Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Noncoding RNA Res ; 9(3): 865-875, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38586316

ABSTRACT

Background: Hypersensitivity pneumonitis (HP) is an inflammatory disorder affecting lung parenchyma and often evolves into fibrosis (fHP). The altered regulation of genes involved in the pathogenesis of the disease is not well comprehended, while the role of microRNAs in lung fibroblasts remains unexplored. Methods: We used integrated bulk RNA-Seq and enrichment pathway bioinformatic analyses to identify differentially expressed (DE)-miRNAs and genes (DEGs) associated with HP lungs. In vitro, we evaluated the expression and potential role of miR-155-5p in the phenotype of fHP lung fibroblasts. Loss and gain assays were used to demonstrate the impact of miR-155-5p on fibroblast functions. In addition, mir-155-5p and its target TP53INP1 were analyzed after treatment with TGF-ß, IL-4, and IL-17A. Results: We found around 50 DEGs shared by several databases that differentiate HP from control and IPF lungs, constituting a unique HP lung transcriptional signature. Additionally, we reveal 18 DE-miRNAs that may regulate these DEGs. Among the candidates likely associated with HP pathogenesis was miR-155-5p. Our findings indicate that increased miR-155-5p in fHP fibroblasts coincides with reduced TP53INP1 expression, high proliferative capacity, and a lack of senescence markers compared to IPF fibroblasts. Induced overexpression of miR-155-5p in normal fibroblasts remarkably increases the proliferation rate and decreases TP53INP1 expression. Conversely, miR-155-5p inhibition reduces proliferation and increases senescence markers. TGF-ß, IL-4, and IL-17A stimulated miR-155-5p overexpression in HP lung fibroblasts. Conclusion: Our findings suggest a distinctive signature of 53 DEGs in HP, including CLDN18, EEF2, CXCL9, PLA2G2D, and ZNF683, as potential targets for future studies. Likewise, 18 miRNAs, including miR-155-5p, could be helpful to establish differences between these two pathologies. The overexpression of miR-155-5p and downregulation of TP53INP1 in fHP lung fibroblasts may be involved in his proliferative and profibrotic phenotype. These findings may help differentiate and characterize their pathogenic features and understand their role in the disease.

2.
J Clin Invest ; 133(6)2023 03 15.
Article in English | MEDLINE | ID: mdl-36919699

ABSTRACT

High mobility group A1 (HMGA1) chromatin regulators are upregulated in diverse tumors where they portend adverse outcomes, although how they function in cancer remains unclear. Pancreatic ductal adenocarcinomas (PDACs) are highly lethal tumors characterized by dense desmoplastic stroma composed predominantly of cancer-associated fibroblasts and fibrotic tissue. Here, we uncover an epigenetic program whereby HMGA1 upregulates FGF19 during tumor progression and stroma formation. HMGA1 deficiency disrupts oncogenic properties in vitro while impairing tumor inception and progression in KPC mice and subcutaneous or orthotopic models of PDAC. RNA sequencing revealed HMGA1 transcriptional networks governing proliferation and tumor-stroma interactions, including the FGF19 gene. HMGA1 directly induces FGF19 expression and increases its protein secretion by recruiting active histone marks (H3K4me3, H3K27Ac). Surprisingly, disrupting FGF19 via gene silencing or the FGFR4 inhibitor BLU9931 recapitulates most phenotypes observed with HMGA1 deficiency, decreasing tumor growth and formation of a desmoplastic stroma in mouse models of PDAC. In human PDAC, overexpression of HMGA1 and FGF19 defines a subset of tumors with extremely poor outcomes. Our results reveal what we believe is a new paradigm whereby HMGA1 and FGF19 drive tumor progression and stroma formation, thus illuminating FGF19 as a rational therapeutic target for a molecularly defined PDAC subtype.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Humans , Mice , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Silencing , HMGA1a Protein/genetics , HMGA1a Protein/metabolism , Pancreatic Neoplasms/pathology
3.
Cells ; 11(19)2022 09 27.
Article in English | MEDLINE | ID: mdl-36230977

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is an aging-associated disease characterized by exacerbated extracellular matrix deposition that disrupts oxygen exchange. Hypoxia and its transcription factors (HIF-1α and 2α) influence numerous circuits that could perpetuate fibrosis by increasing myofibroblasts differentiation and by promoting extracellular matrix accumulation. Therefore, this work aimed to elucidate the signature of hypoxia in the transcriptomic circuitry of IPF-derived fibroblasts. To determine this transcriptomic signature, a gene expression analysis with six lines of lung fibroblasts under normoxia or hypoxia was performed: three cell lines were derived from patients with IPF, and three were from healthy donors, a total of 36 replicates. We used the Clariom D platform, which allows us to evaluate a huge number of transcripts, to analyze the response to hypoxia in both controls and IPF. The control's response is greater by the number of genes and complexity. In the search for specific genes responsible for the IPF fibroblast phenotype, nineteen dysregulated genes were found in lung fibroblasts from IPF patients in hypoxia (nine upregulated and ten downregulated). In this sense, the signaling pathways revealed to be affected in the pulmonary fibroblasts of patients with IPF may represent an adaptation to chronic hypoxia.


Subject(s)
Idiopathic Pulmonary Fibrosis , Fibroblasts/metabolism , Humans , Hypoxia/genetics , Hypoxia/metabolism , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Oxygen/metabolism , Transcription Factors/metabolism , Transcriptome/genetics
4.
Int J Toxicol ; 40(6): 506-516, 2021 12.
Article in English | MEDLINE | ID: mdl-34530646

ABSTRACT

Wood smoke (WS) contains many harmful compounds, including polycyclic aromatic hydrocarbons (PAHs). WS induces inflammation in the airways and lungs and can lead to the development of various acute and chronic respiratory diseases. Pulmonary fibroblasts are the main cells involved in the remodeling of the extracellular matrix (ECM) during the WS-induced inflammatory response. Although fibroblasts remain in a low proliferation state under physiological conditions, they actively participate in ECM remodeling during the inflammatory response in pathophysiological states. Consequently, we used normal human lung fibroblasts (NHLFs) to assess the potential effects of the PAHs-containing wood smoke extract (WSE) on the growth rate, total collagen synthesis, and the expression levels of collagen I and III, matrix metalloproteinase (MMP)-1, MMP-2, MMP-9, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, and the transforming growth factor (TGF)-ß1. We also assessed MMPs activity. The results showed that WSE induced a trimodal behavior in the growth rate curves in NHLFs; the growth rate increased with 0.5-1 % WSE and decreased with 2.5% WSE, without causing cell damage; 5-20% WSE inhibited the growth and induced cell damage. After 3 hours of exposure, 2.5% WSE induced an increase in total collagen synthesis and upregulation of TGF-ß1, collagen I and III, MMP-1, TIMP-1, and TIMP-2 expression. However, MMP-2 expression was downregulated and MMP-9 was not expressed. The gelatinase activity of MMP-2 was also increased. These results suggest that WSE affects the ECM remodeling in NHLFs and indicate the potential involvement of PAHs in this process.


Subject(s)
Extracellular Matrix/drug effects , Fibroblasts/drug effects , Inflammation/chemically induced , Inflammation/physiopathology , Lung Diseases/chemically induced , Plant Extracts/adverse effects , Smoke/adverse effects , Cell Proliferation/drug effects , Humans , Magnoliopsida/chemistry , Wood/chemistry
5.
J Immunol Res ; 2021: 6654220, 2021.
Article in English | MEDLINE | ID: mdl-33977111

