Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 300(1): 105530, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38072048

RESUMO

Fibroblast to myofibroblast transdifferentiation mediates numerous fibrotic disorders, such as idiopathic pulmonary fibrosis (IPF). We have previously demonstrated that non-muscle myosin II (NMII) is activated in response to fibrotic lung extracellular matrix, thereby mediating myofibroblast transdifferentiation. NMII-A is known to interact with the calcium-binding protein S100A4, but the mechanism by which S100A4 regulates fibrotic disorders is unclear. In this study, we show that fibroblast S100A4 is a calcium-dependent, mechanoeffector protein that is uniquely sensitive to pathophysiologic-range lung stiffness (8-25 kPa) and thereby mediates myofibroblast transdifferentiation. Re-expression of endogenous fibroblast S100A4 rescues the myofibroblastic phenotype in S100A4 KO fibroblasts. Analysis of NMII-A/actin dynamics reveals that S100A4 mediates the unraveling and redistribution of peripheral actomyosin to a central location, resulting in a contractile myofibroblast. Furthermore, S100A4 loss protects against murine in vivo pulmonary fibrosis, and S100A4 expression is dysregulated in IPF. Our data reveal a novel mechanosensor/effector role for endogenous fibroblast S100A4 in inducing cytoskeletal redistribution in fibrotic disorders such as IPF.


Assuntos
Fibrose Pulmonar Idiopática , Mecanotransdução Celular , Miofibroblastos , Proteína A4 de Ligação a Cálcio da Família S100 , Animais , Camundongos , Transdiferenciação Celular , Fibrose , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Pulmão/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Proteína A4 de Ligação a Cálcio da Família S100/genética , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo
2.
Am J Respir Cell Mol Biol ; 70(6): 457-467, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38346220

RESUMO

Sepsis is a systemic inflammatory response that requires effective macrophage metabolic functions to resolve ongoing inflammation. Previous work showed that the mechanosensitive cation channel, transient receptor potential vanilloid 4 (TRPV4), mediates macrophage phagocytosis and cytokine production in response to lung infection. Here, we show that TRPV4 regulates glycolysis in a stiffness-dependent manner by augmenting macrophage glucose uptake by GLUT1. In addition, TRPV4 is required for LPS-induced phagolysosome maturation in a GLUT1-dependent manner. In a cecal slurry mouse model of sepsis, TRPV4 regulates sepsis-induced glycolysis as measured by BAL fluid (BALF) lactate and sepsis-induced lung injury as measured by BALF total protein and lung compliance. TRPV4 is necessary for bacterial clearance in the peritoneum to limit sepsis-induced lung injury. It is interesting that BALF lactate is increased in patients with sepsis compared with healthy control participants, supporting the relevance of lung cell glycolysis to human sepsis. These data show that macrophage TRPV4 is required for glucose uptake through GLUT1 for effective phagolysosome maturation to limit sepsis-induced lung injury. Our work presents TRPV4 as a potential target to protect the lung from injury in sepsis.


Assuntos
Transportador de Glucose Tipo 1 , Glicólise , Lesão Pulmonar , Macrófagos , Sepse , Canais de Cátion TRPV , Animais , Canais de Cátion TRPV/metabolismo , Sepse/metabolismo , Sepse/complicações , Transportador de Glucose Tipo 1/metabolismo , Transportador de Glucose Tipo 1/genética , Camundongos , Lesão Pulmonar/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Humanos , Masculino , Glucose/metabolismo , Fagossomos/metabolismo , Líquido da Lavagem Broncoalveolar , Lipopolissacarídeos/farmacologia , Fagocitose , Modelos Animais de Doenças , Pulmão/metabolismo , Pulmão/patologia , Pulmão/imunologia
3.
J Immunol ; 204(5): 1310-1321, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31969384

RESUMO

Mechanical cell-matrix interactions can drive the innate immune responses to infection; however, the molecular underpinnings of these responses remain elusive. This study was undertaken to understand the molecular mechanism by which the mechanosensitive cation channel, transient receptor potential vanilloid 4 (TRPV4), alters the in vivo response to lung infection. For the first time, to our knowledge, we show that TRPV4 protects the lung from injury upon intratracheal Pseudomonas aeruginosa in mice. TRPV4 functions to enhance macrophage bacterial clearance and downregulate proinflammatory cytokine secretion. TRPV4 mediates these effects through a novel mechanism of molecular switching of LPS signaling from predominant activation of the MAPK, JNK, to that of p38. This is accomplished through the activation of the master regulator of inflammation, dual-specificity phosphatase 1. Further, TRPV4's modulation of the LPS signal is mechanosensitive in that both upstream activation of p38 and its downstream biological consequences depend on pathophysiological range extracellular matrix stiffness. We further show the importance of TRPV4 on LPS-induced activation of macrophages from healthy human controls. These data are the first, to our knowledge, to demonstrate new roles for macrophage TRPV4 in regulating innate immunity in a mechanosensitive manner through the modulation of dual-specificity phosphatase 1 expression to mediate MAPK activation switching.


Assuntos
Pulmão , Sistema de Sinalização das MAP Quinases , Ativação de Macrófagos , Macrófagos/imunologia , Pneumonia Bacteriana , Infecções por Pseudomonas , Pseudomonas aeruginosa/imunologia , Canais de Cátion TRPV/imunologia , Animais , Feminino , Humanos , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/patologia , Sistema de Sinalização das MAP Quinases/genética , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos/patologia , Camundongos , Camundongos Mutantes , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/imunologia , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/imunologia , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/prevenção & controle , Infecções por Pseudomonas/genética , Infecções por Pseudomonas/imunologia , Infecções por Pseudomonas/prevenção & controle , Canais de Cátion TRPV/genética
4.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L1074-L1084, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33787326

RESUMO

The transient receptor potential vanilloid 1 (TRPV1) channel is expressed in human bronchial epithelium (HBE), where it transduces Ca2+ in response to airborne irritants. TRPV1 activation results in bronchoconstriction, cough, and mucus production, and may therefore contribute to the pathophysiology of obstructive airway disease. Since children with asthma face the greatest risk of developing virus-induced airway obstruction, we hypothesized that changes in TRPV1 expression, localization, and function in the airway epithelium may play a role in bronchiolitis and asthma in childhood. We sought to measure TRPV1 protein expression, localization, and function in HBE cells from children with versus without asthma, both at baseline and after RSV infection. We determined changes in TRPV1 protein expression, subcellular localization, and function both at baseline and after RSV infection in primary HBE cells from normal children and children with asthma. Basal TRPV1 protein expression was higher in HBE from children with versus without asthma and primarily localized to plasma membranes (PMs). During RSV infection, TRPV1 protein increased more in the PM of asthmatic HBE as compared with nonasthmatic cells. TRPV1-mediated increase in intracellular Ca2+ was greater in RSV-infected asthmatic cells, but this increase was attenuated when extracellular Ca2+ was removed. Nerve growth factor (NGF) recapitulated the effect of RSV on TRPV1 activation in HBE cells. Our data suggest that children with asthma have intrinsically hyperreactive airways due in part to higher TRPV1-mediated Ca2+ influx across epithelial membranes, and this abnormality is further exacerbated by NGF overexpression during RSV infection driving additional Ca2+ from intracellular stores.


Assuntos
Asma/virologia , Cálcio/metabolismo , Transporte de Íons/fisiologia , Canais de Cátion TRPV/metabolismo , Asma/metabolismo , Broncoconstrição/fisiologia , Criança , Pré-Escolar , Células Epiteliais/metabolismo , Epitélio/metabolismo , Humanos , Mucosa Respiratória/metabolismo , Mucosa Respiratória/virologia , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico
5.
BMC Pulm Med ; 20(1): 64, 2020 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-32171287

RESUMO

BACKGROUND: Matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) play important roles in the turnover of extracellular matrix and in the pathogenesis of idiopathic pulmonary fibrosis (IPF). This study aimed to determine the utility of circulating MMPs and TIMPs in distinguishing patients with IPF from controls and to explore associations between MMPs/TIMPs and measures of disease severity in patients with IPF. METHODS: The IPF cohort (n = 300) came from the IPF-PRO Registry, an observational multicenter registry of patients with IPF that was diagnosed or confirmed at the enrolling center in the past 6 months. Controls (n = 100) without known lung disease came from a population-based registry. Generalized linear models were used to compare circulating concentrations of MMPs 1, 2, 3, 7, 8, 9, 12, and 13 and TIMPs 1, 2, and 4 between patients with IPF and controls, and to investigate associations between circulating levels of these proteins and measures of IPF severity. Multivariable models were fit to identify the MMP/TIMPs that best distinguished patients with IPF from controls. RESULTS: All the MMP/TIMPs analyzed were present at significantly higher levels in patients with IPF compared with controls except for TIMP2. Multivariable analyses selected MMP8, MMP9 and TIMP1 as top candidates for distinguishing patients with IPF from controls. Higher concentrations of MMP7, MMP12, MMP13 and TIMP4 were significantly associated with lower diffusion capacity of the lung for carbon monoxide (DLCO) % predicted and higher composite physiologic index (worse disease). MMP9 was associated with the composite physiologic index. No MMP/TIMPs were associated with forced vital capacity % predicted. CONCLUSIONS: Circulating MMPs and TIMPs were broadly elevated among patients with IPF. Select MMP/TIMPs strongly associated with measures of disease severity. Our results identify potential MMP/TIMP targets for further development as disease-related biomarkers.


Assuntos
Fibrose Pulmonar Idiopática/sangue , Metaloproteinases da Matriz Secretadas/sangue , Inibidores Teciduais de Metaloproteinases/sangue , Idoso , Biomarcadores/sangue , Estudos de Casos e Controles , Feminino , Humanos , Fibrose Pulmonar Idiopática/patologia , Modelos Lineares , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Valor Preditivo dos Testes , Capacidade Vital
6.
Pulm Pharmacol Ther ; 59: 101839, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31518649

RESUMO

PURPOSE: Although safety and tolerability of approved antifibrotics has been reported extensively, little is known about their effects on weight. We analyzed predictors of weight change after one year of uninterrupted antifibrotic therapy in patients followed at our institution's interstitial lung disease clinic. METHODS/RESULTS: We identified 80 patients on antifibrotic therapy (44 pirfenidone/36 nintedanib) with at least one year of follow-up and no therapy interruptions. Thirty-five patients (44%) lost more than 5% of their baseline body weight, and 11 (19%) lost more than 10%. A higher proportion of patients on nintedanib experienced a clinically significant weight loss (>5%) versus pirfenidone (61% vs 30%, p = 0.005). Univariate and multivariate analyses identified nintedanib therapy and a higher composite physiologic index (CPI) as predictors of weight loss. CONCLUSIONS: Weight loss is common among IPF patients on antifibrotic therapy. Nintedanib therapy and more advanced disease were identified as predictors of weight loss in this population.


Assuntos
Fibrose Pulmonar Idiopática/tratamento farmacológico , Indóis/administração & dosagem , Piridonas/administração & dosagem , Redução de Peso/efeitos dos fármacos , Idoso , Anti-Inflamatórios não Esteroides/administração & dosagem , Anti-Inflamatórios não Esteroides/efeitos adversos , Feminino , Seguimentos , Humanos , Fibrose Pulmonar Idiopática/fisiopatologia , Indóis/efeitos adversos , Masculino , Pessoa de Meia-Idade , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/efeitos adversos , Piridonas/efeitos adversos , Índice de Gravidade de Doença
8.
J Immunol ; 196(1): 428-36, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26597012

RESUMO

Macrophage phagocytosis of particles and pathogens is an essential aspect of innate host defense. Phagocytic function requires cytoskeletal rearrangements that depend on the interaction between macrophage surface receptors, particulates/pathogens, and the extracellular matrix. In the present study we determine the role of a mechanosensitive ion channel, transient receptor potential vanilloid 4 (TRPV4), in integrating the LPS and matrix stiffness signals to control macrophage phenotypic change for host defense and resolution from lung injury. We demonstrate that active TRPV4 mediates LPS-stimulated murine macrophage phagocytosis of nonopsonized particles (Escherichia coli) in vitro and opsonized particles (IgG-coated latex beads) in vitro and in vivo in intact mice. Intriguingly, matrix stiffness in the range seen in inflamed or fibrotic lung is required to sensitize the TRPV4 channel to mediate the LPS-induced increment in macrophage phagocytosis. Furthermore, TRPV4 is required for the LPS induction of anti-inflammatory/proresolution cytokines. These findings suggest that signaling through TRPV4, triggered by changes in extracellular matrix stiffness, cooperates with LPS-induced signals to mediate macrophage phagocytic function and lung injury resolution. These mechanisms are likely to be important in regulating macrophage function in the context of pulmonary infection and fibrosis.


Assuntos
Lipopolissacarídeos/imunologia , Lesão Pulmonar/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Canais de Cátion TRPV/imunologia , Animais , Células Cultivadas , Citocinas/biossíntese , Citocinas/imunologia , Escherichia coli/imunologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Matriz Extracelular/metabolismo , Imunoglobulina G/imunologia , Lesão Pulmonar/patologia , Fenômenos Mecânicos , Camundongos , Camundongos Endogâmicos C57BL , Microesferas , Fibrose Pulmonar/imunologia , Transdução de Sinais/imunologia
9.
Am J Respir Cell Mol Biol ; 56(5): 667-679, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28459387

RESUMO

Numerous compounds have shown efficacy in limiting development of pulmonary fibrosis using animal models, yet few of these compounds have replicated these beneficial effects in clinical trials. Given the challenges associated with performing clinical trials in patients with idiopathic pulmonary fibrosis (IPF), it is imperative that preclinical data packages be robust in their analyses and interpretations to have the best chance of selecting promising drug candidates to advance to clinical trials. The American Thoracic Society has convened a group of experts in lung fibrosis to discuss and formalize recommendations for preclinical assessment of antifibrotic compounds. The panel considered three major themes (choice of animal, practical considerations of fibrosis modeling, and fibrotic endpoints for evaluation). Recognizing the need for practical considerations, we have taken a pragmatic approach. The consensus view is that use of the murine intratracheal bleomycin model in animals of both genders, using hydroxyproline measurements for collagen accumulation along with histologic assessments, is the best-characterized animal model available for preclinical testing. Testing of antifibrotic compounds in this model is recommended to occur after the acute inflammatory phase has subsided (generally after Day 7). Robust analyses may also include confirmatory studies in human IPF specimens and validation of results in a second system using in vivo or in vitro approaches. The panel also strongly encourages the publication of negative results to inform the lung fibrosis community. These recommendations are for preclinical therapeutic evaluation only and are not intended to dissuade development of emerging technologies to better understand IPF pathogenesis.


Assuntos
Congressos como Assunto , Modelos Animais de Doenças , Fibrose Pulmonar/terapia , Sociedades Médicas , Animais , Determinação de Ponto Final , Feminino , Humanos , Masculino , Organismos Geneticamente Modificados , Reprodutibilidade dos Testes
10.
J Biol Chem ; 291(12): 6083-95, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26763235

RESUMO

Pro-fibrotic mesenchymal cells are known to be the key effector cells of fibroproliferative disease, but the specific matrix signals and the induced cellular responses that drive the fibrogenic phenotype remain to be elucidated. The key mediators of the fibroblast fibrogenic phenotype were characterized using a novel assay system that measures fibroblast behavior in response to actual normal and fibrotic lung tissue. Using this system, we demonstrate that normal lung promotes fibroblast motility and polarization, while fibrotic lung immobilizes the fibroblast and promotes myofibroblast differentiation. These context-specific phenotypes are surprisingly both mediated by myosin II. The role of myosin II is supported by the observation of an increase in myosin phosphorylation and a change in intracellular distribution in fibroblasts on fibrotic lung, as compared with normal lung. Moreover, loss of myosin II activity has opposing effects on protrusive activity in fibroblasts on normal and fibrotic lung. Loss of myosin II also selectively inhibits myofibroblast differentiation in fibroblasts on fibrotic lung. Importantly, these findings are recapitulated by varying the matrix stiffness of polyacrylamide gels in the range of normal and fibrotic lung tissue. Comparison of the effects of myosin inhibition on lung tissue with that of polyacrylamide gels suggests that matrix fiber organization drives the fibroblast phenotype under conditions of normal/soft lung, while matrix stiffness drives the phenotype under conditions of fibrotic/stiff lung. This work defines novel roles for myosin II as a key regulatory effector molecule of the pro-fibrotic phenotype, in response to biophysical properties of the matrix.


Assuntos
Fibroblastos/fisiologia , Miosina Tipo II/fisiologia , Fibrose Pulmonar/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Movimento Celular , Polaridade Celular , Forma Celular , Matriz Extracelular/fisiologia , Feminino , Humanos , Pulmão/metabolismo , Pulmão/patologia , Camundongos Endogâmicos C57BL , Fenótipo , Fibrose Pulmonar/patologia
11.
J Neurooncol ; 131(3): 449-458, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27858267

RESUMO

The circulating levels of soluble tumor necrosis factor receptor-1 (sTNF-R1) and sTNF-R2 are altered in numerous diseases, including several types of cancer. Correlations with the risk of progression in some cancers, as well as systemic manifestations of the disease and therapeutic side-effects, have been described. However, there is very little information on the levels of these soluble receptors in glioblastoma (GBM). Here, we report on an exploratory retrospective study of the levels of sTNF-Rs in the vascular circulation of patients with GBM. Banked samples were obtained from 112 GBM patients (66 untreated, newly-diagnosed patients and 46 with recurrent disease) from two institutions. The levels of sTNF-R1 in the plasma were significantly lower in patients with newly-diagnosed or recurrent GBM than apparently healthy individuals and correlated with the intensity of expression of TNF-R1 on the tumor-associated endothelial cells (ECs) in the corresponding biopsies. Elevated levels of sTNF-R1 in patients with recurrent, but not newly-diagnosed GBM, were significantly associated with a shorter survival, independent of age (p = 0.02) or steroid medication. In contrast, the levels of circulating sTNF-R2 were significantly higher in recurrent GBM than healthy individuals and there was no significant correlation with expression of TNF-R2 on the tumor-associated ECs or survival time. The results indicate that larger, prospective studies are warranted to determine the predictive value of the levels of sTNF-R1 in patients with recurrent GBM and the factors that regulate the levels of sTNF-Rs in the circulation in GBM patients.


Assuntos
Glioblastoma/sangue , Recidiva Local de Neoplasia/sangue , Receptores Tipo II do Fator de Necrose Tumoral/sangue , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Análise de Sobrevida , Adulto Jovem
13.
J Biol Chem ; 289(18): 12791-804, 2014 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-24644284

RESUMO

The urokinase-type plasminogen activator receptor (uPAR) is a glycosylphosphatidylinositol-linked membrane protein with no cytosolic domain that localizes to lipid raft microdomains. Our laboratory and others have documented that lung fibroblasts from patients with idiopathic pulmonary fibrosis (IPF) exhibit a hypermotile phenotype. This study was undertaken to elucidate the molecular mechanism whereby uPAR ligation with its cognate ligand, urokinase, induces a motile phenotype in human lung fibroblasts. We found that uPAR ligation with the urokinase receptor binding domain (amino-terminal fragment) leads to enhanced migration of fibroblasts on fibronectin in a protease-independent, lipid raft-dependent manner. Ligation of uPAR with the amino-terminal fragment recruited α5ß1 integrin and the acylated form of the Src family kinase, Fyn, to lipid rafts. The biological consequences of this translocation were an increase in fibroblast motility and a switch of the integrin-initiated signal pathway for migration away from the lipid raft-independent focal adhesion kinase pathway and toward a lipid raft-dependent caveolin-Fyn-Shc pathway. Furthermore, an integrin homologous peptide as well as an antibody that competes with ß1 for uPAR binding have the ability to block this effect. In addition, its relative insensitivity to cholesterol depletion suggests that the interactions of α5ß1 integrin and uPAR drive the translocation of α5ß1 integrin-acylated Fyn signaling complexes into lipid rafts upon uPAR ligation through protein-protein interactions. This signal switch is a novel pathway leading to the hypermotile phenotype of IPF patient-derived fibroblasts, seen with uPAR ligation. This uPAR dependent, fibrotic matrix-selective, and profibrotic fibroblast phenotype may be amenable to targeted therapeutics designed to ameliorate IPF.


Assuntos
Movimento Celular , Fibroblastos/metabolismo , Integrina alfa5beta1/metabolismo , Microdomínios da Membrana/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Western Blotting , Caveolinas/genética , Caveolinas/metabolismo , Células Cultivadas , Fibroblastos/citologia , Fibronectinas/metabolismo , Humanos , Fibrose Pulmonar Idiopática/sangue , Fibrose Pulmonar Idiopática/metabolismo , Fibrose Pulmonar Idiopática/patologia , Integrina alfa5beta1/genética , Camundongos , Microscopia de Fluorescência , Ligação Proteica , Proteínas Proto-Oncogênicas c-fyn/genética , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Interferência de RNA , Receptores de Ativador de Plasminogênio Tipo Uroquinase/sangue , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Índice de Gravidade de Doença , Proteínas Adaptadoras da Sinalização Shc/genética , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Transdução de Sinais , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
14.
Am J Pathol ; 182(5): 1572-84, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23499373

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease whose underlying molecular mechanisms are largely unknown. Herein, we show that focal adhesion kinase-related nonkinase (FRNK) plays a key role in limiting the development of lung fibrosis. Loss of FRNK function in vivo leads to increased lung fibrosis in an experimental mouse model. The increase in lung fibrosis is confirmed at the histological, biochemical, and physiological levels. Concordantly, loss of FRNK function results in increased fibroblast migration and myofibroblast differentiation and activation of signaling proteins that drive these phenotypes. FRNK-deficient murine lung fibroblasts also have an increased capacity to produce and contract matrix proteins. Restoration of FRNK expression in vivo and in vitro reverses these profibrotic phenotypes. These data demonstrate the multiple antifibrotic actions of FRNK. More important, FRNK expression is down-regulated in human IPF, and down-regulation of FRNK in normal human lung fibroblasts recapitulates the profibrotic phenotype seen in FRNK-deficient cells. The effect of loss and gain of FRNK in the experimental model, when taken together with its down-regulation in human IPF, suggests that FRNK acts as an endogenous negative regulator of lung fibrosis by repressing multiple profibrotic responses.


Assuntos
Proteínas Tirosina Quinases/metabolismo , Fibrose Pulmonar/enzimologia , Fibrose Pulmonar/patologia , Adulto , Animais , Bleomicina , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Humanos , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Pulmão/patologia , Camundongos , Camundongos Endogâmicos C57BL , Miofibroblastos/efeitos dos fármacos , Miofibroblastos/enzimologia , Miofibroblastos/patologia , Proteínas Tirosina Quinases/deficiência , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta1/farmacologia
17.
Ann Am Thorac Soc ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38843487

RESUMO

OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is the third leading cause of death worldwide. Our previous studies have identified that nocturnal hypoxemia causes skeletal muscle loss (i.e. sarcopenia) in in vitro models of COPD. RATIONALE: We aimed to extend our preclinical mechanistic findings by analyzing a large sleep registry to determine whether nocturnal hypoxemia is associated with sarcopenia in COPD patients. METHODS: Sleep studies from COPD patients (n=479) and control subjects without COPD (n=275) were analyzed. Patients with obstructive sleep apnea (OSA), as defined by apnea hypopnea index >5, were excluded. Pectoralis muscle cross sectional area (PMcsa) was quantified using CT scans performed within one year of the sleep study. We defined sarcopenia as less than the lowest 20% residuals for PMcsa of controls, which was adjusted for age, BMI, and stratified by sex. Youden's optimal cutpoint criteria was used to predict sarcopenia based on mean oxygen saturation (mean SaO2) during sleep. Additional measures of nocturnal hypoxemia were analyzed. Pectoralis muscle index (PMI) was defined as PMcsa normalized to BMI. RESULTS: On average, COPD males had 16.6% lower PMI than control males (1.41+0.44 vs 1.69+0.56 cm2/BMI, p<0.001), while COPD females had 9.4% lower PMI than control females (0.96+0.27 vs 1.06+0.33 cm2/BMI, p<0.001). COPD males with nocturnal hypoxemia had a 9.5% decrease in PMI versus COPD with normal O2 (1.33+0.39 vs 1.47+0.46 cm2/BMI, p<0.05), and 23.6% decrease compared to controls (1.33+0.39 vs 1.74+0.56 cm2/BMI, p<0.001). COPD females with nocturnal hypoxemia had a 11.2% decrease versus COPD with normal O2 (0.87+0.26 vs 0.98+0.28 cm2/BMI, p<0.05), and 17.9% decrease compared to controls (0.87+0.26 vs 1.06+0.33 cm2/BMI, p<0.001). These findings were largely replicated using multiple measures of nocturnal hypoxemia. CONCLUSIONS: We defined sarcopenia in the pectoralis muscle using residuals that take into account age, BMI, and sex. We found that COPD patients have lower PMI than non-COPD patients, and that nocturnal hypoxemia was associated with an additional decrease in the PMI of COPD patients. Additional prospective analyses are needed to determine a protective threshold of oxygen saturation to prevent or reverse sarcopenia due to nocturnal hypoxemia in COPD.

18.
Am J Respir Crit Care Med ; 186(1): 88-95, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22561965

RESUMO

RATIONALE: Animal and human studies support the importance of the coagulation cascade in pulmonary fibrosis. OBJECTIVES: In a cohort of subjects with progressive idiopathic pulmonary fibrosis (IPF), we tested the hypothesis that treatment with warfarin at recognized therapeutic doses would reduce rates of mortality, hospitalization, and declines in FVC. METHODS: This was a double-blind, randomized, placebo-controlled trial of warfarin targeting an international normalized ratio of 2.0 to 3.0 in patients with IPF. Subjects were randomized in a 1:1 ratio to warfarin or matching placebo for a planned treatment period of 48 weeks. International normalized ratios were monitored using encrypted home point-of-care devices that allowed blinding of study therapy. MEASUREMENTS AND MAIN RESULTS: The primary outcome measure was the composite outcome of time to death, hospitalization (nonbleeding, nonelective), or a 10% or greater absolute decline in FVC. Due to a low probability of benefit and an increase in mortality observed in the subjects randomized to warfarin (14 warfarin versus 3 placebo deaths; P = 0.005) an independent Data and Safety Monitoring Board recommended stopping the study after 145 of the planned 256 subjects were enrolled (72 warfarin, 73 placebo). The mean follow-up was 28 weeks. CONCLUSIONS: This study did not show a benefit for warfarin in the treatment of patients with progressive IPF. Treatment with warfarin was associated with an increased risk of mortality in an IPF population who lacked other indications for anticoagulation.


Assuntos
Anticoagulantes/uso terapêutico , Fibrose Pulmonar Idiopática/tratamento farmacológico , Varfarina/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Método Duplo-Cego , Feminino , Hospitalização/estatística & dados numéricos , Humanos , Fibrose Pulmonar Idiopática/mortalidade , Fibrose Pulmonar Idiopática/fisiopatologia , Coeficiente Internacional Normatizado , Masculino , Pessoa de Meia-Idade , Falha de Tratamento
20.
J Am Coll Cardiol ; 80(7): 697-718, 2022 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-35953136

RESUMO

BACKGROUND: PVDOMICS (Pulmonary Vascular Disease Phenomics) is a precision medicine initiative to characterize pulmonary vascular disease (PVD) using deep phenotyping. PVDOMICS tests the hypothesis that integration of clinical metrics with omic measures will enhance understanding of PVD and facilitate an updated PVD classification. OBJECTIVES: The purpose of this study was to describe clinical characteristics and transplant-free survival in the PVDOMICS cohort. METHODS: Subjects with World Symposium Pulmonary Hypertension (WSPH) group 1-5 PH, disease comparators with similar underlying diseases and mild or no PH and healthy control subjects enrolled in a cross-sectional study. PH groups, comparators were compared using standard statistical tests including log-rank tests for comparing time to transplant or death. RESULTS: A total of 1,193 subjects were included. Multiple WSPH groups were identified in 38.9% of PH subjects. Nocturnal desaturation was more frequently observed in groups 1, 3, and 4 PH vs comparators. A total of 50.2% of group 1 PH subjects had ground glass opacities on chest computed tomography. Diffusing capacity for carbon monoxide was significantly lower in groups 1-3 PH than their respective comparators. Right atrial volume index was higher in WSPH groups 1-4 than comparators. A total of 110 participants had a mean pulmonary artery pressure of 21-24 mm Hg. Transplant-free survival was poorest in group 3 PH. CONCLUSIONS: PVDOMICS enrolled subjects across the spectrum of PVD, including mild and mixed etiology PH. Novel findings include low diffusing capacity for carbon monoxide and enlarged right atrial volume index as shared features of groups 1-3 and 1-4 PH, respectively; unexpected, frequent presence of ground glass opacities on computed tomography; and sleep alterations in group 1 PH, and poorest survival in group 3 PH. PVDOMICS will facilitate a new understanding of PVD and refine the current PVD classification. (Pulmonary Vascular Disease Phenomics Program PVDOMICS [PVDOMICS]; NCT02980887).


Assuntos
Hipertensão Pulmonar , Doenças Vasculares , Monóxido de Carbono , Estudos Transversais , Humanos , Hipertensão Pulmonar/etiologia , Circulação Pulmonar , Doenças Vasculares/complicações , Doenças Vasculares/diagnóstico , Doenças Vasculares/cirurgia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA