Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 44(3): 582-596, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26921108

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a devastating lung disorder with increasing incidence. Mitochondrial oxidative stress in alveolar macrophages is directly linked to pulmonary fibrosis. Mitophagy, the selective engulfment of dysfunctional mitochondria by autophagasomes, is important for cellular homeostasis and can be induced by mitochondrial oxidative stress. Here, we show Akt1 induced macrophage mitochondrial reactive oxygen species (ROS) and mitophagy. Mice harboring a conditional deletion of Akt1 in macrophages (Akt1(-/-)Lyz2-cre) and Park2(-/-) mice had impaired mitophagy and reduced active transforming growth factor-ß1 (TGF-ß1). Although Akt1 increased TGF-ß1 expression, mitophagy inhibition in Akt1-overexpressing macrophages abrogated TGF-ß1 expression and fibroblast differentiation. Importantly, conditional Akt1(-/-)Lyz2-cre mice and Park2(-/-) mice had increased macrophage apoptosis and were protected from pulmonary fibrosis. Moreover, IPF alveolar macrophages had evidence of increased mitophagy and displayed apoptosis resistance. These observations suggest that Akt1-mediated mitophagy contributes to alveolar macrophage apoptosis resistance and is required for pulmonary fibrosis development.


Asunto(s)
Fibrosis Pulmonar Idiopática/inmunología , Pulmón/patología , Macrófagos Alveolares/fisiología , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Apoptosis/genética , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Fibrosis , Humanos , Fibrosis Pulmonar Idiopática/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Mitofagia/genética , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-akt/genética , Especies Reactivas de Oxígeno/metabolismo , Eliminación de Secuencia/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
2.
Am J Respir Cell Mol Biol ; 55(1): 58-71, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26699812

RESUMEN

M2 macrophages are implicated in the development of pulmonary fibrosis as they generate profibrotic signals. The polarization process, at least in part, is regulated by epigenetic modulation. Because Cu,Zn-superoxide dismutase-induced H2O2 can polarize macrophages to a profibrotic M2 phenotype, we hypothesized that modulation of the redox state of the cell is involved in the epigenetic modulation of the macrophage phenotype. In this study, we show that signal transducer and activator of transcription 6 (STAT6) regulates Jumonji domain containing (Jmjd) 3, a histone H3 lysine 27 demethylase, and mutation of a redox-sensitive cysteine in STAT6 attenuates jmjd3 expression. Moreover, Jmjd3 deficiency abrogates profibrotic M2 gene expression. Treatment with leflunomide, which reduces mitochondrial reactive oxygen species production and tyrosine phosphorylation, inhibits jmjd3 expression and M2 polarization, as well as development of a fibrotic phenotype. Taken together, these observations provide evidence that the redox regulation of Jmjd3 is a unique regulatory mechanism for Cu,Zn-superoxide dismutase-mediated profibrotic M2 polarization. Furthermore, leflunomide, which reduces reactive oxygen species production and tyrosine phosphorylation, may prove to be therapeutic in the treatment of asbestos-induced pulmonary fibrosis.


Asunto(s)
Polaridad Celular , Histona Demetilasas con Dominio de Jumonji/metabolismo , Macrófagos/patología , Superóxido Dismutasa-1/metabolismo , Animales , Línea Celular , Polaridad Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Interleucina-4/metabolismo , Isoxazoles/farmacología , Histona Demetilasas con Dominio de Jumonji/genética , Leflunamida , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/genética , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/metabolismo
3.
FASEB J ; 29(8): 3527-36, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25953850

RESUMEN

Alternative activation of alveolar macrophages is linked to fibrosis following exposure to asbestos. The scavenger receptor, macrophage receptor with collagenous structure (MARCO), provides innate immune defense against inhaled particles and pathogens; however, a receptor for asbestos has not been identified. We hypothesized that MARCO acts as an initial signaling receptor for asbestos, polarizes macrophages to a profibrotic M2 phenotype, and is required for the development of asbestos-induced fibrosis. Compared with normal subjects, alveolar macrophages isolated from patients with asbestosis express higher amounts of MARCO and have greater profibrotic polarization. Arginase 1 (40-fold) and IL-10 (265-fold) were higher in patients. In vivo, the genetic deletion of MARCO attenuated the profibrotic environment and pulmonary fibrosis in mice exposed to chrysotile. Moreover, alveolar macrophages from MARCO(-/-) mice polarize to an M1 phenotype, whereas wild-type mice have higher Ym1 (>3.0-fold) and nearly 7-fold more active TGF-ß1 in bronchoalveolar lavage (BAL) fluid (BALF). Arg(432) and Arg(434) in domain V of MARCO are required for the polarization of macrophages to a profibrotic phenotype as mutation of these residues reduced FIZZ1 expression (17-fold) compared with cells expressing MARCO. These observations demonstrate that a macrophage membrane protein regulates the fibrotic response to lung injury and suggest a novel target for therapeutic intervention.


