Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Sci Rep ; 13(1): 15073, 2023 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-37699920

RESUMO

In the repair of injury, some transforming growth factor-[Formula: see text]s (TGF-[Formula: see text]s) and platelet-derived growth factors (PDGFs) bind to fibroblast receptors as ligands and cause the differentiation of fibroblasts into myofibroblasts. When the injury repair is repeated, the myofibroblasts proliferate excessively, forming fibrotic tissue. We goal to control myofibroblasts proliferation and apoptosis with anti-transforming growth factor-[Formula: see text] (anti-TGF-[Formula: see text]) and anti-platelet-derived growth factor (anti-PDGF) medicines. The novel optimal regulator control problem with two controls (medicines) is proposed to simulate how to the preventing pulmonary fibrosis. Idiopathic pulmonary fibrosis (IPF) consists of restoring a system of cells, protein, and tissue networks with injury and scar. Myofibroblasts proliferation back to its equilibrium position after it has been disturbed by abnormal repair. Thus, the optimal regulator control problem with a parabolic partial differential equation as a constraint, zero flux boundary, and given specific initial conditions, is considered. The myofibroblast diffusion equation stands as a governing dynamic system while the objective function is the summation of myofibroblast, anti-TGF-[Formula: see text] and anti-PDGF medicines for the fixed final time. Here, myofibroblast is a nonlinear state of time while anti-TGF-[Formula: see text] and anti-PDGF are two unknown control functions. In order to solve the corresponding problem a weighted Galerkin method is used. Firstly, we convert the myofibroblast diffusion equation to a system of ordinary differential equations using the Lagrangian interpolation polynomials defined at Gauss-Lobatto integration points. Secondly, by the calculus of variations, the optimal control problem is solved successfully using canonical Hamiltonian and extended Riccati equations. Numerical results are given, and the plots are depicted. Moreover, solutions to the problem in which there is no control are compared. Numerical results show that, over time, the myofibroblast increases and then remains constant when there is no control. In contrast, the current solution decreases and vanishes after 300 days by prescribing controller medicines for anti-TGF-[Formula: see text] and anti-PDGF. The optimal strategy proposed in this paper helps practitioners to reduce myofibroblasts by controlling both anti-TGF-[Formula: see text] and anti-PDGF medicines.


Assuntos
Fibrose Pulmonar Idiopática , Humanos , Fibrose Pulmonar Idiopática/tratamento farmacológico , Fibrose Pulmonar Idiopática/prevenção & controle , Miofibroblastos , Fibroblastos , Apoptose , Fator de Crescimento Derivado de Plaquetas
4.
Int J Mol Sci ; 22(20)2021 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-34681813

RESUMO

Idiopathic pulmonary fibrosis (IPF) is characterized by fibrotic change in alveolar epithelial cells and leads to the irreversible deterioration of pulmonary function. Transforming growth factor-beta 1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) in type 2 lung epithelial cells contributes to excessive collagen deposition and plays an important role in IPF. Atractylodin (ATL) is a kind of herbal medicine that has been proven to protect intestinal inflammation and attenuate acute lung injury. Our study aimed to determine whether EMT played a crucial role in the pathogenesis of pulmonary fibrosis and whether EMT can be utilized as a therapeutic target by ATL treatment to mitigate IPF. To address this topic, we took two steps to investigate: 1. Utilization of anin vitro EMT model by treating alveolar epithelial cells (A549 cells) with TGF-ß1 followed by ATL treatment for elucidating the underlying pathways, including Smad2/3 hyperphosphorylation, mitogen-activated protein kinase (MAPK) pathway overexpression, Snail and Slug upregulation, and loss of E-cadherin. Utilization of an in vivo lung injury model by treating bleomycin on mice followed by ATL treatment to demonstrate the therapeutic effectiveness, such as, less collagen deposition and lower E-cadherin expression. In conclusion, ATL attenuates TGF-ß1-induced EMT in A549 cells and bleomycin-induced pulmonary fibrosis in mice.


Assuntos
Células Epiteliais Alveolares/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Furanos/farmacologia , Fibrose Pulmonar Idiopática/prevenção & controle , Células A549 , Células Epiteliais Alveolares/fisiologia , Animais , Bleomicina/efeitos adversos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Furanos/uso terapêutico , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/genética , Fibrose Pulmonar Idiopática/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/fisiologia
5.
Proc Natl Acad Sci U S A ; 118(35)2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34446559

RESUMO

Perturbation of lung homeostasis is frequently associated with progressive and fatal respiratory diseases, such as pulmonary fibrosis. Leucine-rich repeat kinase 2 (LRRK2) is highly expressed in healthy lungs, but its functions in lung homeostasis and diseases remain elusive. Herein, we showed that LRRK2 expression was clearly reduced in mammalian fibrotic lungs, and LRRK2-deficient mice exhibited aggravated bleomycin-induced pulmonary fibrosis. Furthermore, we demonstrated that in bleomycin-treated mice, LRRK2 expression was dramatically decreased in alveolar type II epithelial (AT2) cells, and its deficiency resulted in profound dysfunction of AT2 cells, characterized by impaired autophagy and accelerated cellular senescence. Additionally, LRRK2-deficient AT2 cells showed a higher capacity of recruiting profibrotic macrophages via the CCL2/CCR2 signaling, leading to extensive macrophage-associated profibrotic responses and progressive pulmonary fibrosis. Taken together, our study demonstrates that LRRK2 plays a crucial role in preventing AT2 cell dysfunction and orchestrating the innate immune responses to protect against pulmonary fibrosis.


Assuntos
Células Epiteliais Alveolares/imunologia , Bleomicina/toxicidade , Fibrose Pulmonar Idiopática/prevenção & controle , Imunidade Inata , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina/fisiologia , Pulmão/imunologia , Macrófagos/imunologia , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Antibióticos Antineoplásicos/toxicidade , Autofagia , Homeostase , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais
6.
Biomed Pharmacother ; 133: 111072, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33378971

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive pulmonary interstitial inflammatory disease of unknown etiology, and is also a sequela in severe patients with the Coronavirus Disease 2019 (COVID-19). Nintedanib and pirfenidone are the only two known drugs which are conditionally recommended for the treatment of IPF by the FDA. However, these drugs pose some adverse side effects such as nausea and diarrhoea during clinical applications. Therefore, it is of great value and significance to identify effective and safe therapeutic drugs to solve the clinical problems associated with intake of western medicine. As a unique medical treatment, Traditional Chinese Medicine (TCM) has gradually exerted its advantages in the treatment of IPF worldwide through a multi-level and multi-target approach. Further, to overcome the current clinical problems of oral and injectable intakes of TCM, pulmonary drug delivery system (PDDS) could be designed to reduce the systemic metabolism and adverse reactions of the drug and to improve the bioavailability of drugs. Through PubMed, Google Scholar, Web of Science, and CNKI, we retrieved articles published in related fields in recent years, and this paper has summarized twenty-seven Chinese compound prescriptions, ten single TCM, and ten active ingredients for effective prevention and treatment of IPF. We also introduce three kinds of inhaling PDDS, which supports further research of TCM combined with PDDS to treat IPF.


Assuntos
COVID-19/complicações , Medicamentos de Ervas Chinesas/uso terapêutico , Fibrose Pulmonar Idiopática/tratamento farmacológico , Medicina Tradicional Chinesa/métodos , Fitoterapia , Composição de Medicamentos , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/química , História do Século XV , História do Século XVI , História do Século XVII , História do Século XVIII , História do Século XIX , História do Século XX , História Antiga , História Medieval , Humanos , Fibrose Pulmonar Idiopática/etiologia , Fibrose Pulmonar Idiopática/prevenção & controle , Medicina Tradicional Chinesa/história , Nebulizadores e Vaporizadores , Terapia Respiratória
7.
Respir Med ; 176: 106259, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33276250

RESUMO

BACKGROUND AND OBJECTIVE: Idiopathic pulmonary fibrosis (IPF) is a progressive and irreversible interstitial lung disease with poor prognosis despite the recent availability of two antifibrotic drugs. Patients are more susceptible to cardiovascular comorbidities. In this study, we aimed to determine the impact of concomitant cardiovascular drugs on disease progression and survival in a modern IPF cohort. METHODS: The database of a tertiary referral centre for interstitial lung diseases in Belgium was reviewed for statin, antiaggregant, anticoagulant and metformin therapy. For a study period of four years, we noted both FVC% and DLCO% trajectories along with survival as outcome measurements. RESULTS: 323 patients were included of which 45% had at least one cardiovascular comorbidity. 274 (86%) patients received antifibrotic therapy. Statin users (n = 171) displayed significantly slower annual FVC% (difference 2.9%, CI 1.6-4.4, p < 0.001) and DLCO% decline (difference 1.3%, CI 0.24-2.3, p = 0.013). Results for antiaggregant therapy (n = 152) were inconclusive: we found a trend for slower FVC decline (p = 0.098) and a numerically decrease in survival rates (HR 1.63, p = 0.074) in a multivariate Cox analysis. Anticoagulant use (n = 49) showed a trend towards worse DLCO decline (difference -1.3%, CI -2.6 - 0.02, p = 0.055).r Metformin (n = 28) therapy did not affect IPF progression in terms of pulmonary function test evolution or survival. CONCLUSION: This retrospective study demonstrated a benefit of statin therapy on IPF progression. Our observations emphasize the need for large clinical trials analysing the effect of statins, as well as other cardiovascular drugs, in the management of IPF patients.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Fibrose Pulmonar Idiopática/etiologia , Fibrose Pulmonar Idiopática/prevenção & controle , Idoso , Doenças Cardiovasculares/tratamento farmacológico , Doenças Cardiovasculares/epidemiologia , Comorbidade , Progressão da Doença , Feminino , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Fibrose Pulmonar Idiopática/epidemiologia , Fibrose Pulmonar Idiopática/fisiopatologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Taxa de Sobrevida , Capacidade Vital
8.
Respir Res ; 21(1): 290, 2020 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-33138822

RESUMO

BACKGROUND: The poor understanding of pathogenesis in idiopathic pulmonary fibrosis (IPF) impaired development of effective therapeutic strategies. The aim of the current study is to investigate the roles of long non-coding RNA H19 (lncRNA H19) in the pulmonary inflammation and fibrosis of IPF. METHODS: Bleomycin was used to induce pulmonary inflammation and fibrosis in mice. The mRNAs and proteins expression in lung tissues was determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western blot. H19 knockout (H19-/-) mice were generated by CRISPR/Cas9. RESULTS: The expression of H19 mRNA was up-regulated in fibrotic lungs patients with IPF as well as in lungs tissues that obtained from bleomycin-treated mice. H19-/- mice suppressed bleomycin-mediated pulmonary inflammation and inhibited the Il6/Stat3 signaling. H19 deficiency ameliorated bleomycin-induced pulmonary fibrosis and repressed the activation of TGF-ß/Smad and S1pr2/Sphk2 in the lungs of bleomycin-treated mice. CONCLUSIONS: Our data suggests that H19 is a profibrotic lncRNA and a potential therapeutic target for IPF.


Assuntos
Fibrose Pulmonar Idiopática/prevenção & controle , Pulmão/metabolismo , Pneumonia/prevenção & controle , RNA Longo não Codificante/metabolismo , Animais , Bleomicina , Estudos de Casos e Controles , Proliferação de Células , Bases de Dados Genéticas , Modelos Animais de Doenças , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Interleucina-6/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Pneumonia/induzido quimicamente , Pneumonia/metabolismo , Pneumonia/patologia , RNA Longo não Codificante/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteínas Smad Reguladas por Receptor/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
10.
Lung India ; 37(4): [359-378], July 1, 2020.
Artigo em Inglês | BIGG | ID: biblio-1117197

RESUMO

Interstitial lung disease (ILD) is a complex and heterogeneous group of acute and chronic lung diseases of several known and unknown causes. While clinical practice guidelines (CPG) for idiopathic pulmonary fibrosis (IPF) have been recently updated, CPG for ILD other than IPF are needed. Methods: A working group of multidisciplinary clinicians familiar with clinical management of ILD (pulmonologists, radiologist, pathologist, and rheumatologist) and three epidemiologists selected by the leaderships of Indian Chest Society and National College of Chest Physicians, India, posed questions to address the clinically relevant situation. A systematic search was performed on PubMed, Embase, and Cochrane databases. A modified GRADE approach was used to grade the evidence. The working group discussed the evidence and reached a consensus of opinions for each question following face-to-face discussions. Results: Statements have been made for each specific question and the grade of evidence has been provided after performing a systematic review of literature. For most of the questions addressed, the available evidence was insufficient and of low to very low quality. The consensus of the opinions of the working group has been presented as statements for the questions and not as an evidence-based CPG for the management of ILD. Conclusion: This document provides the guidelines made by consensus of opinions among experts following discussion of systematic review of evidence pertaining to the specific questions for management of ILD other than IPF. It is hoped that this document will help the clinician understand the accumulated evidence and help better management of idiopathic and nonidiopathic interstitial pneumonias.


Assuntos
Humanos , Doenças Pulmonares Intersticiais/diagnóstico , Doenças Pulmonares Intersticiais/prevenção & controle , Doenças Pulmonares Intersticiais/terapia , Fibrose Pulmonar Idiopática/complicações , Fibrose Pulmonar Idiopática/prevenção & controle
11.
Immunohorizons ; 4(6): 352-362, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32576593

RESUMO

Pulmonary fibrosis is a chronic and generally fatal disorder characterized by progressive formation of scar-like tissue in the lungs. Sialic acids are often found as the terminal sugar on extracellular glycoconjugates such as protein glycosylations. Sialidases, also known as neuraminidases, desialylate glycoconjugates. Serum amyloid P (SAP), a pentameric serum glycoprotein that has two sialic acids on each polypeptide, inhibits the differentiation of monocytes into fibrocytes and promotes human PBMCs to accumulate high extracellular levels of IL-10. When SAP is desialylated with sialidase, the effects of SAP on fibrocyte differentiation and IL-10 accumulation are strongly inhibited. Intriguingly, in patients with pulmonary fibrosis, there are increased levels of sialidase activity in the bronchoalveolar lavage fluid, increased levels of sialidases in the lungs, and decreased levels of SAP in the sera. To elucidate the role of SAP desialylation in idiopathic pulmonary fibrosis (IPF) pathogenesis, we purified SAP from the serum of IPF patients and healthy controls and measured the extent of sialylation and bioactivity of the purified SAP. We find that some IPF patients have abnormally high levels of the sialidase NEU3 in their sera and that the SAP in the sera of IPF patients has an abnormally high extent of desialylation and an abnormally low ability to inhibit fibrocyte differentiation and induce extracellular IL-10 accumulation by PBMC. These results suggest that SAP desialylation may play a role in IPF pathogenesis and that inhibiting NEU3 could be a potential therapeutic target for IPF.


Assuntos
Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/prevenção & controle , Leucócitos Mononucleares/metabolismo , Neuraminidase/metabolismo , Componente Amiloide P Sérico/metabolismo , Idoso , Líquido da Lavagem Broncoalveolar , Estudos de Casos e Controles , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/fisiopatologia , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Neuraminidase/antagonistas & inibidores , Componente Amiloide P Sérico/farmacologia
12.
FASEB J ; 34(8): 9884-9898, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32596871

RESUMO

Idiopathic pulmonary fibrosis (IPF) is characterized by altered epithelial cell phenotypes, which are associated with myofibroblast accumulation in the lung. Atypical alveolar epithelial cells in IPF express molecular markers of airway epithelium. Polymorphisms within and around Toll interacting protein (TOLLIP) are associated with the susceptibility to IPF and mortality. However, the functional role of TOLLIP in IPF is unknown. Using lung tissues from IPF and control subjects, we showed that expression of TOLLIP gene in the lung parenchyma is globally lower in IPF compared to controls. Lung cells expressing significant levels of TOLLIP include macrophages, alveolar type II, and basal cells. TOLLIP protein expression is lower in the parenchyma of IPF lungs but is expressed in the atypical epithelial cells of the distal fibrotic regions. Using overexpression and silencing approaches, we demonstrate that TOLLIP protects cells from bleomycin-induced apoptosis using primary bronchial epithelial cells and BEAS-2B cells. The protective effects are mediated by reducing mitochondrial reactive oxygen species (ROS) levels and upregulating autophagy. Therefore, global downregulation of the TOLLIP gene in IPF lungs may predispose injured lung epithelial cells to apoptosis and to the development of IPF.


Assuntos
Apoptose , Bleomicina/efeitos adversos , Brônquios/citologia , Células Epiteliais/citologia , Fibrose Pulmonar Idiopática/prevenção & controle , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/metabolismo , Substâncias Protetoras , Antibióticos Antineoplásicos/efeitos adversos , Autofagia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
13.
Respir Res ; 21(1): 132, 2020 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-32471489

RESUMO

BACKGROUND: Chronic tissue injury was shown to induce progressive scarring in fibrotic diseases such as idiopathic pulmonary fibrosis (IPF), while an array of repair/regeneration and stress responses come to equilibrium to determine the outcome of injury at the organ level. In the lung, type I alveolar epithelial (ATI) cells constitute the epithelial barrier, while type II alveolar epithelial (ATII) cells play a pivotal role in regenerating the injured distal lungs. It had been demonstrated that eukaryotic cells possess repair machinery that can quickly patch the damaged plasma membrane after injury, and our previous studies discovered the membrane-mending role of Tripartite motif containing 72 (TRIM72) that expresses in a limited number of tissues including the lung. Nevertheless, the role of alveolar epithelial cell (AEC) repair in the pathogenesis of IPF has not been examined yet. METHOD: In this study, we tested the specific roles of TRIM72 in the repair of ATII cells and the development of lung fibrosis. The role of membrane repair was accessed by saponin assay on isolated primary ATII cells and rat ATII cell line. The anti-fibrotic potential of TRIM72 was tested with bleomycin-treated transgenic mice. RESULTS: We showed that TRIM72 was upregulated following various injuries and in human IPF lungs. However, TRIM72 expression in ATII cells of the IPF lungs had aberrant subcellular localization. In vitro studies showed that TRIM72 repairs membrane injury of immortalized and primary ATIIs, leading to inhibition of stress-induced p53 activation and reduction in cell apoptosis. In vivo studies demonstrated that TRIM72 protects the integrity of the alveolar epithelial layer and reduces lung fibrosis. CONCLUSION: Our results suggest that TRIM72 protects injured lungs and ameliorates fibrosis through promoting post-injury repair of AECs.


Assuntos
Células Epiteliais Alveolares/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/prevenção & controle , Pulmão/metabolismo , Proteínas com Motivo Tripartido/biossíntese , Células Epiteliais Alveolares/efeitos dos fármacos , Animais , Bleomicina/toxicidade , Feminino , Células HEK293 , Humanos , Fibrose Pulmonar Idiopática/induzido quimicamente , Pulmão/efeitos dos fármacos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Proteínas Recombinantes/biossíntese
14.
Surg Today ; 50(8): 905-911, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32144483

RESUMO

PURPOSE: Lung cancer in patients with idiopathic pulmonary fibrosis (IPF) is associated with a poor prognosis and postoperative acute exacerbation (AE) of IPF is a fatal complication. Studies have shown that perioperative pirfenidone treatment (PPT) may reduce the incidence of AE-IPF. We evaluated the efficacy of PPT in preventing AE-IPF and improving overall survival. METHODS: The subjects of this study were 56 patients with IPF who underwent resection of lung cancer in our hospital between January, 2011 and September, 2016. Pirfenidone was administered to patients from 4 weeks before the operation and continued for longer periods. Thirty-six patients received PPT and their outcome was compared with that of the other 20 patients who did not. RESULTS: There were no differences in age, gender, smoking history, respiratory function, or surgical procedures between the groups. AE-IPF developed in three patients (8%) in the PPT group and four (20%) patients in the non-PPT group, without a significant difference between the groups. The interval was significantly longer in the PPT group (p = 0.03). PPT reduced postoperative mortality significantly (p = 0.04). CONCLUSIONS: Although perioperative pirfenidone treatment did not obviously prevent postoperative AE-IPF, it may reduce the mortality of lung cancer patients with IPF.


Assuntos
Fibrose Pulmonar Idiopática/prevenção & controle , Neoplasias Pulmonares/cirurgia , Resultados Negativos , Assistência Perioperatória , Complicações Pós-Operatórias/prevenção & controle , Piridonas/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Progressão da Doença , Feminino , Seguimentos , Humanos , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Seleção de Pacientes , Prognóstico , Taxa de Sobrevida
15.
Cell Death Dis ; 11(2): 136, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-32075954

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a group of chronic interstitial pulmonary diseases characterized by myofibroblast proliferation and extracellular matrix deposition with limited treatment options. Based on our previous observation, we hypothesized microcystin-leucine arginine (LR), an environmental cyanobacterial toxin, could potentially suppress pulmonary fibrosis. In this study, we first demonstrated that chronic exposure of microcystin-LR by oral for weeks indeed attenuated the pulmonary fibrosis both on bleomycin-induced rat and fluorescein isothiocyanate-induced mouse models. Our data further indicated that treatment with microcystin-LR substantially reduced TGF-ß1/Smad signaling in rat pulmonary tissues. The experiments in vitro found that microcystin-LR was capable of blocking epithelial-mesenchymal transition (EMT) and fibroblast-myofibroblast transition (FMT) through suppressing the differentiation of CD206+ macrophages. Mechanically, microcystin-LR was found to bind to glucose-regulated protein 78 kDa (GRP78) and suppress endoplasmic reticulum unfolded protein response (UPRER) signaling pathways. These events led to the modulation of M2 polarization of macrophages, which eventually contributed to the alleviation of pulmonary fibrosis. Our results revealed a novel mechanism that may account for therapeutic effect of microcystin-LR on IPF.


Assuntos
Fibroblastos/efeitos dos fármacos , Fibrose Pulmonar Idiopática/prevenção & controle , Lectinas Tipo C/metabolismo , Pulmão/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Lectinas de Ligação a Manose/metabolismo , Toxinas Marinhas/farmacologia , Microcistinas/farmacologia , Receptores de Superfície Celular/metabolismo , Células A549 , Animais , Modelos Animais de Doenças , Chaperona BiP do Retículo Endoplasmático , Fibroblastos/metabolismo , Fibroblastos/patologia , Proteínas de Choque Térmico/metabolismo , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Receptor de Manose , Camundongos , Camundongos Endogâmicos C57BL , Células NIH 3T3 , Fenótipo , Proteína Fosfatase 2/metabolismo , Células RAW 264.7 , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
16.
Curr Opin Allergy Clin Immunol ; 20(2): 103-111, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31895128

RESUMO

PURPOSE OF REVIEW: A recent meta-analysis of data from international case-control studies reports a population attributable fraction of 16% for occupational factors in the cause of idiopathic pulmonary fibrosis (IPF). Smoking, genetic factors and other prevalent diseases only partly explain IPF, and so this review aims to summarize recent progress in establishing which occupational exposures are important in cause. RECENT FINDINGS: IPF is a rare disease, although it is the commonest idiopathic interstitial pneumonia. Epidemiological study suggests that incidence of IPF is increasing, particularly in older men. There are significant associations with IPF and occupational exposures to organic dust, including livestock, birds and animal feed, metal dust, wood dust and silica/minerals. Estimates of effect vary between studies, and are influenced by the distribution of employment, study design and case definition. Inhalation of asbestos fibres is a known cause of usual interstitial pneumonia (as seen histologically in IPF), though there are significant linear relationships between asbestos consumption, and mortality from both IPF and mesothelioma, leading to the hypothesis that low-level asbestos exposure may cause IPF. SUMMARY: Research must focus on exposure-response relationships between asbestos and other occupational inhaled hazards, and IPF. Funding bodies and policy makers should acknowledge the significant occupational burden on IPF.


Assuntos
Efeitos Psicossociais da Doença , Fibrose Pulmonar Idiopática/epidemiologia , Doenças Profissionais/epidemiologia , Exposição Ocupacional/efeitos adversos , Poeira , Humanos , Fibrose Pulmonar Idiopática/etiologia , Fibrose Pulmonar Idiopática/prevenção & controle , Incidência , Exposição por Inalação/efeitos adversos , Metanálise como Assunto , Doenças Profissionais/etiologia , Doenças Profissionais/prevenção & controle , Exposição Ocupacional/prevenção & controle , Prevalência , Fatores de Risco
17.
Tuberk Toraks ; 68(4): 419-429, 2020 Dec.
Artigo em Turco | MEDLINE | ID: mdl-33448739

RESUMO

Though it has been 8 months since the beginning of COVID-19 pandemic, number of cases and deaths are still seriously increasing. We still don't have enough evidence about the prognosis of patients who had COVID-19 pneumonia. In long term follow up we wonder if they will have rapid FVC decline, widespread fibrosis in computed tomography, loss in quality of life or increased mortality that we experience in idiopathic pulmonary fibrosis, chronic hypersensitivity pneumonia or autoimmune interstitial lung diseases. However, in elderly patients less severe dysfunction or non-progressive-fibrosis can cause morbidity and mortality. Therefore, if we consider millions of people who are affected by COVID-19, even a rare complication can cause serious health problem in social scale. Because of the importance of this issue randomized controlled trials should be rapidly planned on post-COVID fibrosis, COVID associated thrombosis, risk factors, prevention and treatment (1). In this review, the frequency, clinical importance, prevention and treatment of possible long-term sequels of COVID-19 pneumonia (pulmonary fibrosis, pulmonary embolism and pulmonary hypertension) will be discussed.


Assuntos
COVID-19/epidemiologia , Fibrose Pulmonar Idiopática/prevenção & controle , Pandemias , SARS-CoV-2 , COVID-19/complicações , Humanos , Fibrose Pulmonar Idiopática/complicações , Fibrose Pulmonar Idiopática/diagnóstico por imagem , Prognóstico , Fatores de Risco , Tomografia Computadorizada por Raios X
18.
Inflammation ; 43(1): 123-134, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31646446

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a chronic lung disease characterized by progressive lung damage. Tyrosine kinase inhibitors are approved to treat people with IPF while bone marrow-derived mesenchymal stem cell therapy was previously suggested to inhibit pulmonary fibrosis through the alveolar epithelial cell repair. The present study aimed to evaluate the anti-inflammatory and anti-fibrotic effect of the bone marrow-derived mesenchymal stem cell (BM-MSC) therapy in comparison with nintedanib, a tyrosine kinase inhibitor, on improving survival in bleomycin-induced lung fibrosis in rats. Moreover, the combined therapy of BM-MSCs and nintedanib will be evaluated. In the present study, IPF was induced through intra-tracheal instillation of bleomycin (5 mg/kg) in rats then treatments were administered 14 days thereafter. Nintedanib (100 mg/kg, I.P.) was administered daily for 28 days, while BM-MSCs were injected once intravenously in tail vein in the dose 1 × 106 cells/ml/rat. In the present study, both treatment regimens effectively inhibited lung fibrosis through several pathways, suppressing tumor growth factor-ß (TGF-ß)/SMAD3 expression which is considered the master signaling pathway. Nintedanib and BLM-MSCs exerted their anti-inflammatory effect through minimizing the expression of TNF-α and IL-6. In addition, the histopathological examination of the lung tissue showed a significant decrease in the alveolar wall thickening, in the inflammatory infiltrate, and in the collagen fiber deposition in response to either nintedanib or BM-MSC and their combination. In conclusion, the therapeutic pulmonary anti-fibrotic activity of nintedanib or BM-MSC is mediated through their anti-inflammatory properties and inhibition of SMAD-3/TGF-ß expression.


Assuntos
Fibrose Pulmonar Idiopática/prevenção & controle , Indóis/farmacologia , Pulmão/efeitos dos fármacos , Transplante de Células-Tronco Mesenquimais , Inibidores de Proteínas Quinases/farmacologia , Animais , Bleomicina , Células Cultivadas , Terapia Combinada , Modelos Animais de Doenças , Colágenos Fibrilares/metabolismo , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Interleucina-6/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Masculino , Ratos Wistar , Transdução de Sinais , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
19.
Toxicol Lett ; 321: 103-113, 2020 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-31706003

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease with no effective medication. Andrographolide (Andro), extracted from Chinese herbal Andrographis paniculata, could attenuate bleomycin (BLM)-induced pulmonary fibrosis via inhibition of inflammation and oxidative stress, however, the anti-fibrotic mechanisms have not been clarified. Myofibroblasts are the primary cell types responsible for the accumulation of extracellular matrix (ECM) in fibrotic diseases, and targeting fibroblast proliferation and differentiation is an important therapeutic strategy for the treatment of IPF. Hence, this study aimed to investigate the effects of Andro on the fibroblast proliferation and differentiation in the in vivo and in vitro models. The results showed that Andro improved pulmonary function and inhibited BLM-induced fibroblast proliferation and differentiation and ECM deposition in the lungs. In vitro, Andro inhibited proliferation and induced apoptosis of TGF-ß1-stimulated NIH 3T3 fibroblasts and primary lung fibroblasts (PLFs). Andro also inhibited TGF-ß1-induced myofibroblast differentiation and ECM deposition in both cells. We also found that Andro suppressed TGF-ß1-induced Smad2/3 and Erk1/2 activation, suggesting that Smad2/3 and Erk1/2 inactivation mediates Andro-induced effects on TGF-ß1-induced fibroblast proliferation and differentiation. These results indicated that Andro has novel and potent anti-fibrotic effects in lung fibroblasts via inhibition of the proliferation and myofibroblast differentiation of fibroblasts and subsequent ECM deposition, which are modulated by TGF-ß1-mediated Smad-dependent and -independent pathways.


Assuntos
Bleomicina , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Diterpenos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibrose Pulmonar Idiopática/prevenção & controle , Pulmão/efeitos dos fármacos , Miofibroblastos/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Fibrose Pulmonar Idiopática/induzido quimicamente , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Pulmão/patologia , Masculino , Camundongos , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Células NIH 3T3 , Ratos Sprague-Dawley , Transdução de Sinais
20.
Rev. chil. enferm. respir ; 35(4): 310-312, dic. 2019.
Artigo em Espanhol | LILACS | ID: biblio-1092713

RESUMO

Clásicamente entendemos como exacerbación de la Fibrosis pulmonar idiopática (FPI) a un deterioro respiratorio, clínicamente significativo, sin causa evidente. En la actualidad se prefiere el concepto de "exacerbación aguda gatillada" para referirnos a aquella que se genera en el contexto de infección, aspiración, toxicidad por drogas, tromboembolismo pulmonar, insuficiencia cardiaca o posterior a procedimientos invasivos. Mientras que se reserva el termino de "exacerbación aguda idiopática" a aquella en la que no encontramos un gatillante. El pronóstico es ominoso, con mortalidad elevada, con cifras que fluctúan entre 50-90% dependiendo de la necesidad de soporte ventilatorio. Por lo que muchas veces una exacerbación aguda puede ser el evento final de un paciente con FPI. El tratamiento no es del todo claro, no existe evidencia robusta del beneficio de terapias, históricamente los corticoides se han utilizados como terapia estándar, sin embargo la evidencia actual cuestiona los beneficios de dicho tratamiento.


Classically we understand as an exacerbation of Idiopathic Pulmonary Fibrosis (IPF) to a clinically significant respiratory deterioration, without obvious cause. At present, the concept of "acute triggered exacerbations" is preferred to refer to those that are generated in the context of infection, aspiration, drug toxicity, pulmonary thromboembolism, heart failure or after invasive procedures. While the term "idiopathic acute exacerbations" is reserved for those in which we do not find a trigger. The prognosis is ominous and the mortality is high, with figures that fluctuate between 50 to 90% depending on the need for ventilatory support. Many times an acute exacerbation can be the final event of a patient with IPF. The treatment is not entirely clear, there is no robust evidence of the benefit of therapies, historically corticosteroids have been used as standard therapy, however current evidence questions the benefits of such a treatment.


Assuntos
Humanos , Fibrose Pulmonar Idiopática/patologia , Fibrose Pulmonar Idiopática/terapia , Prognóstico , Fatores de Risco , Fibrose Pulmonar Idiopática/prevenção & controle
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA