Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Lung Cell Mol Physiol ; 324(5): L639-L651, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36648147

RESUMO

Idiopathic pulmonary fibrosis is increasingly associated with nerve-driven processes and endogenous innate immune ligands such as mitochondrial DNA (mtDNA). Interestingly, a connection between these entities has not been explored. Here, we report that noradrenaline (NA) derived from the lung's adrenergic nerve supply drives α-smooth muscle actin (αSMA)-expressing fibroblast accumulation via mechanisms involving α1 adrenoreceptors and mtDNA. Using the bleomycin model, we compared ablation of the lung's adrenergic nerve supply with surgical adrenal resection and found that NA derived from local but not adrenal sources contributes to experimentally induced lung fibrosis and the emergence of an αSMA+ve fibroblast population expressing adrenoreceptor α-1D (ADRA1D). Therapeutic delivery of the α1 adrenoreceptor antagonist terazosin reversed these changes and suppressed extracellular mtDNA accumulation. Cultured normal human lung fibroblasts displayed α1 adrenoreceptors and in response to costimulation with TGFß1 and NA adopted ACTA2 expression and extracellular mtDNA release. These findings were opposed by terazosin. Evaluation of a previously studied IPF cohort revealed that patients prescribed α1 adrenoreceptor antagonists for nonpulmonary indications demonstrated improved survival and reduced concentrations of plasma mtDNA. Our observations link nerve-derived NA, α1 adrenoreceptors, extracellular mtDNA, and lung fibrogenesis in mouse models, cultured cells, and humans with IPF. Further study of this neuroinnate connection may yield new avenues for investigation in the clinical and basic science realms.


Assuntos
DNA Mitocondrial , Fibrose Pulmonar Idiopática , Camundongos , Animais , Humanos , DNA Mitocondrial/genética , DNA Mitocondrial/metabolismo , Fibrose Pulmonar Idiopática/metabolismo , Pulmão/metabolismo , Transdução de Sinais , Fibroblastos/metabolismo , Bleomicina/farmacologia , Adrenérgicos/metabolismo , Adrenérgicos/farmacologia
2.
J Clin Invest ; 131(1)2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33393489

RESUMO

Fibrosis is a macrophage-driven process of uncontrolled extracellular matrix accumulation. Neuronal guidance proteins such as netrin-1 promote inflammatory scarring. We found that macrophage-derived netrin-1 stimulates fibrosis through its neuronal guidance functions. In mice, fibrosis due to inhaled bleomycin engendered netrin-1-expressing macrophages and fibroblasts, remodeled adrenergic nerves, and augmented noradrenaline. Cell-specific knockout mice showed that collagen accumulation, fibrotic histology, and nerve-associated endpoints required netrin-1 of macrophage but not fibroblast origin. Adrenergic denervation; haploinsufficiency of netrin-1's receptor, deleted in colorectal carcinoma; and therapeutic α1 adrenoreceptor antagonism improved collagen content and histology. An idiopathic pulmonary fibrosis (IPF) lung microarray data set showed increased netrin-1 expression. IPF lung tissues were enriched for netrin-1+ macrophages and noradrenaline. A longitudinal IPF cohort showed improved survival in patients prescribed α1 adrenoreceptor blockade. This work showed that macrophages stimulate lung fibrosis via netrin-1-driven adrenergic processes and introduced α1 blockers as a potentially new fibrotic therapy.


Assuntos
Pulmão/inervação , Pulmão/metabolismo , Macrófagos/metabolismo , Netrina-1/metabolismo , Fibrose Pulmonar/metabolismo , Animais , Bleomicina/efeitos adversos , Bleomicina/farmacologia , Feminino , Pulmão/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Transgênicos , Netrina-1/genética , Norepinefrina/genética , Norepinefrina/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/genética , Fibrose Pulmonar/patologia
3.
Life Sci Alliance ; 2(3)2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31085559

RESUMO

TGF-ß1 is a critical mediator of tissue fibrosis in health and disease whose effects are augmented by chitinase 1 (CHIT1). However, the mechanisms that CHIT1 uses to regulate TGF-ß1-mediated fibrotic responses have not been defined. Here, we demonstrate that CHIT1 enhances TGF-ß1-stimulated fibrotic cellular and tissue responses and TGF-ß1 signaling. Importantly, we also demonstrate that these effects are mediated by the ability of CHIT1 to inhibit TGF-ß1 induction of its feedback inhibitor, SMAD7. CHIT1 also interacted with TGF-ß receptor associated protein 1 (TGFBRAP1) and forkhead box O3 (FOXO3) with TGFBRAP1 playing a critical role in CHIT1 enhancement of TGF-ß1 signaling and effector responses and FOXO3 playing a critical role in TGF-ß1 induction of SMAD7. These pathways were disease relevant because the levels of CHIT1 were increased and inversely correlated with SMAD7 in tissues from patients with idiopathic pulmonary fibrosis or scleroderma-associated interstitial lung disease. These studies demonstrate that CHIT1 regulates TGF-ß1/SMAD7 axis via TGFBRAP1 and FOXO3 and highlight the importance of these pathways in the pathogenesis of pulmonary fibrosis.


Assuntos
Proteína Forkhead Box O3/metabolismo , Hexosaminidases/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fibrose Pulmonar/etiologia , Fibrose Pulmonar/metabolismo , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Hexosaminidases/metabolismo , Humanos , Imuno-Histoquímica , Regiões Promotoras Genéticas , Fibrose Pulmonar/patologia , RNA Interferente Pequeno/genética , Transdução de Sinais
4.
JCI Insight ; 2(24)2017 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-29263297

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a fatal disease of unknown etiology characterized by a compositionally and mechanically altered extracellular matrix. Poor understanding of the origin of α-smooth muscle actin (α-SMA) expressing myofibroblasts has hindered curative therapies. Though proposed as a source of myofibroblasts in mammalian tissues, identification of microvascular pericytes (PC) as contributors to α-SMA-expressing populations in human IPF and the mechanisms driving this accumulation remain unexplored. Here, we demonstrate enhanced detection of α-SMA+ cells coexpressing the PC marker neural/glial antigen 2 in the human IPF lung. Isolated human PC cultured on decellularized IPF lung matrices adopt expression of α-SMA, demonstrating that these cells undergo phenotypic transition in response to direct contact with the extracellular matrix (ECM) of the fibrotic human lung. Using potentially novel human lung-conjugated hydrogels with tunable mechanical properties, we decoupled PC responses to matrix composition and stiffness to show that α-SMA+ PC accumulate in a mechanosensitive manner independent of matrix composition. PC activated with TGF-ß1 remodel the normal lung matrix, increasing tissue stiffness to facilitate the emergence of α-SMA+ PC via MKL-1/MTRFA mechanotranduction. Nintedanib, a tyrosine-kinase inhibitor approved for IPF treatment, restores the elastic modulus of fibrotic lung matrices to reverse the α-SMA+ phenotype. This work furthers our understanding of the role that microvascular PC play in the evolution of IPF, describes the creation of an ex vivo platform that advances the study of fibrosis, and presents a potentially novel mode of action for a commonly used antifibrotic therapy that has great relevance for human disease.


Assuntos
Fibrose Pulmonar Idiopática/patologia , Miofibroblastos/fisiologia , Pericitos/fisiologia , Actinas/metabolismo , Antígenos/metabolismo , Células Cultivadas , Elasticidade , Inibidores Enzimáticos/farmacologia , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Humanos , Fibrose Pulmonar Idiopática/metabolismo , Indóis/farmacologia , Pulmão/metabolismo , Mecanotransdução Celular/efeitos dos fármacos , Mecanotransdução Celular/fisiologia , Metaloproteases/biossíntese , Miofibroblastos/metabolismo , Pericitos/efeitos dos fármacos , Fenótipo , Proteoglicanas/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
5.
FASEB J ; 30(12): 4056-4070, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27609773

RESUMO

Pulmonary fibrosis is a progressive and often fatal condition that is believed to be partially orchestrated by macrophages. Mechanisms that control migration of these cells into and within the lung remain undefined. We evaluated the contributions of the semaphorin receptor, plexin C1 (PLXNC1), and the exocytic calcium sensor, synaptotagmin 7 (Syt7), in these processes. We evaluated the role of PLXNC1 in macrophage migration by using Boyden chambers and scratch tests, characterized its contribution to experimentally induced lung fibrosis in mice, and defined the mechanism for our observations. Our findings reveal that relative to control participants, patients with idiopathic pulmonary fibrosis demonstrate excessive monocyte migration and underexpression of PLXNC1 in the lungs and circulation, a finding that is recapitulated in the setting of scleroderma-related interstitial lung disease. Relative to wild type, PLXNC1-/- mouse macrophages are excessively migratory, and PLXNC1-/- mice show exacerbated collagen accumulation in response to either inhaled bleomycin or inducible lung targeted TGF-ß1 overexpression. These findings are ameliorated by replacement of PLXNC1 on bone marrow-derived cells or by genetic deletion of Syt7. These data demonstrate the previously unrecognized observation that PLXNC1 deficiency permits Syt7-mediated macrophage migration and enhances mammalian lung fibrosis.-Peng, X., Moore, M., Mathur, A., Zhou, Y., Sun, H., Gan, Y., Herazo-Maya, J. D., Kaminski, N., Hu, X., Pan, H., Ryu, C., Osafo-Addo, A., Homer, R. J., Feghali-Bostwick, C., Fares, W. H., Gulati, M., Hu, B., Lee, C.-G., Elias, J. A., Herzog, E. L. Plexin C1 deficiency permits synaptotagmin 7-mediated macrophage migration and enhances mammalian lung fibrosis.


Assuntos
Macrófagos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fibrose Pulmonar/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Virais/metabolismo , Sinaptotagminas/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Pulmão/metabolismo , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Fibrose Pulmonar/genética , Receptores de Superfície Celular/deficiência , Receptores Virais/deficiência , Fator de Crescimento Transformador beta1/metabolismo
6.
Arthritis Rheumatol ; 68(5): 1251-61, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26749424

RESUMO

OBJECTIVE: Fibrocytes are collagen-producing leukocytes that accumulate in patients with systemic sclerosis (SSc; scleroderma)-related interstitial lung disease (ILD) via unknown mechanisms that have been associated with altered expression of neuroimmune proteins. The extracellular matrix (ECM) influences cellular phenotypes. However, a relationship between the lung ECM and fibrocytes in SSc has not been explored. The aim of this study was to use a novel translational platform based on decellularized human lungs to determine whether the lung ECM of patients with scleroderma controls the development of fibrocytes from peripheral blood mononuclear cells. METHODS: We performed biomechanical evaluation of decellularized scaffolds prepared from lung explants from healthy control subjects and patients with scleroderma, using tensile testing and biochemical and proteomic analysis. Cells obtained from healthy controls and patients with SSc-related ILD were cultured on these scaffolds, and CD45+pro-ColIα1+ cells meeting the criteria for fibrocytes were quantified. The contribution of the neuromolecule netrin-1 to fibrosis was assessed using neutralizing antibodies in this system and by administering bleomycin via inhalation to netrin-1(+/-) mice. RESULTS: Compared with control lung scaffolds, lung scaffolds from patients with SSc-related ILD showed aberrant anatomy, enhanced stiffness, and abnormal ECM composition. Culture of control cells in lung scaffolds from patients with SSc-related ILD increased production of pro-ColIα1+ cells, which was stimulated by enhanced stiffness and abnormal ECM composition. Cells from patients with SSc-related ILD demonstrated increased pro-ColIα1 responsiveness to lung scaffolds from scleroderma patients but not enhanced stiffness. Enhanced detection of netrin-1-expressing CD14(low) cells in patients with SSc-related ILD was observed, and antibody-mediated netrin-1 neutralization attenuated detection of CD45+pro-ColIα1+ cells in all settings. Netrin-1(+/-) mice were protected against bleomycin-induced lung fibrosis and fibrocyte accumulation. CONCLUSION: Factors present in the lung matrices of patients with scleroderma regulate fibrocyte accumulation via a netrin-1-dependent pathway. Netrin-1 regulates bleomycin-induced pulmonary fibrosis in mice. Netrin-1 might be a novel therapeutic target in SSc-related ILD.


Assuntos
Doenças Pulmonares Intersticiais/metabolismo , Pulmão/metabolismo , Fatores de Crescimento Neural/metabolismo , Fibrose Pulmonar/metabolismo , Escleroderma Sistêmico/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Antibióticos Antineoplásicos/toxicidade , Anticorpos Neutralizantes/farmacologia , Fenômenos Biomecânicos , Bleomicina/toxicidade , Estudos de Casos e Controles , Diferenciação Celular , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Fibrose , Citometria de Fluxo , Imunofluorescência , Heterozigoto , Humanos , Antígenos Comuns de Leucócito/metabolismo , Leucócitos Mononucleares , Pulmão/efeitos dos fármacos , Pulmão/patologia , Doenças Pulmonares Intersticiais/etiologia , Doenças Pulmonares Intersticiais/patologia , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Varredura , Fatores de Crescimento Neural/antagonistas & inibidores , Fatores de Crescimento Neural/genética , Netrina-1 , Proteômica , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Escleroderma Sistêmico/complicações , Alicerces Teciduais , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/genética
7.
Cancer Res ; 75(3): 487-96, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25511377

RESUMO

The prototypic chitinase-like protein Chi3l1 is induced in cancers and portends a poor prognosis, but whether it contributes to cancer progression is unknown. To address this gap in knowledge, we investigated the production of Chi3l1 in melanoma lung metastases. We found that Chi3l1 was induced during pulmonary melanoma metastasis and that this induction was regulated by the semaphorin Sema7a, interacting in stimulatory or inhibitory ways with its ß1 integrin or Plexin C1 receptors, respectively. In mouse strains with genetic deletions of Chi3l1 or Sema7a, there was a significant reduction in pulmonary metastasis. Notably, antiserum raised against Chi3l1 or Sema7a phenocopied the reduction produced by genetic deletions. Melanoma lung metastasis was also decreased in the absence of IL13Rα2, a recently identified receptor for Chi3l1, consistent with a key role for Chi3l1 in melanoma spread. We confirmed roles for Sema7a and Chi3l1 in pulmonary metastasis of EMT6 breast cancer cells. Taken together, our studies establish a novel pathway through which Sem7a and its receptors regulate Chi3l1, revealing a host axis involving IL13Rα2 that plays a critical role in generating a pulmonary microenvironment that is critical to license metastasis.


Assuntos
Antígenos CD/metabolismo , Regulação Neoplásica da Expressão Gênica , Glicoproteínas/metabolismo , Neoplasias Pulmonares/secundário , Melanoma/patologia , Semaforinas/metabolismo , Animais , Linhagem Celular Tumoral , Proteína 1 Semelhante à Quitinase-3 , Deleção de Genes , Inativação Gênica , Imuno-Histoquímica , Neoplasias Pulmonares/metabolismo , Melanoma/metabolismo , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
8.
Sci Transl Med ; 6(240): 240ra76, 2014 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-24920662

RESUMO

Epithelial injury, alternative macrophage accumulation, and fibroproliferation coexist in the lungs of patients with idiopathic pulmonary fibrosis (IPF). Chitinase 3-like 1 (CHI3L1) is a prototypic chitinase-like protein that has been retained over species and evolutionary time. However, the regulation of CHI3L1 in IPF and its ability to regulate injury and/or fibroproliferative repair have not been fully defined. We demonstrated that CHI3L1 levels were elevated in patients with IPF. High levels of CHI3L1 are associated with progression--as defined by lung transplantation or death--and with scavenger receptor-expressing circulating monocytes in an ambulatory IPF population. In preterminal acute exacerbations of IPF, CHI3L1 levels were reduced and associated with increased levels of apoptosis. We also demonstrated that in bleomycin-treated mice, CHI3L1 expression was acutely and transiently decreased during the injury phase and returned toward and eventually exceeded baseline levels during the fibrotic phase. In this model, CHI3L1 played a protective role in injury by ameliorating inflammation and cell death, and a profibrotic role in the repair phase by augmenting alternative macrophage activation, fibroblast proliferation, and matrix deposition. Using three-dimensional culture system of a human fibroblast cell line, we found that CHI3L1 is sufficient to induce low grade myofibroblast transformation. In combination, these studies demonstrate that CHI3L1 is stimulated in IPF, where it represents an attempt to diminish injury and induce repair. They also demonstrate that high levels of CHI3L1 are associated with disease progression in ambulatory patients and that a failure of the CHI3L1 antiapoptotic response might contribute to preterminal disease exacerbations.


Assuntos
Adipocinas/metabolismo , Lectinas/metabolismo , Pulmão/citologia , Fibrose Pulmonar/metabolismo , Adipocinas/genética , Animais , Apoptose/genética , Apoptose/fisiologia , Transplante de Medula Óssea , Proliferação de Células , Proteína 1 Semelhante à Quitinase-3 , Ensaio de Imunoadsorção Enzimática , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Citometria de Fluxo , Humanos , Marcação In Situ das Extremidades Cortadas , Lectinas/genética , Camundongos , Camundongos Knockout , Fibrose Pulmonar/genética
9.
Am J Physiol Lung Cell Mol Physiol ; 306(6): L463-75, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24337923

RESUMO

Creation of bioartificial organs has been enhanced by the development of strategies involving decellularized mammalian lung. Because fibroblasts critically support lung function through a number of mechanisms, study of these cells in the context of the decellularized lung has the potential to improve the structure and function of tissue-engineered lungs. We characterized the engraftment and survival of a mouse fibroblast cell line in decellularized rat lung slices and found a time-dependent increase in cell numbers assessed by hematoxylin and eosin staining, cell proliferation assessed by Ki67 staining, and minimal cell death assessed by TUNEL staining. We developed a repopulation index to allow quantification of cell survival that accounts for variation in cell density throughout the seeded scaffold. We then applied this method to the study of mouse lung scaffolds and found that decellularization of presliced mouse lungs produced matrices with preserved alveolar architecture and proteinaceous components including fibronectin, collagens I and IV, laminin, and elastin. Treatment with a ß1-integrin-neutralizing antibody significantly reduced the repopulation index after 24 h of culture. Treatment with focal adhesion kinase (FAK) inhibitor and extracellular signal-regulated kinase (ERK) inhibitor further reduced initial repopulation scores while treatment with AKT inhibitor increased initial scores. Rho-associated kinase inhibitor had no discernible effect. These data indicate that initial adhesion and survival of mouse fibroblasts in the decellularized mouse lung occur in a ß1-integrin-dependent, FAK/ERK-dependent manner that is opposed by AKT.


Assuntos
Órgãos Bioartificiais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fibroblastos/transplante , Quinase 1 de Adesão Focal/metabolismo , Integrina beta1/metabolismo , Pulmão/fisiologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Adesão Celular/fisiologia , Linhagem Celular , Proliferação de Células , Sobrevivência Celular , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fibroblastos/fisiologia , Quinase 1 de Adesão Focal/antagonistas & inibidores , Integrina beta1/imunologia , Pulmão/citologia , Camundongos , Fosforilação , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Ratos , Engenharia Tecidual/métodos , Alicerces Teciduais , Quinases Associadas a rho/antagonistas & inibidores
10.
Am J Respir Crit Care Med ; 187(2): 180-8, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23220917

RESUMO

RATIONALE: Lymphocytes are increasingly associated with idiopathic pulmonary fibrosis (IPF). Semaphorin 7a (Sema 7a) participates in lymphocyte activation. OBJECTIVES: To define the relationship between Sema 7a and lymphocytes in IPF. METHODS: We characterized the significance of Sema 7a+ lymphocytes in humans with IPF and in a mouse model of lung fibrosis caused by lung-targeted, transgenic overexpression of TGF-ß1. We determined the site of Sema 7a expression in human and murine lungs and circulation and used adoptive transfer approaches to define the relevance of lymphocytes coexpressing Sema7a and the markers CD19, CD4, or CD4+CD25+FoxP3+ in TGF-ß1-induced murine lung fibrosis. MEASUREMENTS AND MAIN RESULTS: Subjects with IPF show expression of Sema 7a on lung CD4+ cells and circulating CD4+ or CD19+ cells. Sema 7a expression is increased on CD4+ cells and CD4+CD25+FoxP3+ regulatory T cells, but not CD19+ cells, in subjects with progressive IPF. Sema 7a is expressed on lymphocytes expressing CD4 but not CD19 in the lungs and spleen of TGF-ß1-transgenic mice. Sema 7a expressing bone marrow-derived cells induce lung fibrosis and alter the production of T-cell mediators, including IFN-γ, IL-4, IL-17A, and IL-10. These effects require CD4 but not CD19. In comparison to Sema 7a-CD4+CD25+FoxP3+ cells, Sema7a+CD4+CD25+FoxP3+ cells exhibit reduced expression of regulatory genes such as IL-10, and adoptive transfer of these cells induces fibrosis and remodeling in the TGF-ß1-exposed murine lung. CONCLUSIONS: Sema 7a+CD4+CD25+FoxP3+ regulatory T cells are associated with disease progression in subjects with IPF and induce fibrosis in the TGF-ß1-exposed murine lung.


Assuntos
Antígenos CD/fisiologia , Fibrose Pulmonar Idiopática/etiologia , Semaforinas/fisiologia , Linfócitos T Reguladores/fisiologia , Fator de Crescimento Transformador beta1/fisiologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/fisiologia , Modelos Animais de Doenças , Humanos , Fibrose Pulmonar Idiopática/imunologia , Fibrose Pulmonar Idiopática/fisiopatologia , Interleucina-10/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA