Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 129(6): 2293-2304, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30830879

ABSTRACT

Non-apoptotic forms of cell death can trigger sterile inflammation through the release of danger-associated molecular patterns, which are recognized by innate immune receptors. However, despite years of investigation the mechanisms which initiate inflammatory responses after heart transplantation remain elusive. Here, we demonstrate that ferrostatin-1 (Fer-1), a specific inhibitor of ferroptosis, decreases the level of pro-ferroptotic hydroperoxy-arachidonoyl-phosphatidylethanolamine, reduces cardiomyocyte cell death and blocks neutrophil recruitment following heart transplantation. Inhibition of necroptosis had no effect on neutrophil trafficking in cardiac grafts. We extend these observations to a model of coronary artery ligation-induced myocardial ischemia reperfusion injury where inhibition of ferroptosis resulted in reduced infarct size, improved left ventricular systolic function, and reduced left ventricular remodeling. Using intravital imaging of cardiac transplants, we uncover that ferroptosis orchestrates neutrophil recruitment to injured myocardium by promoting adhesion of neutrophils to coronary vascular endothelial cells through a TLR4/TRIF/type I IFN signaling pathway. Thus, we have discovered that inflammatory responses after cardiac transplantation are initiated through ferroptotic cell death and TLR4/Trif-dependent signaling in graft endothelial cells. These findings provide a platform for the development of therapeutic strategies for heart transplant recipients and patients, who are vulnerable to ischemia reperfusion injury following restoration of coronary blood flow.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Ferroptosis/immunology , Heart Transplantation , Myocardial Reperfusion Injury/immunology , Myocardium/immunology , Neutrophil Infiltration , Neutrophils/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Cyclohexylamines/pharmacology , Ferroptosis/drug effects , Ferroptosis/genetics , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Knockout , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Neutrophils/pathology , Phenylenediamines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 4/genetics , Ventricular Function, Left/drug effects , Ventricular Function, Left/genetics , Ventricular Function, Left/immunology
2.
Am J Transplant ; 19(4): 1011-1023, 2019 04.
Article in English | MEDLINE | ID: mdl-30378766

ABSTRACT

Neutrophil extracellular traps (NETs) have been shown to worsen acute pulmonary injury including after lung transplantation. The breakdown of NETs by DNAse-1 can help restore lung function, but whether there is an impact on allograft tolerance remains less clear. Using intravital 2-photon microscopy, we analyzed the effects of DNAse-1 on NETs in mouse orthotopic lung allografts damaged by ischemia-reperfusion injury. Although DNAse-1 treatment rapidly degrades intragraft NETs, the consequential release of NET fragments induces prolonged interactions between infiltrating CD4+ T cells and donor-derived antigen presenting cells. DNAse-1 generated NET fragments also promote human alveolar macrophage inflammatory cytokine production and prime dendritic cells for alloantigen-specific CD4+ T cell proliferation through activating toll-like receptor (TLR) - Myeloid Differentiation Primary Response 88 (MyD88) signaling pathways. Furthermore, and in contrast to allograft recipients with a deficiency in NET generation due to a neutrophil-specific ablation of Protein Arginine Deiminase 4 (PAD4), DNAse-1 administration to wild-type recipients promotes the recognition of allo- and self-antigens and prevents immunosuppression-mediated lung allograft acceptance through a MyD88-dependent pathway. Taken together, these data show that the rapid catalytic release of NET fragments promotes innate immune responses that prevent lung transplant tolerance.


Subject(s)
Extracellular Traps/immunology , Immunity, Innate/immunology , Lung Transplantation , Transplantation Tolerance , Animals , Cells, Cultured , Dendritic Cells/immunology , Deoxyribonucleases/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Traps/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages, Alveolar/cytology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Mice , Mice, Inbred BALB C , Reperfusion Injury
3.
Am J Transplant ; 19(5): 1464-1477, 2019 05.
Article in English | MEDLINE | ID: mdl-30582269

ABSTRACT

Primary graft dysfunction (PGD) is a major limitation in short- and long-term lung transplant survival. Recent work has shown that mitochondrial damage-associated molecular patterns (mtDAMPs) can promote solid organ injury, but whether they contribute to PGD severity remains unclear. We quantitated circulating plasma mitochondrial DNA (mtDNA) in 62 patients, before lung transplantation and shortly after arrival to the intensive care unit. Although all recipients released mtDNA, high levels were associated with severe PGD development. In a mouse orthotopic lung transplant model of PGD, we detected airway cell-free damaged mitochondria and mtDNA in the peripheral circulation. Pharmacologic inhibition or genetic deletion of formylated peptide receptor 1 (FPR1), a chemotaxis sensor for N-formylated peptides released by damaged mitochondria, inhibited graft injury. An analysis of intragraft neutrophil-trafficking patterns reveals that FPR1 enhances neutrophil transepithelial migration and retention within airways but does not control extravasation. Using donor lungs that express a mitochondria-targeted reporter protein, we also show that FPR1-mediated neutrophil trafficking is coupled with the engulfment of damaged mitochondria, which in turn triggers reactive oxygen species (ROS)-induced pulmonary edema. Therefore, our data demonstrate an association between mtDAMP release and PGD development and suggest that neutrophil trafficking and effector responses to damaged mitochondria are drivers of graft damage.


Subject(s)
Alarmins/metabolism , Lung Diseases/immunology , Lung Diseases/surgery , Lung Transplantation/adverse effects , Mitochondria/metabolism , Primary Graft Dysfunction , Aged , Animals , Cell Separation , DNA, Mitochondrial/blood , Female , Flow Cytometry , Graft Survival , Humans , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Middle Aged , Neutrophils/metabolism , Pulmonary Edema/complications , Pulmonary Edema/immunology , Reactive Oxygen Species/metabolism , Receptors, Formyl Peptide/metabolism , Reperfusion Injury , Retrospective Studies , Tissue Donors
4.
J Clin Invest ; 129(2): 556-568, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30561386

ABSTRACT

Antibody-mediated rejection (AMR) is a principal cause of acute and chronic failure of lung allografts. However, mechanisms mediating this oftentimes fatal complication are poorly understood. Here, we show that Foxp3+ T cells formed aggregates in rejection-free human lung grafts and accumulated within induced bronchus-associated lymphoid tissue (BALT) of tolerant mouse lungs. Using a retransplantation model, we show that selective depletion of graft-resident Foxp3+ T lymphocytes resulted in the generation of donor-specific antibodies (DSA) and AMR, which was associated with complement deposition and destruction of airway epithelium. AMR was dependent on graft infiltration by B and T cells. Depletion of graft-resident Foxp3+ T lymphocytes resulted in prolonged interactions between B and CD4+ T cells within transplanted lungs, which was dependent on CXCR5-CXCL13. Blockade of CXCL13 as well as inhibition of the CD40 ligand and the ICOS ligand suppressed DSA production and prevented AMR. Thus, we have shown that regulatory Foxp3+ T cells residing within BALT of tolerant pulmonary allografts function to suppress B cell activation, a finding that challenges the prevailing view that regulation of humoral responses occurs peripherally. As pulmonary AMR is largely refractory to current immunosuppression, our findings provide a platform for developing therapies that target local immune responses.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes , Bronchi , Graft Rejection , Lung Transplantation , Lymphocyte Activation , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Bronchi/immunology , Bronchi/pathology , CD40 Ligand/genetics , CD40 Ligand/immunology , Chemokine CXCL13/genetics , Chemokine CXCL13/immunology , Graft Rejection/genetics , Graft Rejection/immunology , Graft Rejection/pathology , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
5.
J Clin Invest ; 128(7): 2833-2847, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29781811

ABSTRACT

Ischemia-reperfusion injury, a form of sterile inflammation, is the leading risk factor for both short-term mortality following pulmonary transplantation and chronic lung allograft dysfunction. While it is well recognized that neutrophils are critical mediators of acute lung injury, processes that guide their entry into pulmonary tissue are not well understood. Here, we found that CCR2+ classical monocytes are necessary and sufficient for mediating extravasation of neutrophils into pulmonary tissue during ischemia-reperfusion injury following hilar clamping or lung transplantation. The classical monocytes were mobilized from the host spleen, and splenectomy attenuated the recruitment of classical monocytes as well as the entry of neutrophils into injured lung tissue, which was associated with improved graft function. Neutrophil extravasation was mediated by MyD88-dependent IL-1ß production by graft-infiltrating classical monocytes, which downregulated the expression of the tight junction-associated protein ZO-2 in pulmonary vascular endothelial cells. Thus, we have uncovered a crucial role for classical monocytes, mobilized from the spleen, in mediating neutrophil extravasation, with potential implications for targeting of recipient classical monocytes to ameliorate pulmonary ischemia-reperfusion injury in the clinic.


Subject(s)
Interleukin-1beta/immunology , Lung Injury/immunology , Monocytes/immunology , Reperfusion Injury/immunology , Animals , Cell Movement/immunology , Humans , Lung Injury/etiology , Lung Injury/pathology , Lung Transplantation/adverse effects , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Models, Immunological , Monocytes/pathology , Myeloid Differentiation Factor 88/immunology , Neutrophils/immunology , Neutrophils/pathology , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Spleen/immunology , Spleen/pathology , Zonula Occludens-2 Protein/immunology
6.
Arterioscler Thromb Vasc Biol ; 38(5): 1030-1036, 2018 05.
Article in English | MEDLINE | ID: mdl-29567678

ABSTRACT

OBJECTIVE: Aortic arch transplants have advanced our understanding of processes that contribute to progression and regression of atherosclerotic plaques. To characterize the dynamic behavior of monocytes and macrophages in atherosclerotic plaques over time, we developed a new model of cervical aortic arch transplantation in mice that is amenable to intravital imaging. APPROACH AND RESULTS: Vascularized aortic arch grafts were transplanted heterotropically to the right carotid arteries of recipient mice using microsurgical suture techniques. To image immune cells in atherosclerotic lesions during regression, plaque-bearing aortic arch grafts from B6 ApoE-deficient donors were transplanted into syngeneic CX3CR1 GFP reporter mice. Grafts were evaluated histologically, and monocytic cells in atherosclerotic plaques in ApoE-deficient grafts were imaged intravitally by 2-photon microscopy in serial fashion. In complementary experiments, CCR2+ cells in plaques were serially imaged by positron emission tomography using specific molecular probes. Plaques in ApoE-deficient grafts underwent regression after transplantation into normolipidemic hosts. Intravital imaging revealed clusters of largely immotile CX3CR1+ monocytes/macrophages in regressing plaques that had been recruited from the periphery. We observed a progressive decrease in CX3CR1+ monocytic cells in regressing plaques and a decrease in CCR2+ positron emission tomography signal during 4 months. CONCLUSIONS: Cervical transplantation of atherosclerotic mouse aortic arches represents a novel experimental tool to investigate cellular mechanisms that contribute to the remodeling of atherosclerotic plaques.


Subject(s)
Aorta, Thoracic/diagnostic imaging , Aorta, Thoracic/pathology , Aortic Diseases/diagnostic imaging , Aortic Diseases/pathology , Atherosclerosis/diagnostic imaging , Atherosclerosis/pathology , Intravital Microscopy/methods , Microscopy, Fluorescence, Multiphoton , Monocytes/pathology , Plaque, Atherosclerotic , Positron Emission Tomography Computed Tomography , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/transplantation , Aortic Diseases/genetics , Aortic Diseases/metabolism , Atherosclerosis/genetics , Atherosclerosis/metabolism , CX3C Chemokine Receptor 1/genetics , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout, ApoE , Monocytes/metabolism , Receptors, CCR2/metabolism , Time Factors , Red Fluorescent Protein
7.
Ann Am Thorac Soc ; 14(Supplement_3): S216-S219, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28945475

ABSTRACT

Outcomes after lung transplant lag behind those of other solid-organ transplants. A better understanding of the pathways that contribute to rejection and tolerance after lung transplant will be required to develop new therapeutic strategies that take into account the unique immunological features of lungs. Mechanistic immunological investigations in an orthotopic transplant model in the mouse have shed new light on immune responses after lung transplant. Here, we highlight that interactions between immune cells within pulmonary grafts shape their fate. These observations set lungs apart from other organs and help provide the conceptual framework for the development of lung-specific immunosuppression.


Subject(s)
Graft Rejection , Lung Transplantation , Lung/immunology , Humans , Immune Tolerance , Lymphoid Tissue/immunology , T-Lymphocytes, Regulatory/immunology
8.
Curr Opin Organ Transplant ; 22(1): 29-35, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27861263

ABSTRACT

PURPOSE OF REVIEW: To date, outcomes after lung transplantation are far worse than after transplantation of other solid organs. New insights into mechanisms that contribute to graft rejection and tolerance after lung transplantation remain of great interest. This review examines the recent literature on the role of innate and adaptive immunity in shaping the fate of lung grafts. RECENT FINDINGS: Innate and adaptive immune cells orchestrate allograft rejection after transplantation. Innate immune cells such as neutrophils are recruited to the lung graft early after reperfusion and subsequently promote allograft rejection. Although it is widely recognized that CD4 T lymphocytes in concert with CD8 T cells promote graft rejection, regulatory Foxp3 CD4 T, central memory CD8 T cells, and natural killer cells can facilitate tolerance. SUMMARY: This review highlights interactions between innate and adaptive immune pathways and how they contribute to lung allograft rejection. These findings lay a foundation for the design of new therapeutic strategies that target both innate and adaptive immune responses.


Subject(s)
Graft Rejection/immunology , Lung Transplantation/methods , Reperfusion Injury/therapy , Humans
9.
JCI Insight ; 1(12)2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27536731

ABSTRACT

It is well established that maladaptive innate immune responses to sterile tissue injury represent a fundamental mechanism of disease pathogenesis. In the context of cardiac ischemia reperfusion injury, neutrophils enter inflamed heart tissue, where they play an important role in potentiating tissue damage and contributing to contractile dysfunction. The precise mechanisms that govern how neutrophils are recruited to and enter the injured heart are incompletely understood. Using a model of cardiac transplant-mediated ischemia reperfusion injury and intravital 2-photon imaging of beating mouse hearts, we determined that tissue-resident CCR2+ monocyte-derived macrophages are essential mediators of neutrophil recruitment into ischemic myocardial tissue. Our studies revealed that neutrophil extravasation is mediated by a TLR9/MyD88/CXCL5 pathway. Intravital 2-photon imaging demonstrated that CXCL2 and CXCL5 play critical and nonredundant roles in guiding neutrophil adhesion and crawling, respectively. Together, these findings uncover a specific role for a tissue-resident monocyte-derived macrophage subset in sterile tissue inflammation and support the evolving concept that macrophage ontogeny is an important determinant of function. Furthermore, our results provide the framework for targeting of cell-specific signaling pathways in myocardial ischemia reperfusion injury.

10.
Am J Pathol ; 185(12): 3189-201, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26468975

ABSTRACT

Chronic obstructive pulmonary disease is characterized, in part, by chronic inflammation that persists even after smoking cessation, suggesting that a failure to resolve inflammation plays an important role in the pathogenesis of the disease. It is widely recognized that the resolution of inflammation is an active process, governed by specialized proresolving lipid mediators, including lipoxins, resolvins, maresins, and protectins. Here, we report that proresolving signaling and metabolic pathways are disrupted in lung tissue from patients with chronic obstructive pulmonary disease, suggesting that supplementation with proresolving lipid mediators might reduce the development of emphysema by controlling chronic inflammation. Groups of mice were exposed long-term to cigarette smoke and treated with the proresolving mediator resolvin D1. Resolvin D1 was associated with a reduced development of cigarette smoke-induced emphysema and airspace enlargement, with concurrent reductions in inflammation, oxidative stress, and cell death. Interestingly, resolvin D1 did not promote the differentiation of M2 macrophages and did not promote tissue fibrosis. Taken together, our results suggest that cigarette smoking disrupts endogenous proresolving pathways and that supplementation with specialized proresolving lipid mediators is an important therapeutic strategy in chronic lung disease, especially if endogenous specialized proresolving lipid mediator signaling is impaired.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Docosahexaenoic Acids/therapeutic use , Pneumonia/prevention & control , Pulmonary Emphysema/prevention & control , Adult , Aged , Aged, 80 and over , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Chronic Disease , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/pharmacology , Drug Evaluation, Preclinical/methods , Female , Humans , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/pathology , Male , Mice, Inbred C57BL , Middle Aged , Oxidative Stress/drug effects , Pneumonia/metabolism , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Emphysema/metabolism , Pulmonary Emphysema/pathology , Signal Transduction/physiology , Smoking/adverse effects
11.
J Immunol ; 193(10): 4980-7, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25320283

ABSTRACT

The respiratory epithelium consists of lung sentinel cells, which are the first to contact inhaled inflammatory insults, including air pollutants, smoke, and microorganisms. To avoid damaging exuberant or chronic inflammation, the inflammatory process must be tightly controlled and terminated once the insult is mitigated. Inflammation resolution is now known to be an active process involving a new genus of lipid mediators, called "specialized proresolving lipid mediators," that includes resolvin D1 (RvD1). We and others have reported that RvD1 counteracts proinflammatory signaling and promotes resolution. A knowledge gap is that the specific cellular targets and mechanisms of action for RvD1 remain largely unknown. In this article, we identified the mechanism whereby RvD1 disrupts inflammatory mediator production induced by the viral mimic polyinosinic-polycytidylic acid [poly(I:C)] in primary human lung epithelial cells. RvD1 strongly suppressed the viral mimic poly(I:C)-induced IL-6 and IL-8 production and proinflammatory signaling involving MAPKs and NF-κB. Most importantly, we found that RvD1 inhibited the phosphorylation of TAK1 (TGF-ß-activated kinase 1), a key upstream regulatory kinase common to both the MAPK and NF-κB pathways, by inhibiting the formation of a poly(I:C)-induced signaling complex composed of TAK1, TAB1 (TAK1 binding protein), and TRAF6 (TNF receptor-associated factor 6). We confirmed that ALX/FPR2 and GPR32, two RvD1 receptors, were expressed on human small airway epithelial cells. Furthermore, blocking these receptors abrogated the inhibitory action of RvD1. In this article, we present the idea that RvD1 has the potential to be used as an anti-inflammatory and proresolving agent, possibly in the context of exuberant host responses to damaging respirable agents such as viruses.


Subject(s)
Docosahexaenoic Acids/immunology , Epithelial Cells/drug effects , MAP Kinase Kinase Kinases/immunology , Poly I-C/pharmacology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Bronchi/drug effects , Bronchi/immunology , Bronchi/pathology , Docosahexaenoic Acids/genetics , Epithelial Cells/immunology , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , MAP Kinase Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Signal Transduction , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/immunology
12.
PLoS One ; 8(5): e63798, 2013.
Article in English | MEDLINE | ID: mdl-23741300

ABSTRACT

Pulmonary Fibrosis (PF) is a devastating progressive disease in which normal lung structure and function is compromised by scarring. Lung fibrosis can be caused by thoracic radiation, injury from chemotherapy and systemic diseases such as rheumatoid arthritis that involve inflammatory responses. CDDO-Me (Methyl 2-cyano-3,12-dioxooleana-1,9(11)dien-28-oate, Bardoxolone methyl) is a novel triterpenoid with anti-fibrotic and anti-inflammatory properties as shown by our in vitro studies. Based on this evidence, we hypothesized that CDDO-Me would reduce lung inflammation, fibrosis and lung function impairment in a bleomycin model of lung injury and fibrosis. To test this hypothesis, mice received bleomycin via oropharyngeal aspiration (OA) on day zero and CDDO-Me during the inflammatory phase from days -1 to 9 every other day. Bronchoalveolar lavage fluid (BALF) and lung tissue were harvested on day 7 to evaluate inflammation, while fibrosis and lung function were evaluated on day 21. On day 7, CDDO-Me reduced total BALF protein by 50%, alveolar macrophage infiltration by 40%, neutrophil infiltration by 90% (p≤0.01), inhibited production of the inflammatory cytokines KC and IL-6 by over 90% (p≤0.001), and excess production of the pro-fibrotic cytokine TGFß by 50%. CDDO-Me also inhibited α-smooth muscle actin and fibronectin mRNA by 50% (p≤0.05). On day 21, CDDO-Me treatment reduced histological fibrosis, collagen deposition and αSMA production. Lung function was significantly improved at day 21 by treatment with CDDO-Me, as demonstrated by respiratory rate and dynamic compliance. These new findings reveal that CDDO-Me exhibits potent anti-fibrotic and anti-inflammatory properties in vivo. CDDO-Me is a potential new class of drugs to arrest inflammation and ameliorate fibrosis in patients who are predisposed to lung injury and fibrosis incited by cancer treatments (e.g. chemotherapy and radiation) and by systemic autoimmune diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Macrophages, Alveolar/drug effects , Neutrophil Infiltration/drug effects , Oleanolic Acid/analogs & derivatives , Pulmonary Fibrosis/drug therapy , Actins/antagonists & inhibitors , Actins/genetics , Actins/metabolism , Administration, Inhalation , Animals , Bleomycin , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Collagen/antagonists & inhibitors , Collagen/genetics , Collagen/metabolism , Fibronectins/antagonists & inhibitors , Fibronectins/genetics , Fibronectins/metabolism , Gene Expression/drug effects , Humans , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Oleanolic Acid/pharmacology , Pneumonia/chemically induced , Pneumonia/drug therapy , Pneumonia/immunology , Pneumonia/pathology , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Respiratory Function Tests , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
13.
Am J Physiol Lung Cell Mol Physiol ; 305(2): L165-74, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23686858

ABSTRACT

Cigarette smoke is a profound proinflammatory stimulus that causes acute lung inflammation and chronic lung disease, including chronic obstructive pulmonary disease (COPD, emphysema, and chronic bronchitis), via a variety of mechanisms, including oxidative stress. Cigarette smoke contains high levels of free radicals, whereas inflammatory cells, including macrophages and neutrophils, express enzymes, including NADPH oxidase, nitric oxide synthase, and myeloperoxidase, that generate reactive oxygen species in situ and contribute to inflammation and tissue damage. Neu-164 and Neu-107 are small-molecule inhibitors of myeloperoxidase, as well as potent antioxidants. We hypothesized that Neu-164 and Neu-107 would inhibit acute cigarette smoke-induced inflammation. Adult C57BL/6J mice were exposed to mainstream cigarette smoke for 3 days to induce acute inflammation and were treated daily by inhalation with Neu-164, Neu-107, or dexamethasone as a control. Inflammatory cells and cytokines were assessed by bronchoalveolar lavage and histology. mRNA levels of endogenous antioxidant genes heme oxygenase-1 and glutamate-cysteine ligase modifier subunit were determined by qPCR. Cigarette smoke exposure induced acute lung inflammation with accumulation of neutrophils and upregulation of proinflammatory cytokines, including IL-6, macrophage inflammatory protein-2, and keratinocyte-derived cytokine. Both Neu-164 and Neu-107 significantly reduced the accumulation of inflammatory cells and the expression of inflammatory cytokines as effectively as dexamethasone. Upregulation of endogenous antioxidant genes was dampened. Neu-164 and Neu-107 inhibit acute cigarette smoke-induced inflammation by scavenging reactive oxygen species in cigarette smoke and by inhibiting further oxidative stress caused by inflammatory cells. These compounds may have promise in preventing or treating lung disease associated with chronic smoke exposure, including COPD.


Subject(s)
Antioxidants/pharmacology , Caffeic Acids/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Lung/enzymology , Oxidative Stress/drug effects , Peroxidase/antagonists & inhibitors , Pneumonia/enzymology , Smoking/adverse effects , Adult , Animals , Cytokines/biosynthesis , Female , Gene Expression Regulation, Enzymologic/drug effects , Heme Oxygenase-1/biosynthesis , Humans , Lung/pathology , Membrane Proteins/biosynthesis , Mice , Peroxidase/metabolism , Pneumonia/chemically induced , Pneumonia/pathology , Pulmonary Disease, Chronic Obstructive/chemically induced , Pulmonary Disease, Chronic Obstructive/enzymology , Pulmonary Disease, Chronic Obstructive/pathology , RNA, Messenger/biosynthesis , Tobacco Smoke Pollution/adverse effects , Up-Regulation/drug effects
14.
PLoS One ; 8(3): e58258, 2013.
Article in English | MEDLINE | ID: mdl-23484005

ABSTRACT

INTRODUCTION: Cigarette smoke is a profound pro-inflammatory stimulus that contributes to acute lung injuries and to chronic lung disease including COPD (emphysema and chronic bronchitis). Until recently, it was assumed that resolution of inflammation was a passive process that occurred once the inflammatory stimulus was removed. It is now recognized that resolution of inflammation is a bioactive process, mediated by specialized lipid mediators, and that normal homeostasis is maintained by a balance between pro-inflammatory and pro-resolving pathways. These novel small lipid mediators, including the resolvins, protectins and maresins, are bioactive products mainly derived from dietary omega-3 and omega-6 polyunsaturated fatty acids (PUFA). We hypothesize that resolvin D1 (RvD1) has potent anti-inflammatory and pro-resolving effects in a model of cigarette smoke-induced lung inflammation. METHODS: Primary human lung fibroblasts, small airway epithelial cells and blood monocytes were treated with IL-1ß or cigarette smoke extract in combination with RvD1 in vitro, production of pro-inflammatory mediators was measured. Mice were exposed to dilute mainstream cigarette smoke and treated with RvD1 either concurrently with smoke or after smoking cessation. The effects on lung inflammation and lung macrophage populations were assessed. RESULTS: RvD1 suppressed production of pro-inflammatory mediators by primary human cells in a dose-dependent manner. Treatment of mice with RvD1 concurrently with cigarette smoke exposure significantly reduced neutrophilic lung inflammation and production of pro-inflammatory cytokines, while upregulating the anti-inflammatory cytokine IL-10. RvD1 promoted differentiation of alternatively activated (M2) macrophages and neutrophil efferocytosis. RvD1 also accelerated the resolution of lung inflammation when given after the final smoke exposure. CONCLUSIONS: RvD1 has potent anti-inflammatory and pro-resolving effects in cells and mice exposed to cigarette smoke. Resolvins have strong potential as a novel therapeutic approach to resolve lung injury caused by smoke and pulmonary toxicants.


Subject(s)
Docosahexaenoic Acids/metabolism , Pneumonia/drug therapy , Pneumonia/etiology , Pneumonia/metabolism , Smoking/adverse effects , Analysis of Variance , Animals , Bronchoalveolar Lavage Fluid , Chemokines/metabolism , Cytokines/metabolism , DNA Primers/genetics , Female , Fibroblasts , Homeostasis/physiology , Humans , Lung/pathology , Mice , Mice, Inbred C57BL , Phagocytosis/physiology
15.
J Endocrinol ; 213(2): 173-82, 2012 May.
Article in English | MEDLINE | ID: mdl-22393245

ABSTRACT

Early studies have reported the differential roles of androgen receptor (AR) in different types (luminal, basal intermediate, and stromal) of prostate cancer cells. In vivo mouse model tumor studies using the total prostate epithelial knockout mice (pes-ARKO) also revealed that AR played a suppressive role in proliferation of the CK5(+)/CK8(+) progenitor/intermediate cells but a positive role in the CK5(-)/CK8(+) luminal epithelial cells. Using three different resources (one human basal epithelial cell line, one mouse basal epithelial originated progenitor cell line, and a basal epithelium-specific ARKO mouse model), we here demonstrated that the AR in basal epithelial cells of normal prostate plays a suppressive role in their proliferation but a positive role in differentiation into luminal epithelial cells. These results led us to conclude that ARs may play a negative role to suppress CK5(+) basal epithelial and progenitor cell proliferation, yet play an essential role to drive basal epithelial cells into more differentiated states. These results may explain why differential AR expression in different cell types within normal prostate is needed and suggest that ARs in prostate basal epithelial cells, although expressed at a very low level, are necessary to maintain the balance between progenitor cells and differentiated luminal epithelial cells.


Subject(s)
Prostate/cytology , Receptors, Androgen/metabolism , Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Epithelial Cells/cytology , Humans , Male , Mice , Mice, Knockout , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics
16.
Blood ; 112(9): 3661-70, 2008 Nov 01.
Article in English | MEDLINE | ID: mdl-18711002

ABSTRACT

Thrombomodulin (TM), a widely expressing glycoprotein originally identified in vascular endothelium, is an important cofactor in the protein C anticoagulant system. TM appears to exhibit anti-inflammatory ability through both protein C-dependent and -independent pathways. We presently have demonstrated that recombinant N-terminal lectinlike domain of TM (rTMD1) functions as a protective agent against sepsis caused by Gram-negative bacterial infections. rTMD1 caused agglutination of Escherichia coli and Klebsiella pneumoniae and enhanced the macrophage phagocytosis of these Gram-negative bacteria. Moreover, rTMD1 bound to the Klebsiella pneumoniae and lipopolysaccharide (LPS) by specifically interacting with Lewis Y antigen. rTMD1 inhibited LPS-induced inflammatory mediator production via interference with CD14 and LPS binding. Furthermore, rTMD1 modulated LPS-induced mitogen-activated protein kinase and nuclear factor-kappaB signaling pathway activations and inducible nitric oxide synthase expression in macrophages. Administration of rTMD1 protected the host by suppressing inflammatory responses induced by LPS and Gram-negative bacteria, and enhanced LPS and bacterial clearance in sepsis. Thus, rTMD1 can be used to defend against bacterial infection and inhibit LPS-induced inflammatory responses, suggesting that rTMD1 may be valuable in the treatment of severe inflammation in sepsis, especially in Gram-negative bacterial infections.


Subject(s)
Lewis Blood Group Antigens/metabolism , Thrombomodulin/chemistry , Thrombomodulin/metabolism , Animals , Binding Sites , Cell Line , Gram-Negative Bacterial Infections/drug therapy , Humans , Inflammation/drug therapy , Inflammation/prevention & control , Klebsiella pneumoniae/metabolism , Klebsiella pneumoniae/pathogenicity , Ligands , Lipopolysaccharides/metabolism , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , NF-kappa B/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sepsis/drug therapy , Signal Transduction/drug effects , Thrombomodulin/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...