ABSTRACT

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains as a leading infectious cause of death worldwide. The increasing number of multidrug-resistant TB (MDR-TB) cases contributes to the poor control of the TB epidemic. Currently, little is known about the immunological requirements of protective responses against MDR-TB. This is of major relevance to identify immune markers for treatment monitoring and targets for adjuvant immunotherapies. Here, we hypothesized that MDR-TB patients display unique immunophenotypical features and immune cell migration dynamics compared to drug-sensitive TB (DS-TB). Hence, we prospectively conducted an extensive characterization of the immune profile of MDR-TB patients at different time points before and after pharmacological therapy. For this purpose, we focused on the leukocyte expression of chemokine receptors, distribution of different monocyte and lymphocyte subsets, plasma levels of chemotactic factors, and in vitro migration capacity of immune cells. Our comparative cohort consisted of DS-TB patients and healthy volunteer donors (HD). Our results demonstrate some unique features of leukocyte migration dynamics during MDR-TB. These include increased and prolonged circulation of CD3+ monocytes, CCR4+ monocytes, EM CD4+ T cells, EM/CM CD8+ T cells, and CXCR1+CXCR3+ T cells that is sustained even after the administration of anti-TB drugs. We also observed shared characteristics of both MDR-TB and DS-TB that include CCR2+ monocyte depletion in the blood; high plasma levels of MPC-1, CCL-7, and IP-10; and increased responsiveness of leukocytes to chemotactic signals in vitro. Our study contributes to a better understanding of the MDR-TB pathobiology and uncovers immunological readouts of treatment efficacy.


Subject(s)
Antitubercular Agents/pharmacology , Leukocytes, Mononuclear/immunology , Receptors, Chemokine/metabolism , Tuberculosis, Multidrug-Resistant/immunology , Tuberculosis, Pulmonary/immunology , Adult , Antitubercular Agents/therapeutic use , Biomarkers/analysis , Biomarkers/metabolism , Case-Control Studies , Cell Movement/immunology , Drug Monitoring/methods , Follow-Up Studies , Healthy Volunteers , Humans , Leukocytes, Mononuclear/metabolism , Male , Microbial Sensitivity Tests , Middle Aged , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/isolation & purification , Prospective Studies , Receptors, Chemokine/analysis , Tuberculosis, Multidrug-Resistant/blood , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/drug therapy , Tuberculosis, Pulmonary/microbiology
6.
Eur Respir J ; 58(2)2021 08.
Article in English | MEDLINE | ID: mdl-33446609

ABSTRACT

BACKGROUND: Around 8-10% of individuals over 50 years of age present interstitial lung abnormalities (ILAs), but their risk factors are uncertain. METHODS: From 817 individuals recruited in our lung ageing programme at the Mexican National Institute of Respiratory Diseases, 80 (9.7%) showed ILAs and were compared with 564 individuals of the same cohort with normal high-resolution computed tomography to evaluate demographic and functional differences, and with 80 individuals randomly selected from the same cohort for biomarkers. We evaluated MUC5B variant rs35705950, telomere length, and serum levels of matrix metalloproteinase (MMP)-1, MMP-2, MMP-3, MMP-7, MMP-8, MMP-9, MMP-12, MMP-13, interleukin (IL)-6, surfactant protein (SP)-D, α-Klotho and resistin. RESULTS: Individuals with ILAs were usually males (p<0.005), older than controls (p<0.0001), smokers (p=0.01), with a greater frequency of MUC5B rs35705950 (OR 3.5, 95% CI 1.3-9.4; p=0.01), and reduced diffusing capacity of the lung for carbon monoxide and oxygen saturation. Resistin, IL-6, SP-D, MMP-1, MMP-7 and MMP-13 were significantly increased in individuals with ILAs. Resistin (12±5 versus 9±4 ng·mL-1; p=0.0005) and MMP-13 (357±143 versus 298±116 pg·mL-1; p=0.004) were the most increased biomarkers. On follow-up (24±18 months), 18 individuals showed progression which was associated with gastro-oesophageal reflux disease (OR 4.1, 95% CI 1.2-12.9; p=0.02) and in females with diabetes mellitus (OR 5.3, 95% CI 1.0-27.4; p=0.01). CONCLUSIONS: Around 10% of respiratory asymptomatic individuals enrolled in our lung ageing programme show ILAs. Increased serum concentrations of pro-inflammatory molecules and MMPs are associated with ILAs.


Subject(s)
Lung Diseases, Interstitial , Female , Humans , Lung/diagnostic imaging , Male , Matrix Metalloproteinase 7 , Mucin-5B , Risk Factors
7.
Oxid Med Cell Longev ; 2020: 3176375, 2020.
Article in English | MEDLINE | ID: mdl-33149807

ABSTRACT

A hypoxic microenvironment is a hallmark in different types of tumors; this phenomenon participates in a metabolic alteration that confers resistance to treatments. Because of this, it was proposed that a combination of 2-methoxyestradiol (2-ME) and sodium dichloroacetate (DCA) could reduce this alteration, preventing proliferation through the reactivation of aerobic metabolism in lung adenocarcinoma cell line (A549). A549 cells were cultured in a hypoxic chamber at 1% O2 for 72 hours to determine the effect of this combination on growth, migration, and expression of hypoxia-inducible factors (HIFs) by immunofluorescence. The effect in the metabolism was evaluated by the determination of glucose/glutamine consumption and the lactate/glutamate production. The treatment of 2-ME (10 µM) in combination with DCA (40 mM) under hypoxic conditions showed an inhibitory effect on growth and migration. Notably, this reduction could be attributed to 2-ME, while DCA had a predominant effect on metabolic activity. Moreover, this combination decreases the signaling of HIF-3α and partially HIF-1α but not HIF-2α. The results of this study highlight the antitumor activity of the combination of 2-ME 10 µl/DCA 40 mM, even in hypoxic conditions.


Subject(s)
2-Methoxyestradiol/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Dichloroacetic Acid/therapeutic use , Lung Neoplasms/drug therapy , Tumor Hypoxia , Tumor Microenvironment , 2-Methoxyestradiol/pharmacology , A549 Cells , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Dichloroacetic Acid/pharmacology , Glucose/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Glycolysis/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactic Acid/metabolism , Lung Neoplasms/pathology , Repressor Proteins/metabolism , Signal Transduction/drug effects , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects , Wound Healing/drug effects
8.
Int J Med Sci ; 17(15): 2357-2361, 2020.
Article in English | MEDLINE | ID: mdl-32922201

ABSTRACT

Idiopathic pulmonary fibrosis is a chronic and progressive disease of unknown cause. It is characterized by the aberrant activation of the bronchioalveolar epithelium, the formation of fibroblast foci and the excessive production extracellular matrix. The cellular and molecular mechanisms that contribute to the pathobiology of the disease are unclear. The CX3CL1-CX3CR1 axis regulates cellular responses that are known to be relevant in IPF, such as proliferation and collagen production. In this study, we characterize for the first time the expression of CX3CL1 and its receptor in lung tissue from patients with IPF; and its effect on collagen production in IPF fibroblasts. We found that CX3CL1-CX3CR1 axis has a modified expression in the lung tissue, importantly this axis is expressed on fibroblasts, and CX3CL1 decreased the collagen production in pulmonary fibroblasts derived from IPF patients.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/metabolism , Collagen/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung/pathology , CX3C Chemokine Receptor 1/analysis , Cell Line , Cell Proliferation , Chemokine CX3CL1/analysis , Collagen/analysis , Extracellular Matrix/pathology , Fibroblasts/metabolism , Humans , Immunohistochemistry , Lung/cytology , Primary Cell Culture , Signal Transduction
9.
BMC Pulm Med ; 20(1): 164, 2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32517728

ABSTRACT

BACKGROUND: Several lung structural and functional abnormalities may occur associated with aging, including emphysema. In this study, we evaluated the frequency and risk factors associated with emphysema in respiratory asymptomatic individuals enrolled in our Lung Aging Program. From a cohort of 687 subjects, we found by high-resolution computed tomography (HRCT) 29 individuals (4%) with emphysematous changes that were compared with 87 controls (3:1) randomly selected from the same cohort. METHODS: This was a transversal, observational, case-control study where we examined demographics and functional characteristics, as well as telomere length and serum Klotho concentration, two conditions that have been associated with aging and some aging-associated diseases including emphysema. RESULTS: Individuals with subclinical pulmonary emphysema were older (72 ± 9 versus 67 ± 6 years), and primarily smoker males with low body mass index. Despite that they were asymptomatic, two of them exhibited a decrease of forced expiratory volume in 1 s (FEV1), with a lower FEV1/FVC suggesting airway obstruction. Cigarette smoking (OR = 5.43, CI95% 1.8-16.7), family history of lung disease (OR = 4.32, CI95% 1.0-19.0) and lower body mass index (OR 7.22, CI95% 1.2-3.5) were risk factors for the development of lung emphysematous changes. No association was found with telomere length and Klotho serum concentration. CONCLUSION: Our findings reveal that a small but important percentage of older people without respiratory symptoms, present pulmonary emphysema and indicate that smoking exposure and genetic background may contribute to etiological factors.


Subject(s)
Aging , Lung/physiopathology , Pulmonary Emphysema/diagnosis , Pulmonary Emphysema/physiopathology , Aged , Aged, 80 and over , Case-Control Studies , Cigarette Smoking/adverse effects , Female , Glucuronidase/blood , Humans , Klotho Proteins , Logistic Models , Male , Middle Aged , Multivariate Analysis , Pulmonary Emphysema/blood , Respiratory Function Tests , Risk Factors , Telomere/physiology , Tomography, X-Ray Computed
10.
Aging (Albany NY) ; 10(8): 2098-2112, 2018 Aug 27.
Article in English | MEDLINE | ID: mdl-30147026

ABSTRACT

Aging is the main risk factor for the development of idiopathic pulmonary fibrosis (IPF), a progressive and usually lethal lung disorder. Although the pathogenic mechanisms are uncertain, endoplasmic reticulum (ER) stress and impaired proteostasis that have been linked with aging are strongly associated with the pathogenesis of IPF. Using the Atg4b-deficient mice as a model, that partially reproduces the autophagy deficient conditions reported in aging and IPF lungs, we show for the first time how autophagy impairment and ER stress induction, contribute simultaneously to development of lung fibrosis in vivo. Increased expression of ER stress markers, inflammation and apoptosis of alveolar epithelial cells were observed in Atg4b-deficient mice compared to WT mice, when treated with the ER stress inducer tunicamycin. After tunicamycin treatment, Atg4b null lungs showed accumulation of its substrate LC3-I, demonstrating that these mice failed to induce autophagy despite the ER stress conditions. We also showed that compromised autophagy in lungs from Atg4b null mice is associated with exacerbated lung damage, epithelial apoptosis and the development of lung fibrosis at 21 days after tunicamycin treatment. Our findings indicate that ATG4B protein and autophagy are essential to mitigate ER stress and to prevent tunicamycin-induced epithelial apoptosis and lung fibrosis.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/drug effects , Cysteine Endopeptidases/metabolism , Endoplasmic Reticulum/physiology , Epithelial Cells/physiology , Animals , Autophagy/physiology , Autophagy-Related Proteins/genetics , Cell Line , Cysteine Endopeptidases/genetics , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Lung/cytology , Respiratory Mucosa/cytology , Tunicamycin/pharmacology
11.
Am J Respir Cell Mol Biol ; 59(1): 77-86, 2018 07.
Article in English | MEDLINE | ID: mdl-29373068

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive aging-associated disease of unknown etiology. A growing body of evidence indicates that aberrant activated alveolar epithelial cells induce the expansion and activation of the fibroblast population, leading to the destruction of the lung architecture. Some matrix metalloproteinases (MMPs) are upregulated in IPF, indicating that they may be important in the pathogenesis and/or progression of IPF. In the present study, we examined the expression of MMP28 in this disease and evaluated its functional effects in two alveolar epithelial cell lines and in human primary bronchial epithelial cells. We found that the enzyme is expressed in bronchial (apical and cytoplasmic localization) and alveolar (cytoplasmic and nuclear localization) epithelial cells in two different groups of patients with IPF. In vitro MMP28 epithelial silencing decreased the proliferation rate and delayed wound closing, whereas overexpression showed opposite effects, protecting from apoptosis and enhanced epithelial-mesenchymal transition. Our findings demonstrate that MMP28 is upregulated in epithelial cells from IPF lungs, where it may play a role in increasing the proliferative and migratory phenotype in a catalysis-dependent manner.


Subject(s)
Cell Nucleus/metabolism , Epithelium/metabolism , Idiopathic Pulmonary Fibrosis/enzymology , Idiopathic Pulmonary Fibrosis/genetics , Matrix Metalloproteinases, Secreted/genetics , Pulmonary Alveoli/pathology , Up-Regulation/genetics , A549 Cells , Animals , Apoptosis , Biocatalysis , Cell Movement , Cell Proliferation , Cytoprotection , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Epithelium/pathology , Gene Silencing , Humans , Matrix Metalloproteinases, Secreted/metabolism , Protein Transport , Rats
12.
Autophagy ; 11(4): 670-84, 2015 Apr 03.
Article in English | MEDLINE | ID: mdl-25906080

ABSTRACT

Autophagy is a critical cellular homeostatic process that controls the turnover of damaged organelles and proteins. Impaired autophagic activity is involved in a number of diseases, including idiopathic pulmonary fibrosis suggesting that altered autophagy may contribute to fibrogenesis. However, the specific role of autophagy in lung fibrosis is still undefined. In this study, we show for the first time, how autophagy disruption contributes to bleomycin-induced lung fibrosis in vivo using an Atg4b-deficient mouse as a model. Atg4b-deficient mice displayed a significantly higher inflammatory response at 7 d after bleomycin treatment associated with increased neutrophilic infiltration and significant alterations in proinflammatory cytokines. Likewise, we found that Atg4b disruption resulted in augmented apoptosis affecting predominantly alveolar and bronchiolar epithelial cells. At 28 d post-bleomycin instillation Atg4b-deficient mice exhibited more extensive and severe fibrosis with increased collagen accumulation and deregulated extracellular matrix-related gene expression. Together, our findings indicate that the ATG4B protease and autophagy play a crucial role protecting epithelial cells against bleomycin-induced stress and apoptosis, and in the regulation of the inflammatory and fibrotic responses.


Subject(s)
Autophagy/drug effects , Bleomycin/pharmacology , Cysteine Endopeptidases/metabolism , Homeostasis/drug effects , Idiopathic Pulmonary Fibrosis/metabolism , Animals , Apoptosis/genetics , Autophagy/physiology , Autophagy-Related Proteins , Cysteine Endopeptidases/genetics , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Gene Expression/drug effects , Idiopathic Pulmonary Fibrosis/chemically induced , Mice, Knockout
13.
Respir Res ; 15: 74, 2014 Jun 30.
Article in English | MEDLINE | ID: mdl-24980707

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) and C-reactive protein (CRP) are involved in chronic obstructive pulmonary disease (COPD) pathogenesis. The aim of the present work was to determine plasma concentrations of MMPs and CRP in COPD associated to biomass combustion exposure (BE) and tobacco smoking (TS). METHODS: Pulmonary function tests, plasma levels of MMP-1, MMP-7, MMP-9, MMP-9/TIMP-1 and CRP were measured in COPD associated to BE (n = 40) and TS (n =40) patients, and healthy non-smoking (NS) healthy women (controls, n = 40). RESULTS: Plasma levels of MMP-1, MMP-7, MMP-9, and MMP-9/TIMP-1 and CRP were higher in BE and TS than in the NS healthy women (p <0.01). An inverse correlation between MMP-1, MMP-7, MMP-9, MMP-9/TIMP-1 and CRP plasma concentrations and FEV1 was observed. CONCLUSIONS: Increase of MMPs and CRP plasma concentrations in BE suggests a systemic inflammatory phenomenon similar to that observed in COPD associated to tobacco smoking, which may also play a role in COPD pathogenesis.


Subject(s)
C-Reactive Protein/metabolism , Forced Expiratory Volume/physiology , Matrix Metalloproteinase 1/blood , Matrix Metalloproteinase 7/blood , Matrix Metalloproteinase 9/blood , Pulmonary Disease, Chronic Obstructive/blood , Smoking/blood , Aged , Aged, 80 and over , Biomarkers/blood , Biomass , Female , Fuel Oils/adverse effects , Humans , Middle Aged , Pulmonary Disease, Chronic Obstructive/diagnosis , Pulmonary Disease, Chronic Obstructive/epidemiology , Smoke/adverse effects , Smoking/adverse effects , Smoking/epidemiology
14.
J Biol Chem ; 288(36): 25964-25975, 2013 Sep 06.
Article in English | MEDLINE | ID: mdl-23902766

ABSTRACT

Idiopathic pulmonary fibrosis is a devastating lung disorder of unknown etiology. Although its pathogenesis is unclear, considerable evidence supports an important role of aberrantly activated alveolar epithelial cells (AECs), which produce a large variety of mediators, including several matrix metalloproteases (MMPs), which participate in fibroblast activation and lung remodeling. MMP-1 has been shown to be highly expressed in AECs from idiopathic pulmonary fibrosis lungs although its role is unknown. In this study, we explored the role of MMP-1 in several AECs functions. Mouse lung epithelial cells (MLE12) transfected with human Mmp-1 showed significantly increased cell growth and proliferation at 36 and 48 h of culture (p < 0.01). Also, MMP-1 promoted MLE12 cell migration through collagen I, accelerated wound closing, and protected cells from staurosporine- and bleomycin-induced apoptosis compared with mock cells (p < 0.01). MLE12 cells expressing human MMP-1 showed a significant repression of oxygen consumption ratio compared with the cells with the empty vector. As under hypoxic conditions hypoxia-inducible factor-1α (HIF-1α) mediates a transition from oxidative to glycolytic metabolism, we analyzed activation of HIF-1α. Ηigher activation of this factor was detected in MMP-1-transfected cells under normoxia and hypoxia. Likewise, a significant decrease of both total and mitochondrial reactive oxygen species was observed in MMP-1-transfected cells. Paralleling these findings, attenuation of MMP-1 expression by shRNA in A549 (human) AECs markedly reduced proliferation and migration (p < 0.01) and increased the oxygen consumption ratio. These findings indicate that epithelial expression of MMP-1 inhibits mitochondrial function, increases HIF-1α expression, decreases reactive oxygen species production, and contributes to a proliferative, migratory, and anti-apoptotic AEC phenotype.


Subject(s)
Apoptosis/physiology , Cell Movement/physiology , Cell Proliferation , Epithelial Cells/enzymology , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 1/biosynthesis , Mitochondria/metabolism , Oxygen Consumption/physiology , Pulmonary Alveoli/enzymology , Respiratory Mucosa/enzymology , Animals , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Bleomycin/pharmacology , Cell Line , Cell Movement/drug effects , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 13/genetics , Mice , Mitochondria/genetics , Oxygen Consumption/drug effects , Pulmonary Alveoli/cytology , Respiratory Mucosa/cytology , Staurosporine/pharmacology
15.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L295-303, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22707614

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disease of unknown etiology. A conspicuous feature is the formation and persistence of fibroblastic/myofibroblastic foci throughout the lung parenchyma. Mechanisms remain unknown, but data indicate that fibroblasts acquire an antiapoptotic phenotype. We hypothesized that transcriptional silencing of proapoptotic genes may be implicated, and accordingly we evaluated the epigenetic regulation of p14(ARF). The expression of p14(ARF) was analyzed by RT-PCR in IPF (n = 8) and normal derived fibroblasts (n = 4) before and after treatment with 5-aza-2'-deoxycytidine (5-aza) and trichostatin A (TSA). p14(ARF) gene promoter methylation was determined by methylation-specific PCR (MS-PCR) and by DNA digestion with endonuclease McrBc, which cleaves 50% of methylated CpG. Apoptosis was evaluated by Annexin-V and nuclear staining. p14(ARF) expression was significantly decreased in four of the eight IPF fibroblasts lines, which was restored after 5-aza treatment. No changes were found with TSA. MS-PCR of bisulfite-treated genomic DNA showed a correlation between the reduced expression of p14(ARF) and the presence of hypermethylated promoter. No amplification was observed in the DNA treated with the McrBc enzyme, corroborating promoter hypermethylation. p14(ARF)-hypermethylated IPF fibroblasts were significantly more resistant to staurosporine-and S-nitrosoglutathione-induced apoptosis compared with normal and nonmethylated IPF fibroblasts (P < 0.01) and showed reduced levels of p53. Resistance to apoptosis was provoked in fibroblasts when p14(ARF) expression was inhibited by siRNA (P < 0.05). These findings demonstrate that many IPF fibroblasts have reduced expression of the proapoptotic p14(ARF) attributable to promoter hypermethylation and indicate that epigenetic mechanisms may underlie their resistance to apoptosis.


Subject(s)
DNA Methylation/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Silencing , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Tumor Suppressor Protein p14ARF/genetics , Apoptosis/genetics , Blotting, Western , Cell Line , Humans , Middle Aged , Promoter Regions, Genetic/genetics , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism
16.
PLoS Med ; 2(9): e251, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16128620

ABSTRACT

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a progressive and lethal disorder characterized by fibroproliferation and excessive accumulation of extracellular matrix in the lung. METHODS AND FINDINGS: Using oligonucleotide arrays, we identified osteopontin as one of the genes that significantly distinguishes IPF from normal lungs. Osteopontin was localized to alveolar epithelial cells in IPF lungs and was also significantly elevated in bronchoalveolar lavage from IPF patients. To study the fibrosis-relevant effects of osteopontin we stimulated primary human lung fibroblasts and alveolar epithelial cells (A549) with recombinant osteopontin. Osteopontin induced a significant increase of migration and proliferation in both fibroblasts and epithelial cells. Epithelial growth was inhibited by the pentapeptide Gly-Arg-Gly-Asp-Ser (GRGDS) and antibody to CD44, while fibroproliferation was inhibited by GRGDS and antibody to alphavbeta3 integrin. Fibroblast and epithelial cell migration were inhibited by GRGDS, anti-CD44, and anti-alphavbeta3. In fibroblasts, osteopontin up-regulated tissue inhibitor of metalloprotease-1 and type I collagen, and down-regulated matrix metalloprotease-1 (MMP-1) expression, while in A549 cells it caused up-regulation of MMP-7. In human IPF lungs, osteopontin colocalized with MMP-7 in alveolar epithelial cells, and application of weakest link statistical models to microarray data suggested a significant interaction between osteopontin and MMP-7. CONCLUSIONS: Our results provide a potential mechanism by which osteopontin secreted from the alveolar epithelium may exert a profibrotic effect in IPF lungs and highlight osteopontin as a potential target for therapeutic intervention in this incurable disease.


Subject(s)
Lung/metabolism , Pulmonary Fibrosis/metabolism , Sialoglycoproteins/metabolism , Bronchoalveolar Lavage Fluid/chemistry , Cell Movement , Cell Proliferation , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Extracellular Matrix/metabolism , Female , Gene Expression Profiling , Humans , Male , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 7/metabolism , Middle Aged , Molecular Sequence Data , Osteopontin , Pulmonary Fibrosis/pathology , Recombinant Proteins/pharmacology , Sialoglycoproteins/pharmacology , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Up-Regulation
17.
Ginecol. & obstet ; 44(1): 45-53, abr.- 1998. tab, graf
Article in Spanish | LILACS, LIPECS | ID: lil-270743

ABSTRACT

Objetivo: Describir la curva de alerta en parturientas de la altura. Diseño: Se realiza un estudio descriptivo, explicativo y prospectivo de corte transversal de enero a mayo de 1997. Pacientes: 400 parturientas nulíparas y multíparas a término, con presentación cefálica atendidas en el Hospital Víctor Ramos Guardia de Huaraz, seleccionadas con criterios de inclusión y que cursaron con trabajo de parto normal sin uso medicamentoso pre e intra parto. Fueron clasificadas en cuatro grupos de acuerdo al CLAP, cuidando de trazar la curva de alerta a partir de 4 cm de dilatación . El análisis estadístico, procedimiento de datos y la obtención de resultados con programas de EPIINFO 5, calculando el percentil 10 y construyéndose la curva a la izquierda con respecto a la curva del CLAP y aún más a la izquierda si comparamos con la curva a nivel del mar (HAMA), atribuyéndose este comportamiento al estado nutricional y al umbral doloroso de mujeres de la altura. Conclusión: La curva de alerta en parturientas de la altura presenta tiempos menores de dilatación. Palabras clave: Partograma, gestantes en altura.


Subject(s)
Humans , Female , Patients , Pregnancy , Altitude , Natural Childbirth , Cross-Sectional Studies , Prospective Studies , Epidemiology, Descriptive
SELECTION OF CITATIONS
SEARCH DETAIL
...