Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.099
Filtrar
1.
Respir Med ; 227: 107656, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38697229

RESUMEN

RATIONALE: The proportion of patients who develop progressive pulmonary fibrosis (PPF), along with risk factors for progression remain poorly understood. OBJECTIVES: To examine factors associated with an increased risk of developing PPF among patients at a referral center. METHODS: We identified patients with a diagnosis of interstitial lung disease (ILD) seen within the Cleveland Clinic Health System. Utilizing a retrospective observational approach we estimated the risk of developing progression by diagnosis group and identified key clinical predictors using the FVC component of both the original progressive fibrotic interstitial lung disease (PFILD) and the proposed PPF (ATS) criteria. RESULTS: We identified 5934 patients with a diagnosis of ILD. The cumulative incidence of progression over the 24 months was similar when assessed with the PFILD and PPF criteria (33.1 % and 37.9 % respectively). Of those who met the ATS criteria, 9.5 % did not meet the PFILD criteria. Conversely, 4.3 % of patients who met PFILD thresholds did not achieve the 5 % absolute FVC decline criteria. Significant differences in the rate of progression were seen based on underlying diagnosis. Steroid therapy (HR 1.46, CI 1.31-1.62) was associated with an increased risk of progressive fibrosis by both PFILD and PPF criteria. CONCLUSION: Regardless of the definition used, the cumulative incidence of progressive disease is high in patients with ILD in the 24 months following diagnosis. Some differences are seen in the risk of progression when assessed by PFILD and PPF criteria. Further work is needed to identify modifiable risk factors for the development of progressive fibrosis.


Asunto(s)
Progresión de la Enfermedad , Enfermedades Pulmonares Intersticiales , Humanos , Enfermedades Pulmonares Intersticiales/fisiopatología , Enfermedades Pulmonares Intersticiales/epidemiología , Enfermedades Pulmonares Intersticiales/complicaciones , Masculino , Femenino , Estudios Retrospectivos , Capacidad Vital/fisiología , Persona de Mediana Edad , Anciano , Factores de Riesgo , Fibrosis Pulmonar/fisiopatología , Fibrosis Pulmonar/complicaciones , Fibrosis Pulmonar/epidemiología , Incidencia
2.
Lung ; 202(3): 269-273, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38753183

RESUMEN

INTRODUCTION: Pulmonary fibrosis is a characteristic of various interstitial lung diseases (ILDs) with differing etiologies. Clinical trials in progressive pulmonary fibrosis (PPF) enroll patients based on previously described clinical criteria for past progression, which include a clinical practice guideline for PPF classification and inclusion criteria from the INBUILD trial. In this study, we compared the ability of past FVC (forced vital capacity) progression and baseline biomarker levels to predict future progression in a cohort of patients from the PFF Patient Registry. METHODS: Biomarkers previously associated with pathobiology and/or progression in pulmonary fibrosis were selected to reflect cellular senescence (telomere length), pulmonary epithelium (SP-D, RAGE), myeloid activation (CXCL13, YKL40, CCL18, OPN) and fibroblast activation (POSTN, COMP, PROC3). RESULTS: PFF or INBUILD-like clinical criteria was used to separate patients into past progressor and non-past progressor groups, and neither clinical criterion appeared to enrich for patients with greater future lung function decline. All baseline biomarkers measured were differentially expressed in patient groups compared to healthy controls. Baseline levels of SP-D and POSTN showed the highest correlations with FVC slope over one year, though correlations were low. CONCLUSIONS: Our findings provide further evidence that prior decline in lung function may not predict future disease progression for ILD patients, and elevate the need for molecular definitions of a progressive phenotype. Across ILD subtypes, certain shared pathobiologies may be present based on the molecular profile of certain biomarker groups observed. In particular, SP-D may be a common marker of pulmonary injury and future lung function decline across ILDs.


Asunto(s)
Biomarcadores , Progresión de la Enfermedad , Enfermedades Pulmonares Intersticiales , Sistema de Registros , Humanos , Masculino , Femenino , Persona de Mediana Edad , Capacidad Vital , Anciano , Enfermedades Pulmonares Intersticiales/fisiopatología , Enfermedades Pulmonares Intersticiales/diagnóstico , Fibrosis Pulmonar/fisiopatología , Fibrosis Pulmonar/diagnóstico , Proteína D Asociada a Surfactante Pulmonar/sangre , Pulmón/fisiopatología , Valor Predictivo de las Pruebas , Proteína 1 Similar a Quitinasa-3/sangre , Quimiocinas CC , Osteopontina , Receptor para Productos Finales de Glicación Avanzada/sangre , Fibrosis Pulmonar Idiopática/fisiopatología , Fibrosis Pulmonar Idiopática/diagnóstico
3.
Lung ; 202(3): 257-267, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38713420

RESUMEN

PURPOSE: World Trade Center (WTC) exposure is associated with obstructive airway diseases and sarcoidosis. There is limited research regarding the incidence and progression of non-sarcoidosis interstitial lung diseases (ILD) after WTC-exposure. ILD encompasses parenchymal diseases which may lead to progressive pulmonary fibrosis (PPF). We used the Fire Department of the City of New York's (FDNY's) WTC Health Program cohort to estimate ILD incidence and progression. METHODS: This longitudinal study included 14,525 responders without ILD prior to 9/11/2001. ILD incidence and prevalence were estimated and standardized to the US 2014 population. Poisson regression modeled risk factors, including WTC-exposure and forced vital capacity (FVC), associated with ILD. Follow-up time ended at the earliest of incident diagnosis, end of study period/case ascertainment, transplant or death. RESULTS: ILD developed in 80/14,525 FDNY WTC responders. Age, smoking, and gastroesophageal reflux disease (GERD) prior to diagnosis were associated with incident ILD, though FVC was not. PPF developed in 40/80 ILD cases. Among the 80 cases, the average follow-up time after ILD diagnosis was 8.5 years with the majority of deaths occurring among those with PPF (PPF: n = 13; ILD without PPF: n = 6). CONCLUSIONS: The prevalence of post-9/11 ILD was more than two-fold greater than the general population. An exposure-response gradient could not be demonstrated. Half the ILD cases developed PPF, higher than previously reported. Age, smoking, and GERD were risk factors for ILD and PPF, while lung function was not. This may indicate that lung function measured after respirable exposures would not identify those at risk for ILD or PPF.


Asunto(s)
Progresión de la Enfermedad , Enfermedades Pulmonares Intersticiales , Fibrosis Pulmonar , Ataques Terroristas del 11 de Septiembre , Humanos , Estudios Longitudinales , Masculino , Enfermedades Pulmonares Intersticiales/epidemiología , Enfermedades Pulmonares Intersticiales/fisiopatología , Persona de Mediana Edad , Femenino , Incidencia , Capacidad Vital , Adulto , Prevalencia , Factores de Riesgo , Fibrosis Pulmonar/epidemiología , Fibrosis Pulmonar/fisiopatología , Ciudad de Nueva York/epidemiología , Reflujo Gastroesofágico/epidemiología , Exposición Profesional/efectos adversos , Fumar/efectos adversos , Fumar/epidemiología , Anciano , Factores de Tiempo , Socorristas/estadística & datos numéricos
5.
Clin Sci (Lond) ; 137(11): 895-912, 2023 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-37314017

RESUMEN

Circadian regulation causes the activity of biological processes to vary over a 24-h cycle. The pathological effects of this variation are predominantly studied using two different approaches: pre-clinical models or observational clinical studies. Both these approaches have provided useful insights into how underlying circadian mechanisms operate and specifically which are regulated by the molecular oscillator, a key time-keeping mechanism in the body. This review compares and contrasts findings from these two approaches in the context of four common respiratory diseases (asthma, chronic obstructive pulmonary disease, pulmonary fibrosis, and respiratory infection). Potential methods used to identify and measure human circadian oscillations are also discussed as these will be useful outcome measures in future interventional human trials that target circadian mechanisms.


Asunto(s)
Relojes Circadianos , Enfermedades Pulmonares , Humanos , Asma/fisiopatología , Relojes Circadianos/fisiología , Enfermedades Pulmonares/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Fibrosis Pulmonar/fisiopatología , Infecciones del Sistema Respiratorio/fisiopatología , Factores de Tiempo , Ensayos Clínicos como Asunto , Proyectos de Investigación
6.
Int Immunopharmacol ; 113(Pt A): 109339, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36330909

