-CD11a antibodies to block the Flt3L-elicited lung DC accumulation or were made transiently neutropenic and then challenged with E. coli LPS or K. pneumoniae. MEASUREMENTS AND MAIN RESULTS Flt3L-pretreated mice challenged with LPS responded with drastically increased numbers of both lung parenchymal and alveolar DCs together with an aggravated neutrophilic alveolitis, elevated tumor necrosis factor-alpha and IL-12 levels, and a strongly increased lung permeability compared with LPS- or Flt3L-only-treated mice. Anti-CD11a-mediated blockade of lung DC accumulation significantly attenuated the lung permeability developing in response to LPS, whereas transient neutropenia did not affect lung permeability changes. Finally, Flt3L-pretreated mice responded with increased lung permeability and decreased survival upon infection with K. pneumoniae. CONCLUSIONS Lung DCs actively participate in the early inflammatory response to both inhaled bacterial toxins and live bacteria and play a yet unrecognized role in regulating lung barrier integrity.
Collapse
Affiliation(s)
- Werner von Wulffen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Giessen Lung Center, Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Beamer CA, Holian A. Antigen-presenting cell population dynamics during murine silicosis. Am J Respir Cell Mol Biol 2007; 37:729-38. [PMID: 17641296 PMCID: PMC2219550 DOI: 10.1165/rcmb.2007-0099oc] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Silicosis is an occupational lung disease resulting from the inhalation of silica particles over prolonged periods of time, which causes chronic inflammation and progressive pulmonary fibrosis. Alveolar macrophages (AM) are critical effector cells, while less is known about the role and function of pulmonary dendritic cells (DC) in silicosis. We hypothesize that a balance exists between the suppressive nature of AM and the stimulatory capacity of DC to regulate lung immunity, and that this equilibrium may be overcome by silica exposure in vivo. Our results demonstrate that in response to silica exposure, both the percent and absolute number of AM significantly decreased over time, with a concomitant significant increase in DC. Both AM and DC exhibited cellular activation in response to silica, indicated by increased expression of cell surface markers. In the absence of silica-induced AM apoptosis (TNFR 1/2-null and Gld mice), no change was observed in the percent or absolute number of either cell type. Furthermore, bone marrow-derived DC, but not bone marrow-derived macrophages, migrated from the alveoli into the lung parenchyma in response to silica, resulting in significantly increased numbers of activated T lymphocytes. Collectively, the results demonstrate that AM and DC are distinct antigen-presenting cells within the respiratory tract that respond to silica exposure in vivo in unique ways, with significant implications for immune reactivity of the lung in response to environmental pathogens.
Collapse
Affiliation(s)
- Celine A Beamer
- Center for Environmental Health Sciences, Department of Biomedical and Pharmaceutical Sciences, School of Pharmacy and Allied Health Sciences, University of Montana, Missoula, Montana 59812, USA.
| | | |
Collapse
|
25
|
Maes T, Bracke KR, Vermaelen KY, Demedts IK, Joos GF, Pauwels RA, Brusselle GG. Murine TLR4 is implicated in cigarette smoke-induced pulmonary inflammation. Int Arch Allergy Immunol 2006; 141:354-68. [PMID: 16940747 DOI: 10.1159/000095462] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2005] [Accepted: 04/20/2006] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is associated with an abnormal inflammatory response of the lungs to noxious particles or gases. We investigated whether Toll-like receptor 4 (TLR4) is implicated in cigarette smoke (CS)-induced pulmonary inflammation in a murine model of COPD. METHODS C3H/HeOuJ (Tlr4(WT)) and C3H/HeJ (Tlr4(defective)) mice were exposed to air or CS for 5 weeks (subacute) and 26 weeks (chronic), and pulmonary inflammation was evaluated. RESULTS In Tlr4(WT) mice, subacute and chronic CS exposure induced a substantial pulmonary infiltration of macrophages, neutrophils, lymphocytes and dendritic cells (DCs), that was absent in air-exposed mice. CS exposure increased the costimulatory marker expression on DCs, the levels of monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-alpha (TNF-alpha) in bronchoalveolar lavage (BAL) fluid and induced the pulmonary expression of matrix metalloproteinase-12 (MMP-12), TLR4 and TLR2. In contrast, after subacute CS exposure, Tlr4(defective) mice showed a limited (5-fold lower) increase of DCs and lymphocytes in BAL fluid, lower costimulatory marker expression on DCs and lower MCP-1 and TNF-alpha levels in BAL fluid compared to Tlr4(WT) animals. After chronic CS exposure, however, the difference in pulmonary inflammation between Tlr4(WT) and Tlr4(defective) mice was less pronounced and both strains showed similar MCP-1 and TNF-alpha levels in BAL and similar pulmonary MMP-12, TLR4 and TLR2 expression. CONCLUSIONS We demonstrated that the TLR4 mutation in C3H/HeJ mice is protective against CS-induced pulmonary influx of neutrophils, DCs and lymphocytes upon subacute CS exposure. However, TLR4 is only of minor importance in chronic CS-induced inflammation in mice.
Collapse
Affiliation(s)
- Tania Maes
- Department of Respiratory Diseases, Ghent University Hospital, Ghent, Belgium.
| | | | | | | | | | | | | |
Collapse
|
26
|
Herold S, von Wulffen W, Steinmueller M, Pleschka S, Kuziel WA, Mack M, Srivastava M, Seeger W, Maus UA, Lohmeyer J. Alveolar Epithelial Cells Direct Monocyte Transepithelial Migration upon Influenza Virus Infection: Impact of Chemokines and Adhesion Molecules. THE JOURNAL OF IMMUNOLOGY 2006; 177:1817-24. [PMID: 16849492 DOI: 10.4049/jimmunol.177.3.1817] [Citation(s) in RCA: 171] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Influenza A virus pneumonia is characterized by severe lung injury and high mortality. Early infection elicits a strong recruitment of monocytes from the peripheral blood across the endo-/epithelial barrier into the alveolar air space. However, it is currently unclear which of the infected resident lung cell populations, alveolar epithelial cells or alveolar macrophages, elicit monocyte recruitment during influenza A virus infection. In the current study, we investigated whether influenza A virus infection of primary alveolar epithelial cells and resident alveolar macrophages would elicit a basal-to-apical monocyte transepithelial migration in vitro. We found that infection of alveolar epithelial cells with the mouse-adapted influenza A virus strain PR/8 strongly induced the release of monocyte chemoattractants CCL2 and CCL5 followed by a strong monocyte transepithelial migration, and this monocytic response was strictly dependent on monocyte CCR2 but not CCR5 chemokine receptor expression. Analysis of the adhesion molecule pathways demonstrated a role of ICAM-1, VCAM-1, integrin-associated protein (CD47), and junctional adhesion molecule-c on the epithelial cell surface interacting with monocyte beta(1) and beta(2) integrins and integrin-associated protein in the monocyte transmigration process. Importantly, addition of influenza A virus-infected alveolar macrophages further enhanced monocyte transmigration across virus-infected epithelium in a TNF-alpha-dependent manner. Collectively, the data show an active role for virus-infected alveolar epithelium in the regulation of CCL2/CCR2-dependent monocyte transepithelial migration during influenza infection that is essentially dependent on both classical beta(1) and beta(2) integrins but also junctional adhesion molecule pathways.
Collapse
Affiliation(s)
- Susanne Herold
- University of Giessen Lung Center, Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine and Infectious Diseases, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Molfino NA, Jeffery PK. Chronic obstructive pulmonary disease: histopathology, inflammation and potential therapies. Pulm Pharmacol Ther 2006; 20:462-72. [PMID: 16798034 DOI: 10.1016/j.pupt.2006.04.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2005] [Revised: 02/07/2006] [Accepted: 04/18/2006] [Indexed: 12/21/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a major worldwide health burden with increasing morbidity, mortality and health care cost. It is a slowly progressive chronic inflammatory condition that affects the conducting airways (both large and small) and lung parenchyma. In COPD, inflammation is evident early on even in mild disease and increases with disease severity. Recent advances in our knowledge demonstrate, by comparison with asthma, the distinctive, "abnormal" or exaggerated inflammatory processes involved in the pathogenesis of COPD and thus identify novel therapeutic targets that could potentially impact on disease progression. The present review will focus on what is known of the abnormal inflammatory response of COPD in different regions of the conducting airways and lung. Novel, potentially promising approaches to therapy are presented.
Collapse
|
28
|
Maus UA, Janzen S, Wall G, Srivastava M, Blackwell TS, Christman JW, Seeger W, Welte T, Lohmeyer J. Resident alveolar macrophages are replaced by recruited monocytes in response to endotoxin-induced lung inflammation. Am J Respir Cell Mol Biol 2006; 35:227-35. [PMID: 16543608 DOI: 10.1165/rcmb.2005-0241oc] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In the acute respiratory distress syndrome, recruitment of peripheral blood monocytes results in expansion of the total pool of resident alveolar macrophages. The fate of resident macrophages, or whether recruited monocytes are selectively eliminated from the alveolar airspace or differentiate into resident alveolar macrophages during the resolving phase of inflammation, has not been determined. Here, we analyzed the kinetics of resident and recruited macrophage turnover within the alveolar airspace of untreated and LPS-challenged mice. Using bone marrow chimeric CD45.2 mice that were generated by lethal irradiation of CD45.2 alloantigen-expressing recipient mice and bone marrow transplantation from CD45.1 alloantigen-expressing donor mice, we employed a flow cytometric approach to distinguish recipient from donor-type macrophages in bronchoalveolar lavage fluids. Our data show that resident alveolar macrophages of untreated chimeric CD45.2 mice are very slowly replaced by constitutively immigrating CD45.1 positive monocytes, resulting in a replacement rate of approximately 40% by 1 yr. In contrast, more than 85% of the resident CD45.2 positive alveolar and lung homogenate macrophages were exchanged by donor CD45.1-expressing macrophages within 2 mo after treatment with Escherichia coli endotoxin (LPS). Importantly, fluorescence-activated cell sorter analysis of increased annexin V binding to both recipient and donor-type macrophages revealed increased apoptotic events to underlie this endotoxin-driven inflammatory macrophage turnover. Collectively, the data show that under baseline conditions the alveolar macrophage turnover exhibits very slow kinetics, whereas acute lung inflammation in response to treatment with LPS triggers a brisk acceleration of recruitment of monocytes that replace the resident alveolar macrophage population.
Collapse
Affiliation(s)
- Ulrich A Maus
- Laboratory for Experimental Lung Research, Hannover School of Medicine, Feodor-Lynen-Strasse 21, Hannover 30625, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Srivastava M, Jung S, Wilhelm J, Fink L, Bühling F, Welte T, Bohle RM, Seeger W, Lohmeyer J, Maus UA. The Inflammatory versus Constitutive Trafficking of Mononuclear Phagocytes into the Alveolar Space of Mice Is Associated with Drastic Changes in Their Gene Expression Profiles. THE JOURNAL OF IMMUNOLOGY 2005; 175:1884-93. [PMID: 16034132 DOI: 10.4049/jimmunol.175.3.1884] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mononuclear phagocytes enter the lungs both constitutively to maintain alveolar macrophage and dendritic cell homeostasis, as well as during lung inflammation, where the role of these cells is less well defined. We used a transgenic mouse strain (CX3CR1(+/GFP)) that harbors a GFP label in circulating monocytes to identify and sort these cells from the vascular and alveolar compartments under both constitutive and acute lung inflammatory conditions. Using nylon arrays combined with real-time RT-PCR for gene expression profiling, we found that flow-sorted, highly purified mononuclear phagocytes recruited to acutely inflamed mouse lungs showed strongly up-regulated mRNA levels of the neutrophil chemoattractants KC, MIP-2, and IP-10, which contrasted with alveolar mononuclear phagocytes that immigrated in steady state. Similar observations were made for the lysosomal cathepsins B, L, and K being strongly up-regulated in mononuclear phagocytes upon recruitment to inflamed lungs but not during constitutive alveolar immigration. Inflammatory elicited mononuclear phagocytes also demonstrated significantly increased mRNA levels of the cytokine TNF-alpha and the PRR-associated molecules CD14, TLR4, and syndecan-4. Together, inflammatory elicited mononuclear phagocytes exhibit strongly increased neutrophil chemoattractants, lysosomal proteases, and LPS signaling mRNA transcripts, suggesting that these cells may play a major role in acute lung inflammatory processes.
Collapse
Affiliation(s)
- Mrigank Srivastava
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Justus-Liebig-University, Giessen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Maus UA, Wellmann S, Hampl C, Kuziel WA, Srivastava M, Mack M, Everhart MB, Blackwell TS, Christman JW, Schlöndorff D, Bohle RM, Seeger W, Lohmeyer J. CCR2-positive monocytes recruited to inflamed lungs downregulate local CCL2 chemokine levels. Am J Physiol Lung Cell Mol Physiol 2005; 288:L350-8. [PMID: 15516494 DOI: 10.1152/ajplung.00061.2004] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The CC chemokine ligand-2 (CCL2) and its receptor CCR2 are essential for monocyte trafficking under inflammatory conditions. However, the mechanisms that determine the intensity and duration of alveolar monocyte accumulation in response to CCL2 gradients in inflamed lungs have not been resolved. To determine the potential role of CCR2-expressing monocytes in regulating alveolar CCL2 levels, we compared leukocyte recruitment kinetics and alveolar CCL2 levels in wild-type and CCR2-deficient mice in response to intratracheal LPS challenge. In wild-type mice, LPS elicited a dose- and time-dependent alveolar monocyte accumulation accompanied by low CCL2 levels in bronchoalveolar lavage fluid (BALF). In contrast, LPS-treated CCR2-deficient mice lacked alveolar monocyte accumulation, which was accompanied by relatively high CCL2 levels in BALF. Similarly, wild-type mice that were treated systemically with the blocking anti-CCR2 antibody MC21 completely lacked LPS-induced alveolar monocyte trafficking that was associated with high CCL2 levels in BALF. Intratracheal application of anti-CCR2 antibody MC21 to locally block CCR2 on both resident and recruited cells did not affect LPS-induced alveolar monocyte trafficking but led to significantly increased BALF CCL2 levels. Reciprocally bone marrow-transplanted, LPS-treated wild-type and CCR2-deficient mice showed a strict inverse relationship between alveolar monocyte recruitment and BALF CCL2 levels. In addition, freshly isolated human and mouse monocytes were capable of integrating CCL2 in vitro. LPS-induced alveolar monocyte accumulation is accompanied by monocytic CCR2-dependent consumption of CCL2 levels in the lung. This feedback loop may limit the intensity of monocyte recruitment to inflamed lungs and play a role in the maintenance of homeostasis.
Collapse
Affiliation(s)
- Ulrich A Maus
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Justus-Liebig-University, Klinikstr. 36, Giessen 35392, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Spight D, Zhao B, Haas M, Wert S, Denenberg A, Shanley TP. Immunoregulatory effects of regulated, lung-targeted expression of IL-10 in vivo. Am J Physiol Lung Cell Mol Physiol 2005; 288:L251-65. [PMID: 15466252 DOI: 10.1152/ajplung.00122.2004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Regulation of pulmonary inflammation involves an intricate balance of both pro- and anti-inflammatory mediators. Acute lung injury can result from direct pulmonary insults that activate alveolar macrophages to respond with increased cytokine expression. Such cytokine gene expression is mediated in part via NF-κB. IL-10 has been previously identified as an important endogenous anti-inflammatory cytokine in vivo on the basis of inhibiting NF-κB activation; however, the mechanism of this inhibition remains incompletely defined. We hypothesized that IL-10 regulated NF-κB activation in vivo via IκK inhibition. A bitransgenic mouse that allowed for externally regulated, lung-specific human IL-10 overexpression was generated. In the bitransgenic mice, introduction of doxycycline induced lung-specific, human IL-10 overexpression. Acute induction of IL-10 resulted in significant decreases in bronchoalveolar lavage fluid neutrophils (48%, P = 0.03) and TNF (62%, P < 0.01) following intratracheal LPS compared with bitransgenic negative mice. In vitro kinase assays showed this decrease to correlate to diminished lung IκK activity. Furthermore, we also examined the effect of chronic IL-10 overexpression in these transgenic mice. Results show that IL-10 overexpression in lungs of mature mice increased the number of intrapulmonary cells the phenotype of which was skewed toward increased B220+/CD45+ B cells and CD4+ T cells and was associated with increased CC chemokine expression. Thus regulated, lung-specific IL-10 overexpression may have a variety of complex immunologic effects depending on the timing and duration of expression.
Collapse
Affiliation(s)
- Donn Spight
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH 45229-3039, USA
| | | | | | | | | | | |
Collapse
|
32
|
Hoffmann HJ, Iversen M, Takai H, Sigsgaard T, Omland Ø, Dahl R. Exposure to work-related levels of swine dust up-regulates CD106 on human alveolar macrophages. Am J Ind Med 2004; 46:378-80. [PMID: 15376213 DOI: 10.1002/ajim.20078] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Exposure to dust from swine confinement buildings, causes an inflammatory response. Monocyte recruitment to the murine lung after instillation of lipopolysaccharide (LPS) has been shown to partially depend on CD106/VCAM1. We wanted to determine whether this can be confirmed in man. METHODS Non-naïve persons bearing personal air samples were exposed for 3 hr in a swine confinement building, and were bronchoscopied before and after exposure. Blood samples were taken at the time of bronchoscopy. Expression of adhesion molecules on leukocytes was investigated by flow cytometry in bronchoaveloar lavage (BAL) and blood. RESULTS Expression of CD106 on alveolar macrophages, but not on neutrophils or T cells in BAL or blood, was up-regulated in proportion to the amount of LPS that individuals were exposed. CONCLUSIONS This argues for requirement of CD106 during inflammatory recruitment of monocytes to the human lung.
Collapse
|
33
|
Maus UA, Srivastava M, Paton JC, Mack M, Everhart MB, Blackwell TS, Christman JW, Schlöndorff D, Seeger W, Lohmeyer J. Pneumolysin-Induced Lung Injury Is Independent of Leukocyte Trafficking into the Alveolar Space. THE JOURNAL OF IMMUNOLOGY 2004; 173:1307-12. [PMID: 15240724 DOI: 10.4049/jimmunol.173.2.1307] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pneumolysin (PLY) is a major virulence factor released by Streptococcus pneumoniae and has been implicated in the pathogenesis of pneumococcal pneumonia. In this study, we evaluated the contribution of newly recruited neutrophils and monocytes and resident alveolar macrophages to the pathogenesis of PLY-induced lung injury. Mice received either adhesion-blocking Abs to inhibit alveolar leukocyte trafficking or liposomal clodronate to deplete alveolar macrophages before intratracheal application of native PLY or its noncytotoxic derivative PdB. We found that treatment with PLY but not PdB resulted in increased lung vascular permeability. In addition, PLY also induced a decrease in the resident alveolar macrophage population, and the recruitment of peripheral blood neutrophils and monocytes into the alveolar space. Blockade of PLY-induced alveolar leukocyte trafficking by pretreatment of mice with anti-CD18 plus anti-CD49d Abs or depletion of circulating neutrophils did not attenuate the increase in lung permeability observed in response to intratracheal PLY. In addition, depletion of resident alveolar macrophages with clodronated liposomes did not reduce alveolar injury developing in response to PLY. PLY-induced lung injury was associated with only a small increase in bronchoalveolar lavage concentrations of cytokines. These data indicate that PLY-induced lung injury results from direct pneumotoxic effects on the alveolar-capillary barrier and is independent of both resident and recruited phagocytic cells.
Collapse
Affiliation(s)
- Ulrich A Maus
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, Justus-Liebig University, Giessen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Circulating monocytes from healthy individuals and COPD patients. Respir Res 2003; 4:11. [PMID: 14624669 PMCID: PMC239032 DOI: 10.1186/1465-9921-4-11] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2003] [Accepted: 09/22/2003] [Indexed: 11/22/2022] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is characterized by incompletely reversible airflow obstruction associated with inflammation in which monocytes/macrophages are the predominant inflammatory cells. The only known genetic factor related to COPD is inherited PiZZ deficiency of α1-antitrypsin (AAT), an inhibitor of serine proteases. Methods We investigated the basal and LPS-stimulated release of pro-inflammatory molecules from blood monocytes isolated from age and gender matched healthy (n = 30) and COPD (n = 20) individuals with and without AAT deficiency. Results After 18 h of cell culture the basal release of MMP-9 was 2.5-fold, p < 0.02 greater, whereas IL-8 was 1.8-fold (p < 0.01) lower from COPD patient monocytes than from controls. LPS-stimulated release of IL-6 and MCP-1 was greater from COPD patient's monocytes relative to controls, while activation of control cells resulted in enhanced secretion of ICAM-1 and MMP-9 compared to COPD patients. Independent of disease status, monocytes from PiZZ AAT carriers released less TNFα (by 2.3-fold, p < 0.03). Conclusions The basal and LPS-stimulated secretion of specific pro-inflammatory molecules from circulating monocytes differs between healthy and COPD subjects. These findings may be valuable for further studies on the mechanisms involved in recruitment and activation of inflammatory cells in COPD.
Collapse
|
35
|
Maus UA, Waelsch K, Kuziel WA, Delbeck T, Mack M, Blackwell TS, Christman JW, Schlöndorff D, Seeger W, Lohmeyer J. Monocytes are potent facilitators of alveolar neutrophil emigration during lung inflammation: role of the CCL2-CCR2 axis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3273-8. [PMID: 12626586 DOI: 10.4049/jimmunol.170.6.3273] [Citation(s) in RCA: 161] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Coordinated neutrophil and monocyte recruitment is a characteristic feature of acute lung inflammatory responses. We investigated the role of monocyte chemotactic protein-1 (CCL2, JE) and the chemokine receptor CCR2 in regulating alveolar leukocyte traffic. Groups of wild-type (WT) mice, CCR2-deficient mice, lethally irradiated CCR2-deficient and WT mice that were reciprocally bone marrow transplanted (chimeric CCR2 deficient and WT, respectively), chimeric CCR2-deficient mice with an enriched CCR2(+) alveolar macrophage population, and CCR2-deficient mice transfused with CCR2(+) mononuclear cells were treated with intratracheal CCL2 and/or Escherichia coli endotoxin. Our data show that alveolar monocyte recruitment is strictly dependent on CCR2. LPS-induced neutrophil migration to the lungs is CCR2 independent. However, when CCR2-bearing blood monocytes are present, alveolar neutrophil accumulation is accelerated and drastically amplified. We suggest that this hitherto unrecognized cooperativity between monocytes and neutrophils contributes to the strong, coordinated leukocyte efflux in lung inflammation.
Collapse
Affiliation(s)
- Ulrich A Maus
- Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine and Infectious Diseases, Justus-Liebig-University, Giessen, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Tobin MJ. Tuberculosis, lung infections, interstitial lung disease, and journalology in AJRCCM 2002. Am J Respir Crit Care Med 2003; 167:345-55. [PMID: 12554623 DOI: 10.1164/rccm.2212002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Martin J Tobin
- Division of Pulmonary and Critical Care Medicine, Loyola University of Chicago Stritch School of Medicine and Hines Veterans Affairs Hospital, Hines, Illinois 60141, USA.
| |
Collapse
|
37
|
Maus U, von Grote K, Kuziel WA, Mack M, Miller EJ, Cihak J, Stangassinger M, Maus R, Schlöndorff D, Seeger W, Lohmeyer J. The role of CC chemokine receptor 2 in alveolar monocyte and neutrophil immigration in intact mice. Am J Respir Crit Care Med 2002; 166:268-73. [PMID: 12153956 DOI: 10.1164/rccm.2112012] [Citation(s) in RCA: 160] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The CC chemokine ligand 2 (CCL2) (JE, monocyte chemotactic protein-1 [MCP-1]) and its CC chemokine receptor 2 (CCR2) are critical regulators of monocyte/macrophage trafficking. Recently, we demonstrated that application of exogenous CCL2 in the lungs of mice induced monocyte accumulation in the airspace, whereas combined bronchoalveolar instillation of CCL2 and Escherichia coli endotoxin provoked both enhanced monocyte accumulation and extensive neutrophil influx associated with loss of pulmonary endothelial/epithelial barrier function. In this study, we investigated the role of the CCL2 receptor CCR2 in alveolar leukocyte traffic. In CCR2 knockout mice or wild-type mice treated with the anti-CCR2-blocking monoclonal antibody MC21, monocyte accumulation in response to alveolar CCL2 or CCL2 plus endotoxin was inhibited by more than 90%. Unexpectedly, alveolar neutrophil accumulation in the CCL2/lipopolysaccharide (LPS) model was also drastically reduced by both approaches of CCR2 function interference. When wild-type mice treated with anti-Gr-1 monoclonal antibody to deplete neutrophils selectively or treated with antileukinate, a CXC receptor inhibitor, were challenged with alveolar CCL2 plus LPS, alveolar monocyte accumulation was markedly decreased. Wild-type mice treated with MC21 to block CCR2 function or with anti-Gr-1 to deplete neutrophils did not exhibit the vascular leakage that typically accompanies inflammation triggered by CCL2 and LPS in wild-type mice. These findings confirm a central role for CCR2 in the process of alveolar monocyte recruitment in response to CCL2 alone and combined CCL2 plus LPS and reveal a previously unobserved interdependence between monocyte and neutrophil trafficking that has important implications for the concomitant increase in vascular permeability.
Collapse
Affiliation(s)
- Ulrich Maus
- Department of Internal Medicine, Justus-Liebig-University, Klinikstrasse 36, Giessen 35392, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Maus UA, Koay MA, Delbeck T, Mack M, Ermert M, Ermert L, Blackwell TS, Christman JW, Schlöndorff D, Seeger W, Lohmeyer J. Role of resident alveolar macrophages in leukocyte traffic into the alveolar air space of intact mice. Am J Physiol Lung Cell Mol Physiol 2002; 282:L1245-52. [PMID: 12003780 DOI: 10.1152/ajplung.00453.2001] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intratracheal instillation of the monocyte chemoattractant JE/monocyte chemoattractant protein (MCP)-1 in mice was recently shown to cause increased alveolar monocyte accumulation in the absence of lung inflammation, whereas combined JE/MCP-1/lipopolysaccharide (LPS) challenge provoked acute lung inflammation with early alveolar neutrophil and delayed alveolar monocyte influx. We evaluated the role of resident alveolar macrophages (rAM) in these leukocyte recruitment events and related phenomena of lung inflammation. Depletion of rAM by pretreatment of mice with liposomal clodronate did not affect the JE/MCP-1-driven alveolar monocyte accumulation, despite the observation that rAM constitutively expressed the JE/MCP-1 receptor CCR2, as analyzed by flow cytometry and immunohistochemistry. In contrast, depletion of rAM largely suppressed alveolar cytokine release as well as neutrophil and monocyte recruitment profiles upon combined JE/MCP-1/LPS treatment. Despite this strongly attenuated alveolar inflammatory response, increased lung permeability was still observed in rAM-depleted mice undergoing JE/MCP-1/LPS challenge. Lung leakage was abrogated by codepletion of circulating neutrophils or administration of anti-CD18. Collectively, rAM are not involved in JE/MCP-1-driven alveolar monocyte recruitment in noninflamed lungs but largely contribute to the alveolar cytokine response and enhanced early neutrophil and delayed monocyte influx under inflammatory conditions (JE/MCP-1/LPS deposition). Loss of lung barrier function observed under these conditions is rAM independent but involves circulating neutrophils via beta(2)-integrin engagement.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Bronchoalveolar Lavage Fluid/cytology
- Bronchoalveolar Lavage Fluid/immunology
- CD18 Antigens/drug effects
- Cell Count
- Chemokine CCL2/administration & dosage
- Chemotaxis, Leukocyte/drug effects
- Chemotaxis, Leukocyte/immunology
- Chemotaxis, Leukocyte/physiology
- Cytokines/metabolism
- Escherichia coli/immunology
- Female
- Flow Cytometry
- Immunohistochemistry
- Instillation, Drug
- Lipopolysaccharides/administration & dosage
- Macrophages, Alveolar/cytology
- Macrophages, Alveolar/physiology
- Mice
- Mice, Inbred BALB C
- Monocytes/cytology
- Monocytes/drug effects
- Monocytes/immunology
- Monocytes/metabolism
- Pneumonia/chemically induced
- Pneumonia/immunology
- Pneumonia/pathology
- Pulmonary Alveoli/cytology
- Pulmonary Alveoli/drug effects
- Pulmonary Alveoli/metabolism
- Receptors, CCR2
- Receptors, Chemokine/biosynthesis
- Trachea/drug effects
- Trachea/physiology
Collapse
Affiliation(s)
- Ulrich A Maus
- Department of Internal Medicine, Division of Pneumology, Justus Liebig University, 35392 Giessen, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|