Asunto(s)
Macrófagos/metabolismo , Macrófagos/patología , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Receptores Depuradores/metabolismo , Animales , Arginasa/metabolismo , Asbestos Serpentinas/metabolismo , Líquido del Lavado Bronquioalveolar , Línea Celular , Humanos , Interleucina-10/metabolismo , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor de Crecimiento Transformador beta1/metabolismo
4.
Am J Respir Cell Mol Biol ; 52(1): 106-15, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24988374

RESUMEN

Asthma is a disease of acute and chronic inflammation in which cytokines play a critical role in orchestrating the allergic inflammatory response. IL-13 and transforming growth factor (TGF)-ß promote fibrotic airway remodeling, a major contributor to disease severity. Improved understanding is needed, because current therapies are inadequate for suppressing development of airway fibrosis. IL-13 is known to stimulate respiratory epithelial cells to produce TGF-ß, but the mechanism through which this occurs is unknown. Here, we tested the hypothesis that reactive oxygen species (ROS) are a critical signaling intermediary between IL-13 or allergen stimulation and TGF-ß-dependent airway remodeling. We used cultured human bronchial epithelial cells and an in vivo mouse model of allergic asthma to map a pathway where allergens enhanced mitochondrial ROS, which is an essential upstream signal for TGF-ß activation and enhanced collagen production and deposition in airway fibroblasts. We show that mitochondria in airway epithelium are an essential source of ROS that activate TGF-ß expression and activity. TGF-ß from airway epithelium stimulates collagen expression in fibroblasts, contributing to an early fibrotic response to allergen exposure in cultured human airway cells and in ovalbumin-challenged mice. Treatment with the mitochondrial-targeted antioxidant, (2-(2,2,6,6-Tetramethylpiperidin-1-oxyl-4-ylamino)-2-oxoethyl)triphenylphosphonium chloride (mitoTEMPO), significantly attenuated mitochondrial ROS, TGF-ß, and collagen deposition in OVA-challenged mice and in cultured human epithelial cells. Our findings suggest that mitochondria are a critical source of ROS for promoting TGF-ß activity that contributes to airway remodeling in allergic asthma. Mitochondrial-targeted antioxidants may be a novel approach for future asthma therapies.


Asunto(s)
Antioxidantes/farmacología , Asma/tratamiento farmacológico , Asma/metabolismo , Colágeno/biosíntesis , Mitocondrias/metabolismo , Compuestos Organofosforados/farmacología , Piperidinas/farmacología , Factor de Crecimiento Transformador beta/biosíntesis , Animales , Asma/inducido químicamente , Asma/genética , Asma/patología , Células Cultivadas , Colágeno/genética , Modelos Animales de Enfermedad , Humanos , Interleucina-13/metabolismo , Ratones , Ratones Transgénicos , Mitocondrias/patología , Especies Reactivas de Oxígeno/metabolismo , Factor de Crecimiento Transformador beta/genética
5.
J Biol Chem ; 289(52): 36204-19, 2014 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-25378391

RESUMEN

Protein kinase B (Akt) is a key effector of multiple cellular processes, including cell survival. Akt, a serine/threonine kinase, is known to increase cell survival by regulation of the intrinsic pathway for apoptosis. In this study, we found that Akt modulated the mevalonate pathway, which is also linked to cell survival, by increasing Rho GTPase activation. Akt modulated the pathway by phosphorylating mevalonate diphosphate decarboxylase (MDD) at Ser(96). This phosphorylation in macrophages increased activation of Rac1, which enhanced macrophage survival because mutation of MDD (MDDS96A) induced apoptosis. Akt-mediated activation in macrophages was specific for Rac1 because Akt did not increase activity of other Rho GTP-binding proteins. The relationship between Akt and Rac1 was biologically relevant because Akt(+/-) mice had significantly less active Rac1 in alveolar macrophages, and macrophages from Akt(+/-) mice had an increase in active caspase-9 and -3. More importantly, Akt(+/-) mice were significantly protected from the development of pulmonary fibrosis, suggesting that macrophage survival is associated with the fibrotic phenotype. These observations for the first time suggest that Akt plays a critical role in the development and progression of pulmonary fibrosis by enhancing macrophage survival via modulation of the mevalonate pathway.