RESUMEN

Our previous study showed that triggering receptors expressed on myeloid cell-1 (TREM-1) was upregulated in bleomycin (BLM)-induced pulmonary fibrosis (PF) mouse model. However, the role of TREM-1 in the development of PF and its underlying mechanism remain unclear. Herein, we report that the prophylactical blockade of TREM-1 using a decoy peptide dodecapeptide (LR12) exerted protective effects against BLM-induced PF in mice, with a higher survival rate, attenuated tissue injury, and less extracellular matrix deposition. Interestingly, therapeutic blockade of TREM-1 at the early stage of fibrosis also attenuated BLM-induced PF, suggesting a non-inflammatory effect. More importantly, we observed that TREM-1 blockade with LR12 significantly reduced the expression of the senescence-relative protein, including p16, p21, p53, and γ-H2AX in the lungs of PF mice. Notably, TREM-1 was upregulated in alveolar epithelial cells (AECs) and correlated with the levels of senescence markers in BLM-treated mice. In vitro, activating TREM-1 with an agonistic antibody exacerbated BLM-induced senescence in MLE12 cells, a murine AEC cell line. Furthermore, prophylactic or therapeutic blockade of TREM-1 protected MLE12 cells from senescence induced by BLM or H2O2. In conclusion, our findings elucidate a pro-fibrotic effect of TREM-1 by inducing AECs senescence in PF, providing a potential strategy for fibrotic disease treatment.


Asunto(s)
Células Epiteliales Alveolares , Fibrosis Pulmonar , Receptor Activador Expresado en Células Mieloides 1 , Animales , Ratones , Células Epiteliales Alveolares/patología , Bleomicina/toxicidad , Peróxido de Hidrógeno/metabolismo , Células Mieloides , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/fisiopatología , Receptor Activador Expresado en Células Mieloides 1/metabolismo
7.
Proc Natl Acad Sci U S A ; 119(10): e2116279119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35238669

RESUMEN

The warning cytokine interleukin-33 receptor (IL-33R) mediates local inflammatory responses and plays crucial roles in the pathogenesis of immune diseases such as pulmonary fibrosis and rheumatoid arthritis. Whether and how IL-33R is regulated remain enigmatic. Here, we identified ubiquitin-specific protease 38 (USP38) as a negative regulator of IL-33R­mediated signaling. USP38 deficiency promotes interleukin-33 (IL-33)­induced downstream proinflammatory responses in vitro and in vivo. Usp38−/− mice are more susceptible to inflammatory damage and death and developed more serious pulmonary fibrosis after bleomycin treatment. USP38 is constitutively associated with IL-33R and deconjugates its K27-linked polyubiquitination at K511, resulting in its autophagic degradation. We further show that the E3 ubiquitin ligase tumor necrosis factor receptor­associated factor 6 (TRAF6) catalyzes K27-linked polyubiquitination of IL-33R at K511, and that deficiency of TRAF6 inhibits IL-33­mediated signaling. Our findings suggest that K27-linked polyubiquitination and deubiquitination of IL-33R by TRAF6 and USP38 reciprocally regulate IL-33R level and signaling, which represents a critical mechanism in the regulation of IL-33­triggered lung inflammatory response and pulmonary fibrosis.


Asunto(s)
Inflamación/fisiopatología , Interleucina-33/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Fibrosis Pulmonar/fisiopatología , Proteasas Ubiquitina-Específicas/metabolismo , Autofagia , Regulación hacia Abajo , Humanos , Inflamación/metabolismo , Interleucina-33/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Transducción de Señal , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
8.
Toxicol Lett ; 359: 65-72, 2022 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-35143883

RESUMEN

This study aimed to investigate the correlation between prenatal dexamethasone exposure (PDE) and susceptibility to pulmonary fibrosis in offspring. Healthy female Wistar rats were given dexamethasone (0.2 mg/kg.d) or an equal volume of normal saline subcutaneously from 9 to 20 days after conception. Some of their female offspring underwent ovariectomy (OV) at 22 weeks after birth. All animals were euthanized at 28 weeks after birth. The morphological changes related to pulmonary fibrosis and extracellular matrix-related gene expression were detected, and Two-way ANOVA analyzed the interaction between PDE and OV. The results showed that adult offspring rats in FD group (female rats with PDE treatment) had early pulmonary fibrosis changes, such as pulmonary interstitial thickening, and increased expression of type IV collagen (COL4), α -smooth muscle actin (α-SMA) and fibronectin (FN) in lung tissues compared with those in FC group (female rats with saline treatment). In addition, adult offspring rats in FDO group (female rats with PDE and OV treatment) showed signs of pulmonary fibrosis, including apparent extracellular matrix deposition, increased lung injury scores (P<0.01, P<0.05), and extracellular matrix related gene expression (P<0.01, P<0.05), compared with rats in FDS (female rats with PDE treatment alone) or rats in FCO group (female rats with OV treatment alone). Moreover, PDE and OV had an interactive effect on the development of pulmonary fibrosis in female adult offspring. This study first reported the correlation between PDE and susceptibility to pulmonary fibrosis in female offspring rats, as well as the synergistic effect of PDE and OV in this pathological event, which provided a basis for further understanding of the pathogenesis of fetal originated pulmonary fibrosis.


Asunto(s)
Dexametasona/toxicidad , Susceptibilidad a Enfermedades/inducido químicamente , Desarrollo Fetal/efectos de los fármacos , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Desarrollo Fetal/genética , Humanos , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/genética , Ratas , Ratas Wistar , Factores Sexuales
9.
Biomed Pharmacother ; 145: 112245, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34772578

RESUMEN

Cathepsin S (CTSS), a lysosomal protease, belongs to a family of cysteine cathepsin proteases that promote degradation of damaged proteins in the endolysosomal pathway. Aberrant CTSS expression and regulation are associated with the pathogenesis of several diseases, including lung diseases. CTSS overexpression causes a variety of pathological processes, including pulmonary fibrosis, with increased CTSS secretion and accelerated extracellular matrix remodeling. Compared to many other cysteine cathepsin family members, CTSS has unique features that it presents limited tissue expression and retains its enzymatic activity at a neutral pH, suggesting its decisive involvement in disease microenvironments. In this review, we investigated the role of CTSS in lung disease, exploring recent studies that have indicated that CTSS mediates fibrosis in unique ways, along with its structure, substrates, and distinct regulation. We also outlined examples of CTSS inhibitors in clinical and preclinical development and proposed CTSS as a potential therapeutic target for pulmonary fibrosis.


Asunto(s)
Catepsinas/antagonistas & inhibidores , Fibrosis Pulmonar/tratamiento farmacológico , Animales , Catepsinas/metabolismo , Desarrollo de Medicamentos , Matriz Extracelular/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Terapia Molecular Dirigida , Fibrosis Pulmonar/fisiopatología
11.
Exp Mol Pathol ; 124: 104737, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34953919

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a chronic interstitial disease of the lung tissue that causes symptoms such as coughing and asthma. It is caused by inflammatory factors and oxidative stress. In vivo model of IPF is induced by bleomycin (BLM,) a chemotherapeutic agent. We have investigated the effect of dapsone on bleomycin-induced IPF in adult male Wistar rats due to its anti-inflammatory and anti-oxidative stress effects. The animals were randomly divided into 5 groups (Control, BLM, BLM + dapsone 1, BLM + Dapsone 3, BLM + Dapsone 10). The control group received normal water and food. In the fibrosis group, bleomycin (BLM) (5 mg/kg) was used to induce pulmonary fibrosis by intratracheal administration. Three groups of animals were treated daily with single doses of 1, 3, and 10 mg dapsone by intraperitoneal injection 1 h after receiving BLM for 2 weeks. The activity of antioxidant enzymes superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), and oxidative stress markers such as myeloperoxidase (MPO), malondialdehyde (MDA), protein carbonyl (PC) and nitrite were measured to evaluate bleomycin and therapeutic effect of dapsone. The histological assays of lung tissues were done by Hematoxylin-eosin (H & E) and Masson's trichrome staining. BLM reduced the activity of oxidative enzymes and increased the oxidative stress markers, while treatment with dapsone has reversed the results. In addition, the total number of cells as inflammatory cells such as neutrophils and eosinophils were examined. It has been indicated BLM increased these cells, and dapsone decreased them. The results of H & E and Masson's trichrome staining showed that dapsone reduced inflammation and alveolar wall thickness and BLM-induced pulmonary fibrosis. According to the findings of this study, dapsone seems to have therapeutic effects on pulmonary fibrosis through its anti-inflammatory and anti-oxidative stress properties and reduction of the toxic effects of bleomycin.


Asunto(s)
Bleomicina/efectos adversos , Dapsona/farmacología , Fibrosis Pulmonar , Animales , Antiinflamatorios/administración & dosificación , Antiinflamatorios/farmacología , Antioxidantes/administración & dosificación , Antioxidantes/farmacología , Bleomicina/toxicidad , Catalasa/metabolismo , Dapsona/administración & dosificación , Modelos Animales de Enfermedad , Glutatión Peroxidasa/metabolismo , Histocitoquímica , Pulmón/citología , Pulmón/patología , Estrés Oxidativo/efectos de los fármacos , Peroxidasa/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/fisiopatología , Ratas , Ratas Wistar , Superóxido Dismutasa/metabolismo
12.
Arch Biochem Biophys ; 715: 109087, 2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34801472

RESUMEN

The epithelial-mesenchymal transition (EMT) is an important pathological process in the occurrence of pulmonary fibrosis. Changes in histone methylation modifications of key genes play an important role in this process. As a histone methyltransferase, the regulatory mechanism and role of SET domain bifurcated 1 (SETDB1) in pulmonary fibrosis remain unclear. We found that SETDB1 inhibited EMT and that cells attenuated the expression of SETDB1 to relieve this inhibition during transforming growth factor-ß (TGF-ß)-induced EMT. Silencing SETDB1 expression significantly enhanced the mesenchymal phenotype induced by TGF-ß and the expression and deposition of fibronectin and significantly reduced the expression of E-cadherin. The decrease in E-cadherin expression and the induction of EMT led to increased lipid reactive oxygen species (ROS) and ferrous ions, which induced ferroptosis. Chromatin immunoprecipitation (ChIP) results showed that SETDB1 regulates the expression of Snai1 by catalyzing the histone H3 lysine 9 trimethylation (H3K9me3) of Snai1, the main transcription factor that initiates the process of EMT, and thus, indirectly regulates E-cadherin. Surprisingly, when examining the effect of overexpressed SETDB1 on EMT, we found that overexpressed SETDB1 alleviated EMT and also caused ferroptosis. We suggest that the overexpression of SETDB1 partially reverses the mesenchymal phenotype to an epithelial state, while those cells that fail to reverse are depleted by ferroptosis. In conclusion, the histone methylase SETDB1 regulates Snai1 epigenetically, driving EMT gene reprogramming and ferroptosis in response to TGF-ß. However, there are unexplored links between the epigenetic reprogramming and transcriptional processes that regulate EMT in a TGF-ß-dependent manner.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Ferroptosis/fisiología , N-Metiltransferasa de Histona-Lisina/metabolismo , Fibrosis Pulmonar/fisiopatología , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Células A549 , Animales , Bleomicina , Transición Epitelial-Mesenquimal/efectos de los fármacos , Técnicas de Silenciamiento del Gen , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Regiones Promotoras Genéticas/fisiología , Fibrosis Pulmonar/inducido químicamente , Ratas Sprague-Dawley
13.
Int J Mol Sci ; 22(23)2021 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-34884731

RESUMEN

Pulmonary fibrosis (PF) is a progressive disease that disrupts the mechanical homeostasis of the lung extracellular matrix (ECM). These effects are particularly relevant in the lung context, given the dynamic nature of cyclic stretch that the ECM is continuously subjected to during breathing. This work uses an in vivo model of pulmonary fibrosis to characterize the macro- and micromechanical properties of lung ECM subjected to stretch. To that aim, we have compared the micromechanical properties of fibrotic ECM in baseline and under stretch conditions, using a novel combination of Atomic Force Microscopy (AFM) and a stretchable membrane-based chip. At the macroscale, fibrotic ECM displayed strain-hardening, with a stiffness one order of magnitude higher than its healthy counterpart. Conversely, at the microscale, we found a switch in the stretch-induced mechanical behaviour of the lung ECM from strain-hardening at physiological ECM stiffnesses to strain-softening at fibrotic ECM stiffnesses. Similarly, we observed solidification of healthy ECM versus fluidization of fibrotic ECM in response to stretch. Our results suggest that the mechanical behaviour of fibrotic ECM under stretch involves a potential built-in mechanotransduction mechanism that may slow down the progression of PF by steering resident fibroblasts away from a pro-fibrotic profile.


Asunto(s)
Matriz Extracelular/fisiología , Mecanotransducción Celular , Fibrosis Pulmonar/fisiopatología , Animales , Bleomicina , Modelos Animales de Enfermedad , Elasticidad , Masculino , Microscopía de Fuerza Atómica , Ratas Sprague-Dawley
14.
Front Immunol ; 12: 740260, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34745111

RESUMEN

Increased left ventricular fibrosis has been reported in patients hospitalized with coronavirus disease 2019 (COVID-19). It is unclear whether this fibrosis is a consequence of severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2) infection or a risk factor for severe disease progression. We observed increased fibrosis in the left ventricular myocardium of deceased COVID-19 patients, compared with matched controls. We also detected increased mRNA levels of soluble interleukin-1 receptor-like 1 (sIL1-RL1) and transforming growth factor ß1 (TGF-ß1) in the left ventricular myocardium of deceased COVID-19 patients. Biochemical analysis of blood sampled from patients admitted to the emergency department (ED) with COVID-19 revealed highly elevated levels of TGF-ß1 mRNA in these patients compared to controls. Left ventricular strain measured by echocardiography as a marker of pre-existing cardiac fibrosis correlated strongly with blood TGF-ß1 mRNA levels and predicted disease severity in COVID-19 patients. In the left ventricular myocardium and lungs of COVID-19 patients, we found increased neuropilin-1 (NRP-1) RNA levels, which correlated strongly with the prevalence of pulmonary SARS-CoV-2 nucleocapsid. Cardiac and pulmonary fibrosis may therefore predispose these patients to increased cellular viral entry in the lung, which may explain the worse clinical outcome observed in our cohort. Our study demonstrates that patients at risk of clinical deterioration can be identified early by echocardiographic strain analysis and quantification of blood TGF-ß1 mRNA performed at the time of first medical contact.


Asunto(s)
COVID-19/fisiopatología , Ventrículos Cardíacos/patología , Miocardio/patología , Fibrosis Pulmonar/fisiopatología , SARS-CoV-2/fisiología , Adulto , Anciano , COVID-19/inmunología , Femenino , Fibrosis , Ventrículos Cardíacos/metabolismo , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Masculino , Persona de Mediana Edad , Miocardio/metabolismo , Neuropilina-1/genética , Neuropilina-1/metabolismo , Fibrosis Pulmonar/inmunología , Riesgo , Índice de Severidad de la Enfermedad , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Carga Viral
15.
Indian J Pathol Microbiol ; 64(4): 644-650, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34673580

RESUMEN

BACKGROUND: Silicosis is a public health issue in developing countries for long and cannot be completely cured. OBJECTIVE: To study the changes of ion content with TNF-α and TGF-ß expression in alveolar lavage fluid (BALF) at different time points in rats exposed to silica and to investigate their correlation with pulmonary fibrosis. METHODS: 42 rats were randomly divided into control group (n = 12) and exposure group (n = 30). Tissues of right lower lungs were collected and fixed for further Hematoxylin-eosin (HE) and Masson staining. We collected the BALF to examine the inflammatory cytokines of TNF-α and TGF-ß and measured the ion contents in BALF. RESULTS: The increase of TNF-α level was earlier than TGF-ß. The content of silica in BALF was significantly increased after exposure and reached the maximum at 7th day, similar to the curve of cytokine TGF-ß level. However, phosphorus ions increased quickly after gradual decline of silicon ion and roughly proportional to the curve of degree of fibrosis. CONCLUSIONS: Crystalline silica exposure can cause changes in TGF-ß and TNF-α in BALF and accompanied with fibrosis and ions content variation. The abnormal expression of phosphorus ion may have significance in the occurrence and development of silicosis.


Asunto(s)
Exposición a Riesgos Ambientales/efectos adversos , Fósforo/análisis , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/fisiopatología , Dióxido de Silicio/efectos adversos , Silicio/análisis , Silicosis/diagnóstico , Adulto , Animales , Biomarcadores/análisis , Femenino , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Profesionales/diagnóstico , Enfermedades Profesionales/fisiopatología , Ratas , Ratas Sprague-Dawley , Silicosis/fisiopatología
16.
Pharmacol Res ; 174: 105924, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34607005

RESUMEN

Pulmonary fibrosis is a devastating lung disease with multifactorial etiology characterized by alveolar injury, fibroblast proliferation and excessive deposition of extracellular matrix proteins, which progressively results in respiratory failure and death. Accumulating evidence from experimental and clinical studies supports a central role of the renin angiotensin aldosterone system (RAAS) in the pathogenesis and progression of idiopathic pulmonary fibrosis. Angiotensin II (Ang II), a key vasoactive peptide of the RAAS mediates pro-inflammatory and pro-fibrotic effects on the lungs, adversely affecting organ function. Recent years have witnessed seminal discoveries in the field of RAAS. Identification of new enzymes, peptides and receptors has led to the development of several novel concepts. Of particular interest is the establishment of a protective axis of the RAAS comprising of Angiotensin converting enzyme 2 (ACE2), Angiotensin-(1-7) [Ang-(1-7)], and the Mas receptor (the ACE2/Ang-(1-7)/Mas axis), and the discovery of a functional role for the Angiotensin type 2 (AT2) receptor. Herein, we will review our current understanding of the role of RAAS in lung fibrogenesis, provide evidence on the anti-fibrotic actions of the newly recognized RAAS components (the ACE2/Ang-(1-7)/Mas axis and AT2 receptor), discuss potential strategies and translational efforts to convert this new knowledge into effective therapeutics for PF.


Asunto(s)
Pulmón/metabolismo , Fibrosis Pulmonar/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Angiotensina I/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , Antifibróticos/uso terapéutico , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Pulmón/fisiopatología , Terapia Molecular Dirigida , Fragmentos de Péptidos/metabolismo , Proto-Oncogenes Mas/metabolismo , Fibrosis Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/fisiopatología , Transducción de Señal
17.
Bull Exp Biol Med ; 171(4): 523-531, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34542758

RESUMEN

Idiopathic pulmonary fibrosis can be caused by different factors, including accumulation of pathological extracellular matrix (ECM) with abnormal composition, stiffness, and architecture in the lung tissue. We studied the effect of ECM produced by lung fibroblasts of healthy mice or mice with bleomycin-induced pulmonary fibrosis on the process of endothelialto- mesenchymal transition, one of the main sources of effector myofibroblasts in fibrosis progression. Despite stimulation of spontaneous and TGFß-1-induced differentiation of fibroblasts into myofibroblasts by fibrotic ECM, the appearance of α-SMA, the main marker of myofibroblasts, and its integration in stress fibrils in endotheliocytes were not observed under similar conditions. However, the expression of transcription factors SNAI1 and SNAI2/Slug and the production of components of fibrotic ECM (specific EDA-fibronectin splice form and collagen type I) were increased in endotheliocytes cultured on fibrotic ECM. Endothelium also demonstrated increased cell velocity in the models of directed cell migration. These data indicate activation of the intermediate state of the endothelial-to-mesenchymal transition in endotheliocytes upon contact with fibrotic, but not normal stromal matrix. In combination with the complex microenvironment that develops during fibrosis progression, it can lead to the replenishment of myofibroblasts pool from the resident endothelium.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Matriz Extracelular/fisiología , Fibrosis Pulmonar/patología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Matriz Extracelular Descelularizada/química , Matriz Extracelular Descelularizada/metabolismo , Matriz Extracelular Descelularizada/farmacología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Fibroblastos/fisiología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Miofibroblastos/efectos de los fármacos , Miofibroblastos/fisiología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/fisiopatología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Andamios del Tejido
18.
Respir Med ; 188: 106602, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34536697

RESUMEN

INTRODUCTION: Survivors of COVID-19 infection may develop post-covid pulmonary fibrosis (PCF) and suffer from long term multi-system complications. The magnitude and risk factors associated with these are unknown. OBJECTIVES: We investigated the prevalence and risk factors associated with PCF and other complications in patients discharged after COVID-19 infection. METHODS: Patients had phone assessment 6 weeks post hospital discharge after COVID-19 infection using a set protocol. Those with significant respiratory symptoms were investigated with a CTPA, Pulmonary Function Tests and echocardiogram. Prevalence of myalgia, fatigue, psychological symptoms and PCF was obtained. Risk factors associated with these were investigated. RESULTS: A large number of patients had persistent fatigue (45.1%), breathlessness (36.5%), myalgia (20.5%) and psychological symptoms (19.5%). PCF was seen in 9.5% of the patients and was associated with persistent breathlessness at 6 weeks and inpatient ventilation [adjusted OR 5.02(1.76-14.27) and 4.45(1.27-15.58)] respectively. It was more common in men and in patients with peak CRP >171.5 mg/L, peak WBC count ≥12 × 10 9/L, severe inpatient COVID-19 CXR changes and CT changes. Ventilation was also a risk factor for persisting fatigue and myalgia, the latter was also more common in those with severe cytokine storm and severe COVID-19 inpatient CXR changes. CONCLUSIONS: All the patients discharged after COVID-19 should be assessed using a set protocol by a multidisciplinary team. Patients who had severe COVID-19 infection particularly those who were intubated and who have persistent breathlessness are at risk of developing PCF. They should have a CT Chest and have respiratory follow-up.


Asunto(s)
COVID-19/complicaciones , Pulmón/fisiopatología , Pandemias , Alta del Paciente/tendencias , Fibrosis Pulmonar/etiología , SARS-CoV-2 , Adulto , COVID-19/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Prevalencia , Fibrosis Pulmonar/diagnóstico , Fibrosis Pulmonar/fisiopatología , Pruebas de Función Respiratoria , Factores de Riesgo , Reino Unido/epidemiología
19.
Toxicology ; 461: 152925, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34481903

RESUMEN

Silica dust is a common pollutant in the occupational environment, such as coal mines. Inhalation of silica dust can cause progressive pulmonary fibrosis and then silicosis. Silicosis is still one of the most harmful occupational diseases in the world, so the study of its pathogenesis is necessary for the treatment of silicosis. In this study, we constructed a mouse model of pulmonary fibrosis via intratracheal instillation of silica particles and identified the decreased expression of miR-138 in fibrotic lung tissues of mice. Moreover, the overexpression of miR-138 retarded the process of epithelial-mesenchymal transition (EMT) in a mouse model of silica particles exposure and epithelial cells stimulated by silica particles. Further studies showed that ZEB2 was one of the potential targets of miR-138, and the up-regulation of miR-138 reduced ZEB2 levels in mouse lung tissues and in epithelial cells. We next found that the expression levels of ɑ-SMA and Vimentin were significantly increased and E-cadherin levels were decreased after transfection with miR-138 inhibitor in epithelial cells. However, these effects were abated by the knockdown of ZEB2. Consistently, the increased migration ability of epithelial cells by miR-138 inhibitor transfection was also reversed by the knockdown of ZEB2. Collectively, we revealed that miR-138 significantly targeted ZEB2, thus inhibited the EMT process and mitigated the development of pulmonary fibrosis. miR-138 may be a potential target for the treatment of pulmonary fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal/efectos de los fármacos , MicroARNs/genética , Fibrosis Pulmonar/inducido químicamente , Dióxido de Silicio/toxicidad , Células A549 , Animales , Movimiento Celular/genética , Modelos Animales de Enfermedad , Contaminantes Ambientales/administración & dosificación , Contaminantes Ambientales/toxicidad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fibrosis/patología , Técnicas de Silenciamiento del Gen , Humanos , Exposición por Inhalación/efectos adversos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/fisiopatología , Dióxido de Silicio/administración & dosificación , Silicosis/fisiopatología , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo
20.
PLoS One ; 16(9): e0257281, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34529707

RESUMEN

The development of more effective, better tolerated drug treatments for progressive pulmonary fibrosis (of which idiopathic pulmonary fibrosis is the most common and severe form) is a research priority. The peroxisome proliferator-activated receptor gamma (PPAR-γ) is a key regulator of inflammation and fibrosis and therefore represents a potential therapeutic target. However, the use of synthetic PPAR-γ agonists may be limited by their potentially severe adverse effects. In a mouse model of bleomycin (BLM)-induced pulmonary fibrosis, we have demonstrated that the non-racemic selective PPAR-γ modulator GED-0507 is able to reduce body weight loss, ameliorate clinical and histological features of pulmonary fibrosis, and increase survival rate without any safety concerns. Here, we focused on the biomolecular effects of GED-0507 on various inflammatory/fibrotic pathways. We demonstrated that preventive and therapeutic administration of GED-0507 reduced the BLM-induced mRNA expression of several markers of fibrosis, including transforming growth factor (TGF)-ß, alpha-smooth muscle actin, collagen and fibronectin as well as epithelial-to-mesenchymal transition (EMT) and expression of mucin 5B. The beneficial effect of GED-0507 on pulmonary fibrosis was confirmed in vitro by its ability to control TGFß-induced myofibroblast activation in the A549 human alveolar epithelial cell line, the MRC-5 lung fibroblast line, and primary human lung fibroblasts. Compared with the US Food and Drug Administration-approved antifibrotic drugs pirfenidone and nintedanib, GED-0507 displayed greater antifibrotic activity by controlling alveolar epithelial cell dysfunction, EMT, and extracellular matrix remodeling. In conclusion, GED-0507 demonstrated potent antifibrotic properties and might be a promising drug candidate for the treatment of pulmonary fibrosis.


Asunto(s)
Transdiferenciación Celular , Miofibroblastos/citología , Propionatos/farmacología , Fibrosis Pulmonar/tratamiento farmacológico , Células A549 , Animales , Bleomicina , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Técnicas In Vitro , Inflamación , Pulmón/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , PPAR gamma/metabolismo , Fibrosis Pulmonar/fisiopatología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA