Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
2.
Vaccine ; 38(6): 1505-1512, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31848051

ABSTRACT

BACKGROUND: Despite the fact that vaccines save 2-3 million lives worldwide every year, a percentage of children are not getting appropriately vaccinated, thus leading to disease outbreaks. One of the major reasons of low vaccine uptake in Europe is vaccine hesitancy, contributing to the recent measles outbreaks. Monitoring of vaccine hesitancy is valuable in early identification of vaccine concerns. METHODS: We performed an eighteen country European survey on parents' attitudes and behaviors regarding their children's immunization. Parents having at least one child 1-4 years old were mostly recruited by primary care paediatricians to reply to a web-based questionnaire. The questionnaire was developed by the European Academy of Paediatrics Research in Ambulatory Setting Network steering committee, based on similar surveys. An individual level hesitancy score was constructed using the answers to 21 questions, and correlations of the score with socio-demographic characteristics and types of providers were explored. To assess inter country differences, a country level self -reported confidence was defined. RESULTS: Fifty six percent and 24% of 5736 respondents defined themselves as "not at all hesitant", and "somewhat hesitant", respectively. Parents who consulted general practitioners were more hesitant than parents who consulted pediatricians (p < 0.05). Consultation with homeopathists was associated with the highest reported hesitancy (p < 0.05). Vaccine confidence was highest in Portugal and Cyprus, and lowest in Bulgaria and Poland. CONCLUSION: The majority of parents in Europe believe in the importance of childhood vaccination. However, significant lack of confidence was found in certain European countries, highlighting the need for continuous monitoring, awareness and response plans. The possible influence of different types of healthcare providers on parental decisions demonstrated for the first time in our survey, calls for further research. Monitoring and continuous medical education efforts aimed mostly at those professionals who might not be likely to recommend vaccination are suggested.


Subject(s)
Health Knowledge, Attitudes, Practice , Parents/psychology , Vaccination/psychology , Vaccines , Bulgaria , Child, Preschool , Cyprus , Europe , Humans , Infant , Poland , Portugal , Surveys and Questionnaires
3.
Am J Med Genet A ; 173(11): 3098-3103, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28884921

ABSTRACT

Mutations in GLE1 underlie Lethal Congenital Contracture syndrome (LCCS) and Lethal Arthrogryposis with Anterior Horn Cell Disease (LAAHD). Both LCCS and LAAHD are characterized by reduced fetal movements, congenital contractures, and a severe form of motor neuron disease that results in fetal death or death in the perinatal period, respectively. We identified bi-allelic mutations in GLE1 in two unrelated individuals with motor delays, feeding difficulties, and respiratory insufficiency who survived beyond the perinatal period. Each affected child had missense variants predicted to result in amino acid substitutions near the C-terminus of GLE1 that are predicted to disrupt protein-protein interaction or GLE1 protein targeting. We hypothesize that mutations that preserve function of the coiled-coil domain of GLE1 cause LAAHD whereas mutations that abolish the function of the coiled-coil domain cause LCCS. The phenotype of LAAHD is now expanded to include multiple individuals surviving into childhood suggesting that LAAHD is a misnomer and should be re-named Arthrogryposis with Anterior Horn Cell Disease (AAHD).


Subject(s)
Arthrogryposis/genetics , Motor Disorders/genetics , Motor Neuron Disease/genetics , Nucleocytoplasmic Transport Proteins/genetics , Arthrogryposis/physiopathology , Child, Preschool , Female , Genetic Predisposition to Disease , Humans , Male , Motor Disorders/physiopathology , Motor Neuron Disease/physiopathology , Mutation , Pedigree , Pregnancy
4.
Respir Res ; 17: 36, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27044262

ABSTRACT

In human lung fibrotic lesions, fibroblasts were shown to be closely associated with immature dendritic cell (DC) accumulation. The aim of the present pilot study was to characterize the role of pulmonary fibroblasts on DC phenotype and function, using co-culture of lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) and from control patients, with a DC cell line MUTZ-3. We observed that co-culture of lung control and IPF fibroblasts with DCs reduced the expression of specific DC markers and down-regulated their T-cell stimulatory activity. This suggests that pulmonary fibroblasts might sustain chronic inflammation in the fibrotic lung by maintaining in situ a pool of immature DCs.


Subject(s)
Cell Communication/immunology , Dendritic Cells/cytology , Fibroblasts/cytology , Fibroblasts/immunology , Lung/cytology , Lung/immunology , Cell Proliferation/physiology , Cells, Cultured , Cytokines/immunology , Humans , In Vitro Techniques , Phenotype , Pilot Projects
5.
PLoS One ; 8(1): e53736, 2013.
Article in English | MEDLINE | ID: mdl-23341987

ABSTRACT

BACKGROUND: Fibrocytes are circulating precursors for fibroblasts. Blood fibrocytes are increased in patients with idiopathic pulmonary fibrosis (IPF). The aim of this study was to determine whether alveolar fibrocytes are detected in broncho-alveolar lavage (BAL), to identify their prognostic value, and their potential association with culture of fibroblasts from BAL. METHODS: We quantified fibrocytes in BAL from 26 patients with IPF, 9 patients with Systemic Sclerosis(SSc)-interstitial lung disease (ILD), and 11 controls. BAL cells were cultured to isolate alveolar fibroblasts. RESULTS: Fibrocytes were detected in BAL in 14/26 IPF (54%) and 5/9 SSc patients (55%), and never in controls. Fibrocytes were in median 2.5% [0.4-19.7] and 3.0% [2.7-3.7] of BAL cells in IPF and SSc-ILD patients respectively. In IPF patients, the number of alveolar fibrocytes was correlated with the number of alveolar macrophages and was associated with a less severe disease but not with a better outcome. Fibroblasts were cultured from BAL in 12/26 IPF (46%), 5/9 SSc-ILD (65%) and never in controls. The detection of BAL fibrocytes did not predict a positive culture of fibroblasts. CONCLUSION: Fibrocytes were detected in BAL fluid in about half of the patients with IPF and SSc-ILD. Their number was associated with less severe disease in IPF patients and did not associate with the capacity to grow fibroblasts from BAL fluid.


Subject(s)
Fibroblasts/pathology , Idiopathic Pulmonary Fibrosis/pathology , Pulmonary Alveoli/pathology , Scleroderma, Systemic/pathology , Aged , Aged, 80 and over , Bronchoalveolar Lavage , Cell Count , Female , Humans , Idiopathic Pulmonary Fibrosis/therapy , Male , Middle Aged , Scleroderma, Systemic/therapy , Time Factors
6.
FASEB J ; 27(4): 1549-60, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23288928

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease characterized by myofibroblast proliferation. Transition of epithelial/mesothelial cells into myofibroblasts [epithelial-to-mesenchymal transition (EMT)] occurs under the influence of transforming growth factor (TGF)-ß1, with Snail being a major transcription factor. We study here the role of the heat-shock protein HSP27 in fibrogenesis and EMT. In vitro, we have up- and down-modulated HSP27 expression in mesothelial and epithelial cell lines and studied the expression of different EMT markers induced by TGF-ß1. In vivo, we inhibited HSP27 with the antisense oligonucleotide OGX-427 (in phase II clinical trials as anticancer agent) in our rat subpleural/pulmonary fibrosis models. We demonstrate that HSP27 is strongly expressed during the fibrotic process in patients with IPF and in different in vivo models. We showed that HSP27 binds to and stabilizes Snail and consequently induces EMT. Conversely, HSP27 knockdown leads to Snail proteasomal degradation, thus inhibiting TGF-ß1-induced EMT. Inhibition of HSP27 with OGX-427 efficiently blocks EMT and fibrosis development. Controls in vivo were an empty adenovirus that did not induce fibrosis and a control antisense oligonucleotide. The present work opens the possibility of a new therapeutic use for HSP27 inhibitors against IPF, for which there is no conclusively effective treatment.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , HSP27 Heat-Shock Proteins/antagonists & inhibitors , Snails/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cadherins/metabolism , Cell Line , Epithelial Cells/metabolism , Fibrosis/metabolism , Humans , Oligonucleotides, Antisense/pharmacology , Rats , Rats, Sprague-Dawley , Thionucleotides/pharmacology , Transcription Factors/metabolism
7.
Am J Pathol ; 181(6): 2126-37, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23031257

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating disease of unknown cause. Key signaling developmental pathways are aberrantly expressed in IPF. The hedgehog pathway plays a key role during fetal lung development and may be involved in lung fibrogenesis. We determined the expression pattern of several Sonic hedgehog (SHH) pathway members in normal and IPF human lung biopsies and primary fibroblasts. The effect of hedgehog pathway inhibition was assayed by lung fibroblast proliferation and differentiation with and without transforming growth factor (TGF)-ß1. We showed that the hedgehog pathway was reactivated in the IPF lung. Importantly, we deciphered the cross talk between the hedgehog and TGF-ß pathway in human lung fibroblasts. TGF-ß1 modulated the expression of key components of the hedgehog pathway independent of Smoothened, the obligatory signal transducer of the pathway. Smoothened was required for TGF-ß1-induced myofibroblastic differentiation of control fibroblasts, but differentiation of IPF fibroblasts was partially resistant to Smoothened inhibition. Furthermore, functional hedgehog pathway machinery from the primary cilium, as well as GLI-dependent transcription in the nucleus, was required for the TGF-ß1 effects on normal and IPF fibroblasts during myofibroblastic differentiation. These data identify the GLI transcription factors as potential therapeutic targets in lung fibrosis.


Subject(s)
Cell Differentiation , Hedgehog Proteins/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Transforming Growth Factor beta/metabolism , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Proliferation/drug effects , Cilia/drug effects , Cilia/pathology , Female , Gene Expression Regulation/drug effects , Hedgehog Proteins/genetics , Humans , Idiopathic Pulmonary Fibrosis/genetics , Immunohistochemistry , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Middle Aged , Models, Biological , Myofibroblasts/drug effects , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Veratrum Alkaloids/pharmacology
8.
Proc Am Thorac Soc ; 9(3): 158-63, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22802291

ABSTRACT

Idiopathic pulmonary fibrosis is currently believed to be driven by alveolar epithelial cells, with abnormally activated alveolar epithelial cells accumulating in an attempt to repair injured alveolar epithelium (1). Thus, targeting the alveolar epithelium to prevent or inhibit the development of pulmonary fibrosis might be an interesting therapeutic option in this disease. Hepatocyte growth factor (HGF) is a growth factor for epithelial and endothelial cells, which is secreted by different cell types, especially fibroblasts and neutrophils. HGF has mitogenic, motogenic, and morphogenic properties and exerts an antiapoptotic action on epithelial and endothelial cells. HGF has also proangiogenic effect. In vitro, HGF inhibits epithelial-to-mesenchymal cell transition and promotes myofibroblast apoptosis. In vivo, HGF has antifibrotic properties demonstrated in experimental models of lung, kidney, heart, skin, and liver fibrosis. Hence, the modulation of HGF may be an attractive target for the treatment of lung fibrosis.


Subject(s)
Hepatocyte Growth Factor/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Apoptosis , Cell Survival , Endothelial Cells/metabolism , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Fibroblasts/metabolism , Humans , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , Transforming Growth Factor beta/metabolism , Wound Healing
9.
Crit Care Med ; 40(7): 2041-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22713216

ABSTRACT

OBJECTIVES: Fibroblast migration is an initiating step in fibroproliferation; its involvement during acute lung injury and acute respiratory distress syndrome remains poorly understood. The aims of this study were: 1) to determine whether bronchoalveolar lavage fluids from patients with acute lung injury/acute respiratory distress syndrome modulate lung fibroblast migration; 2) to assess lung fibroblast migration's clinical relevance; and 3) to evaluate the role of the platelet-derived growth factor pathway in this effect. DESIGN: Prospective cohort study. SETTING: Three intensive care units of a large tertiary referral center. PATIENTS: Ninety-three ventilated patients requiring bronchoalveolar lavage fluids were enrolled (48 with acute respiratory distress syndrome, 33 with acute lung injury, and 12 ventilated patients without acute lung injury/acute respiratory distress syndrome). INTERVENTIONS: After bronchoalveolar lavage fluids collection during standard care, the patients were followed up for 28 days and clinical outcomes were recorded. Migration assays were performed by using a Transwell model; bronchoalveolar lavage fluids platelet-derived growth factor and soluble platelet-derived growth factor receptor-α were characterized by Western blot and measured by ELISA. MEASUREMENTS AND MAIN RESULTS: Most of the bronchoalveolar lavage fluids inhibited basal fibroblast migration. Bronchoalveolar lavage fluids chemotactic index increased with severity of lung injury (28% in patients without acute lung injury/acute respiratory distress syndrome and with acute lung injury vs. 91% in acute respiratory distress syndrome patients; p = .016). In acute lung injury/acute respiratory distress syndrome patients, inhibition of basal fibroblast migration by bronchoalveolar lavage fluids below 52% was independently associated with a lower 28-day mortality (odds ratio [95% confidence interval] 0.313 [0.10-0.98], p = .046). Platelet-derived growth factor-related peptides and soluble platelet-derived growth factor-Rα were detected in all bronchoalveolar lavage fluids from acute lung injury/acute respiratory distress syndrome patients. The effect of bronchoalveolar lavage fluids stimulating migration was inhibited by a specific platelet-derived growth factor receptor inhibitor (AG1296). Bronchoalveolar lavage fluids inhibiting migration reversed the effect of rh-platelet-derived growth factor-BB and reduced by 40% the binding of 125I-platelet-derived growth factor-BB to fibroblast cell surface in favor of a role for platelet-derived growth factor-sRα. CONCLUSIONS: : Together, our results suggest that during acute lung injury, fibroblast migration is modulated by bronchoalveolar lavage fluids through a platelet-derived growth factor/platelet-derived growth factor-sRα balance. Migration is associated with clinical severity and patient 28-day mortality.


Subject(s)
Acute Lung Injury/metabolism , Bronchoalveolar Lavage Fluid/cytology , Cell Movement/physiology , Fibroblasts/physiology , Platelet-Derived Growth Factor/metabolism , Respiratory Distress Syndrome/metabolism , Acute Lung Injury/epidemiology , Aged , Blotting, Western , Bronchoalveolar Lavage , Cell Line , Enzyme Inhibitors/pharmacology , Female , Humans , Intensive Care Units , Interleukin-8/metabolism , Male , Middle Aged , Multiple Organ Failure/mortality , Multivariate Analysis , Neutrophils/metabolism , Prospective Studies , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Respiration, Artificial , Respiratory Distress Syndrome/epidemiology , Severity of Illness Index , Tyrphostins/pharmacology
10.
Crit Care Med ; 40(1): 21-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21926612

ABSTRACT

OBJECTIVE: Fibrocytes are mesenchymal progenitors involved in normal and pathologic repair. The aims of this study were: 1) to quantify fibrocytes in bronchoalveolar lavage fluid from patients with or without acute lung injury and acute respiratory distress syndrome; and 2) to evaluate the prognostic value of bronchoalveolar lavage fibrocyte percentage in patients with acute lung injury and acute respiratory distress syndrome. DESIGN: Prospective cohort study. SETTING: Three intensive care units of a large tertiary referral center. PATIENTS: One hundred twenty-two ventilated patients requiring bronchoalveolar lavage were enrolled (62 acute respiratory distress syndrome, 30 acute lung injury, 30-ventilated patients without acute lung injury and acute respiratory distress syndrome). INTERVENTIONS: After bronchoalveolar lavage collection during standard care, the patients were followed up for 28 days and clinical outcome was recorded. Fibrocytes (CD45+/collagen 1+) were quantified in bronchoalveolar lavage by flow cytometry. Comparison of bronchoalveolar lavage fibrocyte percentage from patients with or without acute lung injury and acute respiratory distress syndrome was performed using a Wilcoxon test. A multivariate analysis using a Cox model was performed to study the independent predictors of survival. MEASUREMENTS AND MAIN RESULTS: Fibrocytes were detected in 90 of 92 (98%) bronchoalveolar lavages from patients with acute lung injury and acute respiratory distress syndrome. The median percentage of bronchoalveolar lavage fibrocytes was significantly higher in patients with acute lung injury and acute respiratory distress syndrome (5.0%) in comparison with ventilated control subjects (0.9%, p < .0001). After adjustment for age, comorbidity of malignancy, and severity of illness, a bronchoalveolar lavage fibrocyte percentage >6% was independently associated with a higher 28-day mortality in patients with acute lung injury and acute respiratory distress syndrome (hazard ratio [95% confidence interval] 6.15 [2.78-13.64], p ≤ .0001). Addition of bronchoalveolar lavage fibrocyte percentage in a clinical model predicting mortality in patients with acute lung injury and acute respiratory distress syndrome improved global fit and discriminatory capacity (c-statistic, 0.78-0.85; p = .007). CONCLUSIONS: Fibrocytes are detectable in bronchoalveolar lavage during acute lung injury and acute respiratory distress syndrome. A bronchoalveolar lavage fibrocyte percentage >6% provides an additive prognostic value to clinical predictors and may be useful to identify patients with acute lung injury and acute respiratory distress syndrome at highest risk of an adverse outcome.


Subject(s)
Acute Lung Injury/diagnosis , Pulmonary Alveoli/cytology , Acute Lung Injury/pathology , Age Factors , Aged , Bronchoalveolar Lavage Fluid/cytology , Female , Flow Cytometry , Humans , Male , Middle Aged , Prognosis , Prospective Studies , Pulmonary Alveoli/pathology , Respiration, Artificial , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/pathology , Treatment Outcome
11.
Am J Respir Crit Care Med ; 183(6): 759-66, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-20935114

ABSTRACT

RATIONALE: Injury to alveolar epithelial cells is central to the pathophysiology of idiopathic pulmonary fibrosis (IPF). An abnormal autoimmune response directed against antigens of the alveolar epithelium may contribute to the disease. OBJECTIVES: To detect circulating autoantibodies (autoAbs) directed against epithelial structures. METHODS: We performed immunoblot by separating human placental amnion extract or alveolar epithelial cell (A549 cell line) proteins on polyacrylamide gels, blotting on nitrocellulose membranes, and incubating with serum from patients with IPF (n = 40) or healthy subjects (n = 40). Proteomic analysis and mass spectrometry characterized the target protein. Inhibition experiments performed with the correspondent recombinant protein confirmed our results. MEASUREMENTS AND MAIN RESULTS: We identified IgG autoAbs recognizing a 200-kD protein in the serum of patients with IPF. Proteomic analysis identified this protein as human periplakin (PPL), a component of desmosomes. Anti-PPL Abs were found by immunoblot in both serum and bronchoalveolar lavage in patients with IPF: 16/40 (40%) of them were positive versus none of the control subjects. Immunohistochemistry revealed that PPL was strongly expressed in bronchial and alveolar epithelium, but that PPL exhibited changes in intracellular localization among normal and fibrotic alveolar epithelium. In an alveolar epithelial wound repair assay, an anti-PPL IgG decreased cell migration. Recombinant PPL induced bronchoalveolar lavage T lymphocyte proliferation. Patients with IPF with anti-PPL Abs had a more severe respiratory disease, despite no difference in survival. CONCLUSIONS: We found a new circulating autoAb directed against PPL in patients with IPF, associated with a more severe disease.


Subject(s)
Autoantibodies/blood , Autoimmunity , Idiopathic Pulmonary Fibrosis/immunology , Immunoglobulin G/blood , Plakins/blood , Adult , Aged , Biomarkers/blood , Case-Control Studies , Female , Humans , Idiopathic Pulmonary Fibrosis/blood , Idiopathic Pulmonary Fibrosis/physiopathology , Male , Middle Aged , Respiratory Mucosa/immunology , Severity of Illness Index
12.
PLoS One ; 5(5): e10886, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20526373

ABSTRACT

BACKGROUND: Few data concern the pathophysiology of primary spontaneous pneumothorax (PSP), which is associated with alveolar hypoxia/reoxygenation. This study tested the hypothesis that PSP is associated with oxidative stress in lung macrophages. We analysed expression of the oxidative stress marker 4-HNE; the antioxidant and anti-inflammatory proteins heme oxygenase-1 (HO-1), biliverdin reductase (BVR) and heavy chain of ferritin (H-ferritin); and the transcription factors controlling their expression Nrf2 and HIF-1alpha, in lung samples from smoker and nonsmoker patients with PSP (PSP-S and PSP-NS), cigarette smoke being a risk factor of recurrence of the disease. METHODOLOGY/PRINCIPAL FINDINGS: mRNA was assessed by RT-PCR and proteins by western blot, immunohistochemistry and confocal laser analysis. 4-HNE, HO-1, BVR and H-ferritin were increased in macrophages from PSP-S as compared to PSP-NS and controls (C). HO-1 increase was associated with increased expression of HIF-1alpha mRNA and protein in alveolar macrophages in PSP-S patients, whereas Nrf2 was not modified. To understand the regulation of HO-1, BVR and H-ferritin, THP-1 macrophages were exposed to conditions mimicking conditions in C, PSP-S and PSP-NS patients: cigarette smoke condensate (CS) or air exposure followed or not by hypoxia/reoxygenation. Silencing RNA experiments confirmed that HIF-1alpha nuclear translocation was responsible for HO-1, BVR and H-ferritin induction mediated by CS and hypoxia/reoxygenation. CONCLUSIONS/SIGNIFICANCE: PSP in smokers is associated with lung macrophage oxidative stress. The response to this condition involves HIF-1alpha-mediated induction of HO-1, BVR and H-ferritin.


Subject(s)
Apoferritins/metabolism , Heme Oxygenase-1/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/enzymology , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pneumothorax/enzymology , Smoking/metabolism , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Apoferritins/genetics , Biopsy , Blood Proteins/genetics , Blood Proteins/metabolism , Cell Hypoxia , Cell Line , Enzyme Induction , Eosinophil Major Basic Protein , Gene Expression Regulation , Gene Silencing , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lung/pathology , Macrophages/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , Oxygen/metabolism , Pneumothorax/pathology , Proteoglycans/genetics , Proteoglycans/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism
13.
Am J Respir Crit Care Med ; 182(3): 385-95, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20395561

ABSTRACT

RATIONALE: Lung dendritic cells (DCs) have been shown to accumulate in human fibrotic lung disease, but little is known concerning a role for DCs in the pathogenesis of fibrotic lung. OBJECTIVES: To characterize lung DCs in an in vivo model of bleomycin-induced pulmonary fibrosis in mice. METHODS: We characterized the kinetics and activation of pulmonary DCs during the course of bleomycin-induced lung injury by flow cytometry on lung single-cell suspensions. We also characterized the lymphocytes accumulating in bleomycin lung and the chemokines susceptible to favor the recruitment of immune cells. MEASUREMENTS AND MAIN RESULTS: We show, for the first time, that increased numbers of CD11c(+)/major histocompatibility complex class II(+) DCs, including CD11b(hi) monocyte-derived inflammatory DCs, infiltrate the lung of treated animals during the fibrotic phase of the response to bleomycin. These DCs are mature DCs expressing CD40, CD86, and CD83. They are associated with increased numbers of recently activated memory T cells expressing CD44, CD40L, and CD28, suggesting that fully mature DCs and Ag-experienced T cells can drive an efficient effector immune response within bleomycin lung. Most importantly, when DCs are inactivated with VAG539, a recently described new immunomodulator, VAG539 treatment attenuates the hallmarks of bleomycin lung injury. CONCLUSIONS: These findings identify lung DCs as key proinflammatory cells potentially able to sustain pulmonary inflammation and fibrosis in the bleomycin model.


Subject(s)
Dendritic Cells/metabolism , Lung/pathology , Pulmonary Fibrosis/pathology , Animals , Antigens, CD/metabolism , Bleomycin/pharmacology , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid/cytology , Chemokines/metabolism , Disease Models, Animal , Flow Cytometry , Immunologic Factors/pharmacology , Lung/metabolism , Major Histocompatibility Complex/physiology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , T-Lymphocytes/metabolism
14.
EMBO Mol Med ; 2(1): 26-37, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20043279

ABSTRACT

Sodium transport via epithelial sodium channels (ENaC) expressed in alveolar epithelial cells (AEC) provides the driving force for removal of fluid from the alveolar space. The membrane-bound channel-activating protease 1 (CAP1/Prss8) activates ENaC in vitro in various expression systems. To study the role of CAP1/Prss8 in alveolar sodium transport and lung fluid balance in vivo, we generated mice lacking CAP1/Prss8 in the alveolar epithelium using conditional Cre-loxP-mediated recombination. Deficiency of CAP1/Prss8 in AEC induced in vitro a 40% decrease in ENaC-mediated sodium currents. Sodium-driven alveolar fluid clearance (AFC) was reduced in CAP1/Prss8-deficient mice, due to a 48% decrease in amiloride-sensitive clearance, and was less sensitive to beta(2)-agonist treatment. Intra-alveolar treatment with neutrophil elastase, a soluble serine protease activating ENaC at the cell surface, fully restored basal AFC and the stimulation by beta(2)-agonists. Finally, acute volume-overload increased alveolar lining fluid volume in CAP1/Prss8-deficient mice. This study reveals that CAP1 plays a crucial role in the regulation of ENaC-mediated alveolar sodium and water transport and in mouse lung fluid balance.


Subject(s)
Epithelial Sodium Channels/metabolism , Extravascular Lung Water/metabolism , Lung/metabolism , Pulmonary Alveoli/metabolism , Serine Endopeptidases/metabolism , Water-Electrolyte Balance , Adrenergic beta-Agonists/pharmacology , Animals , Cells, Cultured , Epithelial Cells/metabolism , Epithelial Sodium Channels/genetics , Gene Deletion , Gene Expression , Mice , Pulmonary Alveoli/cytology , Pulmonary Edema/metabolism , Respiratory Mucosa/metabolism , Serine Endopeptidases/genetics , Water-Electrolyte Balance/drug effects
15.
Am J Respir Cell Mol Biol ; 42(3): 286-93, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19448157

ABSTRACT

Hepatocyte growth factor (HGF) is a growth factor for alveolar epithelial cells. Activation of pro-HGF to HGF is regulated by the HGF activator (HGFA), a serine protease, and a specific inhibitor (HGFA inhibitor-1, HAI-1). An imbalance in the HGFA/HAI-1 system might contribute to lung fibrosis. Pro-HGF activation capacity from bronchoalveolar lavage (BAL) fluid was evaluated 3, 7, and 14 days after the intratracheal bleomycin injection (Bleo) in mice with or without thrombin. BAL fluid from naïve mice was used as control. HGFA and HAI-1 mRNA were evaluated by QPCR in the whole lung or by Western blot in BAL fluid. BAL fluid from control mice and Bleo mice activated pro-HGF in vitro at a similar degree. Thrombin accelerated proHGF activation by Bleo BAL on Day 3 and Day 7, but not on Day 14, or in control BAL. Incubation of pro-HGF with BAL from Bleo Day 3 and Day 7 mice increased phosphorylation of HGFR on A549 cells. Thrombin-induced pro-HGF activation was inhibited by an anti-HGFA antibody and accelerated by an anti-HAI-1 antibody. Active HGFA was not detected in control BAL and was strongly induced in Bleo BAL. HGFA concentrations were higher on Day 3 and Day 7 than on Day 14. HAI-1 was detected at low levels in control BAL and increased strongly by Day 3 with stable concentrations until Day 14. By demonstrating an imbalance between HGFA and HAI-1 expression in BAL fluid, our results highlight a defective thrombin-dependent proHGF activation system at the fibrotic phase of bleomycin-induced pulmonary fibrosis.


Subject(s)
Bleomycin , Hepatocyte Growth Factor/metabolism , Protein Precursors/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Animals , Antibodies, Neutralizing/immunology , Bronchoalveolar Lavage Fluid , Cell Line , Epithelial Cells/metabolism , Gene Expression Regulation , Humans , Lung/metabolism , Lung/pathology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Proteinase Inhibitory Proteins, Secretory , Proto-Oncogene Proteins c-met/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Pulmonary Fibrosis/pathology , Serine Endopeptidases/genetics , Serine Endopeptidases/immunology , Serine Endopeptidases/metabolism
16.
Rev Prat ; 58(10): 1063-9, 2008 May 31.
Article in French | MEDLINE | ID: mdl-18652404

ABSTRACT

Sarcoidosis results from an uncontrolled cell-mediated immune reaction involving activated T cells and monocytes-macrophages, characterized by the formation of typical granulomas at the sites of the lesions. The interactions between both cell types induce the release of several mediators that play a central role in the development of granulomas, allowing to design new therapies. The aetiology of sarcoidosis remains unknown, but the disease is currently viewed as the consequence of a chronic immunological response associating a genetic susceptibility and specific environmental factors.


Subject(s)
Sarcoidosis/physiopathology , Cytokines/metabolism , Environment , Genetic Predisposition to Disease , Granuloma/physiopathology , Humans , Sarcoidosis/etiology
17.
Orphanet J Rare Dis ; 2: 46, 2007 Nov 19.
Article in English | MEDLINE | ID: mdl-18021432

ABSTRACT

Sarcoidosis is a multisystemic disorder of unknown cause characterized by the formation of immune granulomas in involved organs. It is an ubiquitous disease with incidence (varying according to age, sex, race and geographic origin) estimated at around 16.5/100,000 in men and 19/100,000 in women. The lung and the lymphatic system are predominantly affected but virtually every organ may be involved. Other severe manifestations result from cardiac, neurological, ocular, kidney or laryngeal localizations. In most cases, sarcoidosis is revealed by persistent dry cough, eye or skin manifestations, peripheral lymph nodes, fatigue, weight loss, fever or night sweats, and erythema nodosum. Abnormal metabolism of vitamin D3 within granulomatous lesions and hypercalcemia are possible. Chest radiography is abnormal in about 90% of cases and shows lymphadenopathy and/or pulmonary infiltrates (without or with fibrosis), defining sarcoidosis stages from I to IV. The etiology remains unknown but the prevailing hypothesis is that various unidentified, likely poorly degradable antigens of either infectious or environmental origin could trigger an exaggerated immune reaction in genetically susceptible hosts. Diagnosis relies on compatible clinical and radiographic manifestations, evidence of non-caseating granulomas obtained by biopsy through tracheobronchial endoscopy or at other sites, and exclusion of all other granulomatous diseases. The evolution and severity of sarcoidosis are highly variable. Mortality is estimated at between 0.5-5%. In most benign cases (spontaneous resolution within 24-36 months), no treatment is required but a regular follow-up until recovery is necessary. In more serious cases, a medical treatment has to be prescribed either initially or at some point during follow-up according to clinical manifestations and their evolution. Systemic corticosteroids are the mainstay of treatment of sarcoidosis. The minimal duration of treatment is 12 months. Some patients experience repeated relapses and may require long-term low-dose corticosteroid therapy during years. Other treatments (immunosuppressive drugs and aminoquinolins) may be useful in case of unsatisfactory response to corticosteroids, poor tolerance and as sparing agents when high doses of corticosteroids are needed for a long time. In some strictly selected cases refractory to standard therapy, specific antiTNF-alpha agents may offer precious improvement. Some patients benefit from topical corticosteroids.


Subject(s)
Sarcoidosis/diagnosis , Sarcoidosis/physiopathology , Diagnosis, Differential , Humans , Sarcoidosis/therapy
18.
Am J Respir Crit Care Med ; 176(10): 1007-14, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17717200

ABSTRACT

RATIONALE: There is growing evidence that resident cells, such as fibroblasts and epithelial cells, can drive the persistent accumulation of dendritic cells (DCs) in chronically inflamed tissue, leading to the organization and the maintenance of ectopic lymphoid aggregates. This phenomenon, occurring through a chemokine-mediated retention mechanism, has been documented in various disorders, but not in fibrotic interstitial lung disorders in which the presence of organized lymphoid follicles has been documented. OBJECTIVES: To characterize the distribution of DCs in fibrotic lung, and to analyze the expression of the main chemokines known to regulate DC recruitment. METHODS: Lung resection tissue (lungs with idiopathic pulmonary fibrosis; n = 12; lungs with nonspecific interstitial pneumonia, n = 5; control lungs, n = 5) was snap-frozen for subsequent immunohistochemical techniques on serial sections and reverse transcriptase-polymerase chain reaction analysis. MEASUREMENTS AND MAIN RESULTS: Results were similar in idiopathic pulmonary fibrosis and nonspecific interstitial pneumonia lungs, which were heavily infiltrated by immature DCs in established fibrosis and in areas of epithelial hyperplasia. Altered epithelial cells and fibroblasts, particularly in fibroblastic foci, frankly expressed all chemokines (CCL19, CCL20, CCL22, and CXCL12) susceptible to favor the recruitment of immune cells. Lymphoid follicles were infiltrated by maturing DCs, which could originate from the pool of DCs accumulating in their vicinity. CONCLUSIONS: These findings suggest that resident cells in pulmonary fibrosis can sustain chronic inflammation by driving the accumulation of DCs with the potential to mature locally within ectopic lymphoid follicles. Future strategies should consider DCs or chemokines as therapeutic targets in the treatment of pulmonary fibrosis.


Subject(s)
Chemokines, CC/metabolism , Dendritic Cells/physiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Adult , Aged , Case-Control Studies , Cell Count , Cell Movement , Epithelial Cells/physiology , Female , Fibroblasts/physiology , Humans , Male , Middle Aged
19.
Rev Prat ; 57(20): 2222-6, 2007 Dec 31.
Article in French | MEDLINE | ID: mdl-18320741

ABSTRACT

Despite the continuous and renewed interest in IPF, the precise biological mechanisms underlying the development of fibrosis and leading to the irreversible destruction of the lung are still unknown. Inflammation seems to play a minor role at the initiation of the disease. Identification of excessive apoptosis of alveolar epithelial cells led to the hypothesis that the disorder results from repeated alveolar epithelial cell injury and activation. In turn, alveolar epithelial cells induce the recruitment, proliferation, and activation of mesenchymal cells with the formation of fibroblastic foci and the abnormal accumulation of extracellular matrix. Fibroblastic foci are connected in a tridimensional reticulum. Circulating mesenchymal precursors called fibrocytes, and transdifferenciation of epithelial cells, endothelial cells and/or mesothelial cells, may all contribute to the accumulation of fibroblasts in the lung.


Subject(s)
Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/physiopathology , Apoptosis , Cell Proliferation , Cell Transdifferentiation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Fibroblasts/metabolism , Humans , Inflammation/physiopathology
20.
J Nucl Med ; 47(8): 1281-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16883006

ABSTRACT

UNLABELLED: Idiopathic pulmonary fibrosis (IPF) is characterized by an uncontrolled accumulation and activation of lung fibroblasts. A modulation of fibroblast activation has been observed in various systems with octreotide, a synthetic somatostatin analog with strong affinity for the somatostatin receptor subtype 2 (sst2). One aim of our study was to evaluate the expression of somatostatin receptors in the lungs of patients with IPF. A second aim was to evaluate the relationship between 111In-octreotide uptake and the effect of pulmonary fibrosis as assessed by lung function tests and parameters and by radiologic findings. METHODS: We investigated 11 patients with IPF, 6 patients with pulmonary fibrosis associated with systemic sclerosis (SSc), and 19 patients with disease not of the lung (control patients). The expression of somatostatin receptors was evaluated in vivo using 111In-octreotide scintigraphy. We evaluated the relationship between 111In-octreotide uptake and the activity of pulmonary fibrosis as assessed by lung function tests, bronchoalveolar lavage (BAL) cellularity, and high-resolution CT (HRCT) of the chest. Planar images and thoracic SPECT (24 h) were performed after injection of 222 MBq of 111In-octreotide. Lung uptake was quantified using the lung-to-background ratio (L/B). In addition, the expression of sst2 was evaluated in vitro, in frozen lung-tissue samples using autoradiography, and in human cultures of lung fibroblasts using a ligand-binding assay. RESULTS: Compared with lung uptake in control patients (median L/B, 1.25; range, 1.14-1.49), lung uptake was increased in all 11 IPF patients (median L/B, 2.63; range, 1.59-3.13; P < 0.001) and in 4 of 6 SSc patients (median L/B, 1.68; range, 1.42-2.16). The L/B was lower in SSc patients than in IPF patients (P = 0.011). Increased uptake correlated with the alteration of lung function (carbon monoxide diffusing capacity [rho = -0.655; P = 0.038], diffusing capacity for carbon monoxide and alveolar volume ratio [rho = -0.627; P = 0.047], vital capacity [rho = -0.609; P = 0.054], and total lung capacity [rho = -0.598; P = 0.058]) and with the intensity of alveolitis (total BAL cellularity [rho = 0.756; P = 0.045], neutrophil counts [rho = 0.738; P = 0.05]), and HRCT fibrosis score (rho = 0.673; P = 0.007). Autoradiography suggested that vascular structures were a prominent binding site. Lung fibroblasts expressed somatostatin receptors in vitro as measured by binding assay. CONCLUSION: Our preliminary results identified an increased expression of sst2 in (mainly idiopathic) pulmonary fibrosis. Lung uptake correlates with the alteration of lung function and with the intensity of alveolitis.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Indium Radioisotopes/pharmacokinetics , Octreotide/therapeutic use , Pulmonary Fibrosis/diagnostic imaging , Pulmonary Fibrosis/diagnosis , Adult , Aged , Bronchoalveolar Lavage Fluid , Cells, Cultured/metabolism , Female , Fibroblasts/metabolism , Humans , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Middle Aged , Radionuclide Imaging , Receptors, Somatostatin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...