Asunto(s)
Macrófagos Alveolares/fisiología , Ácido Mevalónico/metabolismo , Proteínas Proto-Oncogénicas c-akt/fisiología , Fibrosis Pulmonar/enzimología , Secuencia de Aminoácidos , Animales , Vías Biosintéticas , Polaridad Celular , Supervivencia Celular , Células Cultivadas , Activación Enzimática , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/metabolismo , Neuropéptidos/metabolismo , Estrés Oxidativo , Fosforilación , Procesamiento Proteico-Postraduccional , Fibrosis Pulmonar/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Proteína de Unión al GTP rac1/metabolismo
6.
J Biol Chem ; 289(48): 33391-403, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25324550

RESUMEN

Although the mechanisms for fibrosis development remain largely unknown, recent evidence indicates that endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) may act as an important fibrotic stimulus in diseased lungs. ER stress is observed in lungs of patients with idiopathic pulmonary fibrosis. In this study we evaluated if ER stress and the UPR was present in macrophages exposed to chrysotile asbestos and if ER stress in macrophages was associated with asbestos-induced pulmonary fibrosis. Macrophages exposed to chrysotile had elevated transcript levels of several ER stress genes. Macrophages loaded with the Ca(2+)-sensitive dye Fura2-AM showed that cytosolic Ca(2+) increased significantly within minutes after chrysotile exposure and remained elevated for a prolonged time. Chrysotile-induced increases in cytosolic Ca(2+) were partially inhibited by either anisomycin, an inhibitor of passive Ca(2+) leak from the ER, or 1,2-bis(2-aminophenoxyl)ethane-N,N,N',N'-tetraacetic acid (BAPTA-AM), an intracellular Ca(2+) chelator known to deplete ER Ca(2+) stores. Anisomycin inhibited X-box-binding protein 1 (XBP1) mRNA splicing and reduced immunoglobulin-binding protein (BiP) levels, whereas BAPTA-AM increased XBP1 splicing and BiP expression, suggesting that ER calcium depletion may be one factor contributing to ER stress in cells exposed to chrysotile. To evaluate ER stress in vivo, asbestos-exposed mice showed fibrosis development, and alveolar macrophages from fibrotic mice showed increased expression of BiP. Bronchoalveolar macrophages from asbestosis patients showed increased expression of several ER stress genes compared with normal subjects. These findings suggest that alveolar macrophages undergo ER stress, which is associated with fibrosis development.


Asunto(s)
Asbestos Serpentinas/toxicidad , Asbestosis/metabolismo , Calcio/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Macrófagos Alveolares/metabolismo , Fibrosis Pulmonar/metabolismo , Adolescente , Adulto , Animales , Asbestosis/patología , Proteínas de Unión al ADN/metabolismo , Femenino , Humanos , Macrófagos Alveolares/patología , Masculino , Ratones , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/patología , Empalme del ARN/efectos de los fármacos , Factores de Transcripción del Factor Regulador X , Factores de Transcripción/metabolismo , Proteína 1 de Unión a la X-Box
7.
J Anat ; 227(6): 732-45, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25409684

RESUMEN

Scaffolds which aim to provide an optimised environment to regenerate bone tissue require a balance between mechanical properties and architecture known to be conducive to enable tissue regeneration, such as a high porosity and a suitable pore size. Using freeze-dried collagen-based scaffolds as an analogue of native ECM, we sought to improve the mechanical properties by incorporating hydroxyapatite (HA) in different ways while maintaining a pore architecture sufficient to allow cell infiltration, vascularisation and effective bone regeneration. Specifically we sought to elucidate the effect of different hydroxyapatite incorporation methods on the mechanical, morphological, and cellular response of the resultant collagen-HA scaffolds. The results demonstrated that incorporating either micron-sized (CHA scaffolds) or nano-sized HA particles (CnHA scaffolds) prior to freeze-drying resulted in moderate increases in stiffness (2.2-fold and 6.2-fold, respectively, vs. collagen-glycosaminoglycan scaffolds, P < 0.05, a scaffold known to support osteogenesis), while enabling good cell attachment, and moderate mesenchymal stem cell (MSC)-mediated calcium production after 28 days' culture (2.1-fold, P < 0.05, and 1.3-fold, respectively, vs. CG scaffolds). However, coating of collagen scaffolds with a hydroxyapatite precipitate after freeze-drying (CpHA scaffolds) has been shown to be a highly effective method to increase the compressive modulus (26-fold vs. CG controls, P < 0.001) of scaffolds while maintaining a high porosity (~ 98%). The coating of the ligand-dense collagen structure results in a lower cell attachment level (P < 0.05), although it supported greater cell-mediated calcium production (P < 0.0001) compared with other scaffold variants after 28 days' culture. The comparatively good mechanical properties of these high porosity scaffolds is obtained partially through highly crosslinking the scaffolds with both a physical (DHT) and chemical (EDAC) crosslinking treatment. Control of scaffold microstructure was examined via alterations in freezing temperature. It was found that the addition of HA prior to freeze-drying generally reduced the pore size and so the CpHA scaffold fabrication method offered increased control over the resulting scaffolds microstructure. These findings will help guide future design considerations for composite biomaterials and demonstrate that the method of HA incorporation can have profound effects on the resulting scaffold structural and biological response.


Asunto(s)
Regeneración Ósea , Colágeno/química , Durapatita/química , Andamios del Tejido/química , Animales , Calcio/metabolismo , Reactivos de Enlaces Cruzados/química , Módulo de Elasticidad , Etildimetilaminopropil Carbodiimida/química , Liofilización , Glicosaminoglicanos/química , Masculino , Células Madre Mesenquimatosas/metabolismo , Nanopartículas/química , Osteoblastos/citología , Osteogénesis , Porosidad , Ratas , Ratas Wistar , Temperatura
8.
J Biol Chem ; 288(28): 20745-57, 2013 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-23720777

RESUMEN

Macrophages not only initiate and accentuate inflammation after tissue injury, but they are also involved in resolution and repair. This difference in macrophage activity is the result of a differentiation process to either M1 or M2 phenotypes. M1 macrophages are pro-inflammatory and have microbicidal and tumoricidal activity, whereas the M2 macrophages are involved in tumor progression and tissue remodeling and can be profibrotic in certain conditions. Because mitochondrial Cu,Zn-superoxide dismutase (Cu,Zn-SOD)-mediated H2O2 is crucial for development of pulmonary fibrosis, we hypothesized that Cu,Zn-SOD modulated the macrophage phenotype. In this study, we demonstrate that Cu,Zn-SOD polarized macrophages to an M2 phenotype, and Cu,Zn-SOD-mediated H2O2 levels modulated M2 gene expression at the transcriptional level by redox regulation of a critical cysteine in STAT6. Furthermore, overexpression of Cu,Zn-SOD in mice resulted in a profibrotic environment and accelerated the development of pulmonary fibrosis, whereas polarization of macrophages to the M1 phenotype attenuated pulmonary fibrosis. Taken together, these observations provide a novel mechanism of Cu,Zn-SOD-mediated and Th2-independent M2 polarization and provide a potential therapeutic target for attenuating the accelerated development of pulmonary fibrosis.


Asunto(s)
Activación de Macrófagos/genética , Macrófagos/metabolismo , Fibrosis Pulmonar/metabolismo , Superóxido Dismutasa/metabolismo , Adolescente , Adulto , Animales , Línea Celular , Polaridad Celular/genética , Citocinas/genética , Citocinas/metabolismo , Progresión de la Enfermedad , Expresión Génica , Humanos , Peróxido de Hidrógeno/metabolismo , Immunoblotting , Macrófagos/clasificación , Macrófagos/enzimología , Macrófagos Alveolares/clasificación , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Mitocondrias/enzimología , Mitocondrias/genética , Mitocondrias/metabolismo , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Superóxido Dismutasa/genética , Células Th2/metabolismo , Adulto Joven
9.
J Biol Chem ; 287(5): 3301-12, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22157762

RESUMEN

The generation of reactive oxygen species, particularly H(2)O(2), from alveolar macrophages is causally related to the development of pulmonary fibrosis. Rac1, a small GTPase, is known to increase mitochondrial H(2)O(2) generation in macrophages; however, the mechanism by which this occurs is not known. This study shows that Rac1 is localized in the mitochondria of alveolar macrophages from asbestosis patients, and mitochondrial import requires the C-terminal cysteine of Rac1 (Cys-189), which is post-translationally modified by geranylgeranylation. Furthermore, H(2)O(2) generation mediated by mitochondrial Rac1 requires electron transfer from cytochrome c to a cysteine residue on Rac1 (Cys-178). Asbestos-exposed mice harboring a conditional deletion of Rac1 in macrophages demonstrated decreased oxidative stress and were significantly protected from developing pulmonary fibrosis. These observations demonstrate that mitochondrial import and direct electron transfer from cytochrome c to Rac1 modulates mitochondrial H(2)O(2) production in alveolar macrophages pulmonary fibrosis.


Asunto(s)
Citocromos c/metabolismo , Macrófagos Alveolares/enzimología , Proteínas Mitocondriales/metabolismo , Neuropéptidos/metabolismo , Fibrosis Pulmonar/enzimología , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Adolescente , Adulto , Anciano , Animales , Amianto/toxicidad , Carcinógenos/toxicidad , Citocromos c/genética , Transporte de Electrón/efectos de los fármacos , Transporte de Electrón/genética , Electrones , Femenino , Humanos , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Mutantes , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Neuropéptidos/genética , Prenilación de Proteína/efectos de los fármacos , Prenilación de Proteína/genética , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/genética , Proteínas de Unión al GTP rac/genética , Proteína de Unión al GTP rac1/genética
10.
Biochem J ; 445(2): 229-36, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22519702

RESUMEN

Rac1, a small GTPase, regulates macrophage MMP (matrix metalloproteinase)-9 in an ERK (extracellular-signal-regulated kinase)- and SP (specificity protein)-1-dependent manner. SP-1 contains a PEST (Pro-Glu-Ser-Thr) domain that may modulate protein stability. We hypothesize that Thr578, Ser586 and/or Ser587 in the PEST domain are required for SP-1 stability and MMP-9 expression secondary to activation of ERK, a serine/threonine kinase. We determined the effects of Rac1 and ERK on MMP-9 expression driven by SP-1WT (wild-type) and the SP-1 mutants T578A, S586A and S587A. Expression of WT and mutant SP-1 increased MMP9 promoter activity in alveolar macrophages. However, constitutively active Rac1 suppressed MMP9 promoter activity in cells expressing SP-1WT, SP-1T578A and SP-1S587A, but not SP-1S586A. Furthermore, constitutive ERK activation, which was inhibited by Rac1, significantly increased MMP9 transcription in cells expressing SP-1WT, but not SP-1S586A. As Rac1 activation and ERK inactivation increased degradation of SP-1WT and not SP-1S586A, the results of the present study suggest that SP-1 stability mediated at Ser586 regulates MMP9 transcription. Ex vivo, alveolar macrophages obtained from patients with asbestosis had less MMP-9 expression that was associated with decreased SP-1 expression and ERK activation. These observations demonstrate that Ser586 in the PEST domain of SP-1 is important for MMP9 gene expression in alveolar macrophages and highlight the importance of these proteins in pulmonary fibrosis.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Macrófagos Alveolares/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Serina/metabolismo , Factor de Transcripción Sp1/metabolismo , Adolescente , Adulto , Animales , Western Blotting , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación/genética , Neuropéptidos/fisiología , Estructura Terciaria de Proteína , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina/química , Serina/genética , Transducción de Señal , Factor de Transcripción Sp1/genética , Adulto Joven , Proteínas de Unión al GTP rac/fisiología , Proteína de Unión al GTP rac1
11.
J Biol Chem ; 286(17): 15597-607, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21393238

RESUMEN

The release of H(2)O(2) from alveolar macrophages has been linked to the development of pulmonary fibrosis, but little is known about its source or mechanism of production. We found that alveolar macrophages from asbestosis patients spontaneously produce high levels of H(2)O(2) and have high expression of Cu,Zn-superoxide dismutase (SOD). Because Cu,Zn-SOD is found in the mitochondrial intermembrane space (IMS), we hypothesized that mitochondrial Cu,Zn-SOD-mediated H(2)O(2) generation contributed to pulmonary fibrosis. Asbestos-induced translocation of Cu,Zn-SOD to the IMS was unique to macrophages and dependent on functional mitochondrial respiration and the presence of at least one of the conserved cysteines required for disulfide bond formation. These conserved cysteine residues were also necessary for enzyme activation and H(2)O(2) generation. Cu,Zn-SOD-mediated H(2)O(2) generation was inhibited by knockdown of the iron-sulfur protein, Rieske, in complex III. The role of Cu,Zn-SOD was biologically relevant in that Cu,Zn-SOD(-/-) mice generated significantly less H(2)O(2) and had less oxidant stress in bronchoalveolar lavage fluid and lung parenchyma. Furthermore, Cu,Zn-SOD(-/-) mice did not develop pulmonary fibrosis, and knockdown of Cu,Zn-SOD in monocytes attenuated collagen I deposition by lung fibroblasts. Our findings demonstrate a novel mechanism for the pathogenesis of pulmonary fibrosis where the antioxidant enzyme Cu,Zn-SOD translocates to the mitochondrial IMS to increase H(2)O(2) generation in alveolar macrophages.


Asunto(s)
Peróxido de Hidrógeno/metabolismo , Fibrosis Pulmonar/etiología , Superóxido Dismutasa/metabolismo , Adolescente , Adulto , Animales , Amianto/farmacología , Humanos , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Mitocondriales , Estrés Oxidativo , Transporte de Proteínas , Regulación hacia Arriba , Adulto Joven
12.
Mater Sci Eng C Mater Biol Appl ; 120: 111657, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33545824

RESUMEN

Core-shell scaffolds offer a promising regenerative solution to debilitating injuries to anterior cruciate ligament (ACL) thanks to a unique biphasic structure. Nevertheless, current core-shell designs are impaired by an imbalance between permeability, biochemical and mechanical cues. This study aimed to address this issue by creating a porous core-shell construct which favors cell infiltration and matrix production, while providing mechanical stability at the site of injury. The developed core-shell scaffold combines an outer shell of electrospun poly(caprolactone) fibers with a freeze-dried core of type I collagen doped with proteoglycans (biglycan, decorin) or glycosaminoglycans (chondroitin sulphate, dermatan sulphate). The aligned fibrous shell achieved an elastic modulus akin of the human ACL, while the porous collagen core is permeable to human mesenchymal stem cell (hMSC). Doping of the core with the aforementioned biomolecules led to structural and mechanical changes in the pore network. Assessment of cellular metabolic activity and scaffold contraction shows that hMSCs actively remodel the matrix at different degrees, depending on the core's doping formulation. Additionally, immunohistochemical staining and mRNA transcript levels show that the collagen-chondroitin sulphate formulation has the highest matrix production activity, while the collagen-decorin formulation featured a matrix production profile more characteristic of the undamaged tissue. Together, this demonstrates that scaffold doping with target biomolecules leads to distinct levels of cell-mediated matrix remodeling. Overall, this work resulted in the development of a versatile and robust platform with a combination of mechanical and biochemical features that have a significant potential in promoting the repair process of ACL tissue.


Asunto(s)
Glicosaminoglicanos , Andamios del Tejido , Colágeno , Humanos , Poliésteres , Ingeniería de Tejidos
13.
Biomaterials ; 269: 120651, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33476892

RESUMEN

This study addresses a crucial gap in the literature by characterising the relationship between urethral tissue mechanics, composition and gross structure. We then utilise these data to develop a biomimetic urethral scaffold with physical properties that more accurately mimic the native tissue than existing gold standard scaffolds; small intestinal submucosa (SIS) and urinary bladder matrix (UBM). Nine human urethra samples were mechanically characterised using pressure-diameter and uniaxial extension testing. The composition and gross structure of the tissue was determined using immunohistological staining. A pressure stiffening response is observed during the application of intraluminal pressure. The elastic and viscous tissue responses to extension are free of regional or directional variance. The elastin and collagen content of the tissue correlates significantly with tissue mechanics. Building on these data, a biomimetic urethral scaffold was fabricated from collagen and elastin in a ratio that mimics the composition of the native tissue. The resultant scaffold is comprised of a dense inner layer and a porous outer layer that structurally mimic the submucosa and corpus spongiosum layers of the native tissue, respectively. The porous outer layer facilitated more uniform cell infiltration relative to SIS and UBM when implanted subcutaneously (p < 0.05). The mechanical properties of the biomimetic scaffold better mimic the native tissue compared to SIS and UBM. The tissue characterisation data presented herein paves the way for the development of biomimetic urethral grafts, and the novel scaffold we develop demonstrates positive findings that warrant further in vivo evaluation.


Asunto(s)
Biomimética , Uretra , Colágeno , Humanos , Masculino , Ingeniería de Tejidos , Andamios del Tejido
14.
Acta Biomater ; 112: 52-61, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32525053

RESUMEN

This article describes the development of a hierarchical biofabrication technique suitable to create large but complex structures, such as vascular mimicking grafts, using facile lyophilisation technology amenable to multiple other biomaterial classes. The combination of three fabrication techniques together, namely solvent evaporation, lyophilisation, and crosslinking together allows highly tailorable structures from the microstructure up to the macrostructure, and with the ability to independently crosslink each layer it allows great flexibility to match desired native mechanical properties independently of the micro/macrostructure. We have demonstrated the flexibility of this biofabrication technique by independently optimising each of the layers to create a multi-layered arterial structure with tailored architectural and biophysical/biochemical properties using a collagen-elastin composite. Taken together, the facile biofabrication methodology developed has led to the development of a biomimetic bilayered scaffold suitable for use as a tissue engineered vascular graft (for haemodialysis access or peripheral/coronary bypass), or as an in vitro test platform to examine disease progression, pharmacological toxicity, or cardiovascular medical device testing. STATEMENT OF SIGNIFICANCE: The ability to grow large complex tissues such as blood vessels for transplantation is often hampered by the limitations of the selected biofabrication technique. Here, we sought to overcome some of the fabrication limitations for naturally occurring cardiovascular polymers (collagen/elastin) via a hierarchical approach to fabrication where each layer is built upon the previous. This approach enabled the flexibility to modify and tailor each layer's properties independently via control over polymer concentration, microstructure, and crosslinking. This simple approach facilitated us to fabricate multi-layered vascular grafts which were remodelled into high-density vascular tissue after 21-days. The fabrication approach could be translated to a myriad of other tissues while the engineered vascular graft could also be used as a test platform for drugs/medical devices or as a tissue engineering scaffold for vascular grafting for different indications.


Asunto(s)
Elastina , Injerto Vascular , Biomimética , Prótesis Vascular , Colágeno , Ingeniería de Tejidos , Andamios del Tejido
15.
Biomaterials ; 197: 405-416, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30708184

RESUMEN

The bone infection osteomyelitis (typically by Staphylococcus aureus) usually requires a multistep procedure of surgical debridement, long-term systemic high-dose antibiotics, and - for larger defects - bone grafting. This, combined with the alarming rise in antibiotic resistance, necessitates development of alternative approaches. Herein, we describe a one-step treatment for osteomyelitis that combines local, controlled release of non-antibiotic antibacterials with a regenerative collagen-based scaffold. To maximise efficacy, we utilised bioactive glass, an established osteoconductive material with immense capacity for bone repair, as a delivery platform for copper ions (proven antibacterial, angiogenic, and osteogenic properties). Multifunctional collagen-copper-doped bioactive glass scaffolds (CuBG-CS) were fabricated with favourable microarchitectural and mechanical properties (up to 1.9-fold increase in compressive modulus over CS) within the ideal range for bone tissue engineering. Scaffolds demonstrated antibacterial activity against Staphylococcus aureus (up to 66% inhibition) whilst also enhancing osteogenesis (up to 3.6-fold increase in calcium deposition) and angiogenesis in vitro. Most significantly, when assessed in a chick embryo in vivo model, CuBG-CS not only demonstrated biocompatibility, but also a significant angiogenic and osteogenic response, consistent with in vitro studies. Collectively, these results indicate that the CuBG-CS developed here show potential as a one-step osteomyelitis treatment: reducing infection, whilst enhancing bone healing.


Asunto(s)
Inductores de la Angiogénesis/administración & dosificación , Antibacterianos/administración & dosificación , Colágeno/química , Cobre/administración & dosificación , Osteogénesis/efectos de los fármacos , Andamios del Tejido/química , Inductores de la Angiogénesis/farmacología , Animales , Antibacterianos/farmacología , Materiales Biocompatibles/química , Línea Celular , Embrión de Pollo , Cobre/farmacología , Sistemas de Liberación de Medicamentos , Vidrio/química , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos
16.
Adv Mater ; 30(15): e1706442, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29504165

RESUMEN

Electroconductive substrates are emerging as promising functional materials for biomedical applications. Here, the development of biohybrids of collagen and pristine graphene that effectively harness both the biofunctionality of the protein component and the increased stiffness and enhanced electrical conductivity (matching native cardiac tissue) obtainable with pristine graphene is reported. As well as improving substrate physical properties, the addition of pristine graphene also enhances human cardiac fibroblast growth while simultaneously inhibiting bacterial attachment (Staphylococcus aureus). When embryonic-stem-cell-derived cardiomyocytes (ESC-CMs) are cultured on the substrates, biohybrids containing 32 wt% graphene significantly increase metabolic activity and cross-striated sarcomeric structures, indicative of the improved substrate suitability. By then applying electrical stimulation to these conductive biohybrid substrates, an enhancement of the alignment and maturation of the ESC-CMs is achieved. While this in vitro work has clearly shown the potential of these materials to be translated for cardiac applications, it is proposed that these graphene-based biohybrid platforms have potential for a myriad of other applications-particularly in electrically sensitive tissues, such as neural and neural and musculoskeletal tissues.


Asunto(s)
Materiales Biocompatibles/química , Colágeno , Conductividad Eléctrica , Grafito , Humanos , Miocitos Cardíacos
17.
ACS Biomater Sci Eng ; 3(7): 1221-1235, 2017 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-33440511

RESUMEN

Peripheral nerve injuries have high incidence rates, limited treatment options and poor clinical outcomes, rendering a significant socioeconomic burden. For effective peripheral nerve repair, the gap or site of injury must be structurally bridged to promote correct reinnervation and functional regeneration. However, effective repair becomes progressively more difficult with larger gaps. Autologous nerve grafting remains the best clinical option for the repair of large gaps (20-80 mm) despite being associated with numerous limitations including permanent donor site morbidity, a lack of available tissue and the formation of neuromas. To meet the clinical demand of large gap repair and overcome these limitations, tissue engineering has led to the development of nerve guidance conduit-based therapeutics. This review focuses on the advances of nerve guidance conduit-based therapeutics in terms of their structural properties including biomimetic composition, permeability, architecture, and surface modifications. Associated biochemical properties, pertaining to the incorporation of cells and neurotrophic factors, are also reviewed. After reviewing the progress in the field, we conclude by presenting an outlook on their clinical translatability and the next generation of therapeutics.

18.
Curr Opin Pharmacol ; 36: 66-71, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28865291

RESUMEN

Islet transplantation for the treatment of type 1 diabetes (T1D) is hampered by the shortage of donor tissue and the need for life-long immunosuppression. The engineering of materials to limit host immune rejection opens the possibilities of utilising allogeneic and even xenogeneic cells without the need for systemic immunosuppression. Here we discuss the most recent developments in immunoisolation of transplanted cells using advanced polymeric biomaterials, utilising macroscale to nanoscale approaches, to limit aberrant immune responses.


Asunto(s)
Reacción a Cuerpo Extraño/prevención & control , Trasplante de Islotes Pancreáticos/instrumentación , Animales , Cuerpos Extraños , Humanos , Islotes Pancreáticos , Polímeros/administración & dosificación , Polímeros/química
19.
Adv Healthc Mater ; 6(24)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28975768

RESUMEN

Clinically available hollow nerve guidance conduits (NGCs) have had limited success in treating large peripheral nerve injuries. This study aims to develop a biphasic NGC combining a physicochemically optimized collagen outer conduit to bridge the transected nerve, and a neuroconductive hyaluronic acid-based luminal filler to support regeneration. The outer conduit is mechanically optimized by manipulating crosslinking and collagen density, allowing the engineering of a high wall permeability to mitigate the risk of neuroma formation, while also maintaining physiologically relevant stiffness and enzymatic degradation tuned to coincide with regeneration rates. Freeze-drying is used to seamlessly integrate the luminal filler into the conduit, creating a longitudinally aligned pore microarchitecture. The luminal stiffness is modulated to support Schwann cells, with laminin incorporation further enhancing bioactivity by improving cell attachment and metabolic activity. Additionally, this biphasic NGC is shown to support neurogenesis and gliogenesis of neural progenitor cells and axonal outgrowth from dorsal root ganglia. These findings highlight the paradigm that a successful NGC requires the concerted optimization of both a mechanical support phase capable of bridging a nerve defect and a neuroconductive phase with an architecture capable of supporting both Schwann cells and neurons in order to achieve functional regenerative outcome.


Asunto(s)
Regeneración Nerviosa , Nervios Periféricos/cirugía , Prótesis e Implantes , Animales , Materiales Biocompatibles/química , Línea Celular , Colágeno/química , Ganglios Espinales/metabolismo , Laminina/metabolismo , Masculino , Neurogénesis , Fenobarbital/química , Ratas , Ratas Endogámicas Lew , Células de Schwann/metabolismo , Ingeniería de Tejidos
20.
Drug Discov Today ; 21(9): 1437-1445, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27117348

RESUMEN

The field of tissue engineering is developing biomimetic biomaterial scaffolds that are showing increasing therapeutic potential for the repair of cardiovascular tissues. However, a major opportunity exists to use them as 3D in vitro models for the study of cardiovascular tissues and disease in addition to drug development and testing. These in vitro models can span the gap between 2D culture and in vivo testing, thus reducing the cost, time, and ethical burden of current approaches. Here, we outline the progress to date and the requirements for the development of ideal in vitro 3D models for blood vessels, heart valves, and myocardial tissue.


Asunto(s)
Vasos Sanguíneos , Válvulas Cardíacas , Modelos Biológicos , Miocardio , Animales , Enfermedades Cardiovasculares , Humanos , Ingeniería de Tejidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA