Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
J Immunol ; 207(6): 1627-1640, 2021 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-34433619

RESUMEN

Silicosis is a lethal pneumoconiosis for which no therapy is available. Silicosis is a global threat, and more than 2.2 million people per year are exposed to silica in the United States. The initial response to silica is mediated by innate immunity. Phagocytosis of silica particles by macrophages is followed by recruitment of mitochondria to phagosomes, generation of mitochondrial reactive oxygen species, and cytokine (IL-1ß, TNF-α, IFN-ß) release. In contrast with LPS, the metabolic remodeling of silica-exposed macrophages is unclear. This study contrasts mitochondrial and metabolic alterations induced by LPS and silica on macrophages and correlates them with macrophage viability and cytokine production, which are central to the pathogenesis of silicosis. Using high-resolution respirometer and liquid chromatography-high-resolution mass spectrometry, we determined the effects of silica and LPS on mitochondrial respiration and determined changes in central carbon metabolism of murine macrophage cell lines RAW 264.7 and IC-21. We show that silica induces metabolic reprogramming of macrophages. Silica, as well as LPS, enhances glucose uptake and increases aerobic glycolysis in macrophages. In contrast with LPS, silica affects mitochondria respiration, reducing complex I and enhancing complex II activity, to sustain cell viability. These mitochondrial alterations are associated in silica, but not in LPS-exposed macrophages, with reductions of tricarboxylic acid cycle intermediates, including succinate, itaconate, glutamate, and glutamine. Furthermore, in contrast with LPS, these silica-induced metabolic adaptations do not correlate with IL-1ß or TNF-α production, but with the suppressed release of IFN-ß. Our data highlight the importance of complex II activity and tricarboxylic acid cycle remodeling to macrophage survival and cytokine-mediated inflammation in silicosis.


Asunto(s)
Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Dióxido de Silicio/química , Dióxido de Silicio/farmacología , Silicosis/inmunología , Animales , Supervivencia Celular/efectos de los fármacos , Ciclo del Ácido Cítrico/efectos de los fármacos , Cristalización , Citocinas/biosíntesis , Inflamación/inmunología , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fagocitosis/efectos de los fármacos , Fagosomas/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Silicosis/metabolismo
2.
Int J Mol Sci ; 24(6)2023 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-36982887

RESUMEN

COPD is a chronic lung disease that affects millions of people, declining their lung function and impairing their life quality. Despite years of research and drug approvals, we are still not capable of halting progression or restoring normal lung function. Mesenchymal stem cells (MSC) are cells with extraordinary repair capacity, and MSC-based therapy brings future hope for COPD treatment, although the best source and route of administration are unclear. MSC from adipose tissue (AD-MSC) represents an option for autologous treatment; however, they could be less effective than donor MSC. We compared in vitro behavior of AD-MSC from COPD and non-COPD individuals by migration/proliferation assay, and tested their therapeutic potential in an elastase mouse model. In addition, we tested intravenous versus intratracheal routes, inoculating umbilical cord (UC) MSC and analyzed molecular changes by protein array. Although COPD AD-MSC have impaired migratory response to VEGF and cigarette smoke, they were as efficient as non-COPD in reducing elastase-induced lung emphysema. UC-MSC reduced lung emphysema regardless of the administration route and modified the inflammatory profile in elastase-treated mice. Our data demonstrate equal therapeutic potential of AD-MSC from COPD and non-COPD subjects in the pre-clinical model, thus supporting their autologous use in disease.


Asunto(s)
Enfisema , Células Madre Mesenquimatosas , Enfisema Pulmonar , Animales , Ratones , Elastasa Pancreática , Enfisema Pulmonar/inducido químicamente , Enfisema Pulmonar/terapia , Células Madre Mesenquimatosas/fisiología , Fenómenos Fisiológicos Respiratorios
3.
Cytotherapy ; 23(5): 373-380, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33934807

RESUMEN

Mesenchymal stromal/stem cells (MSCs) have been widely tested against many diseases, with more than 1000 registered clinical trials worldwide. Despite many setbacks, MSCs have been approved for the treatment of graft-versus-host disease and Crohn disease. However, it is increasingly clear that MSCs exert their therapeutic functions in a paracrine manner through the secretion of small extracellular vesicles (sEVs) of 50-200 nm in diameter. Unlike living cells that can persist long-term, sEVs are non-living and non-replicative and have a transient presence in the body. Their small size also renders sEV preparations highly amenable to sterilization by filtration. Together, acellular MSC-sEV preparations are potentially safer and easier to translate into the clinic than cellular MSC products. Nevertheless, there are inherent challenges in the development of MSC-sEV drug products. MSC-sEVs are products of living cells, and living cells are sensitive to changes in the external microenvironment. Consequently, quality control metrics to measure key identity and potency features of MSC-sEV preparations have to be specified during development of MSC-sEV therapeutics. The authors have previously described quantifiable assays to define the identity of MSC-sEVs. Here the authors discuss requirements for prospective potency assays to predict the therapeutic effectiveness of the drug substance in accordance with International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use guidelines. Although potency assays should ideally reflect the mechanism of action (MoA), this is challenging because the MoA for the reported efficacy of MSC-sEV preparations against multiple diseases of diverse underlying pathology is likely to be complex and different for each disease and difficult to fully elucidate. Nevertheless, robust potency assays could be developed by identifying the EV attribute most relevant to the intended biological activity in EV-mediated therapy and quantifying the EV attribute. Specifically, the authors highlight challenges and mitigation measures to enhance the manufacture of consistent and reproducibly potent sEV preparations, to identify and select the appropriate EV attribute for potency assays despite a complex "work-in-progress" MoA and to develop assays likely to be compliant with regulatory guidance for assay validation.


Asunto(s)
Vesículas Extracelulares , Enfermedad Injerto contra Huésped , Células Madre Mesenquimatosas , Humanos , Estudios Prospectivos
4.
Cytotherapy ; 22(9): 482-485, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32425691

RESUMEN

STATEMENT: The International Society for Cellular and Gene Therapies (ISCT) and the International Society for Extracellular Vesicles (ISEV) recognize the potential of extracellular vesicles (EVs, including exosomes) from mesenchymal stromal cells (MSCs) and possibly other cell sources as treatments for COVID-19. Research and trials in this area are encouraged. However, ISEV and ISCT do not currently endorse the use of EVs or exosomes for any purpose in COVID-19, including but not limited to reducing cytokine storm, exerting regenerative effects or delivering drugs, pending the generation of appropriate manufacturing and quality control provisions, pre-clinical safety and efficacy data, rational clinical trial design and proper regulatory oversight.


Asunto(s)
Vesículas Extracelulares , Células Madre Mesenquimatosas/citología , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/inmunología , Exosomas/trasplante , Vesículas Extracelulares/trasplante , Humanos , Sociedades Científicas , Tratamiento Farmacológico de COVID-19
5.
Am J Respir Crit Care Med ; 198(7): 914-927, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29727583

RESUMEN

RATIONALE: Idiopathic pulmonary fibrosis (IPF) is a progressive, fibrotic interstitial lung disease characterized by (myo)fibroblast accumulation and collagen deposition. Resistance to Fas-induced apoptosis is thought to facilitate (myo)fibroblast persistence in fibrotic lung tissues by poorly understood mechanisms. OBJECTIVES: To test the hypothesis that PTPN13 (protein tyrosine phosphatase-N13) is expressed by IPF lung (myo)fibroblasts, promotes their resistance to Fas-induced apoptosis, and contributes to the development of pulmonary fibrosis. METHODS: PTPN13 was localized in lung tissues from patients with IPF and control subjects by immunohistochemical staining. Inhibition of PTPN13 function in primary IPF and normal lung (myo)fibroblasts was accomplished by: 1) downregulation with TNF-α (tumor necrosis factor-α)/IFN-γ, 2) siRNA knockdown, or 3) a cell-permeable Fas/PTPN13 interaction inhibitory peptide. The role of PTPN13 in the development of pulmonary fibrosis was assessed in mice with genetic deficiency of PTP-BL, the murine ortholog of PTPN13. MEASUREMENTS AND MAIN RESULTS: PTPN13 was constitutively expressed by (myo)fibroblasts in the fibroblastic foci of patients with IPF. Human lung (myo)fibroblasts, which are resistant to Fas-induced apoptosis, basally expressed PTPN13 in vitro. TNF-α/IFN-γ or siRNA-mediated PTPN13 downregulation and peptide-mediated inhibition of the Fas/PTPN13 interaction in human lung (myo)fibroblasts promoted Fas-induced apoptosis. Bleomycin-challenged PTP-BL-/- mice, while developing inflammatory lung injury, exhibited reduced pulmonary fibrosis compared with wild-type mice. CONCLUSIONS: These findings suggest that PTPN13 mediates the resistance of human lung (myo)fibroblasts to Fas-induced apoptosis and promotes pulmonary fibrosis in mice. Our results suggest that strategies aimed at interfering with PTPN13 expression or function may represent a novel strategy to reduce fibrosis in IPF.


Asunto(s)
Apoptosis/genética , Bleomicina/farmacología , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/patología , Miofibroblastos/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 13/genética , Animales , Biopsia con Aguja , Estudios de Casos y Controles , Regulación hacia Abajo , Farmacorresistencia Microbiana , Femenino , Humanos , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Inmunohistoquímica , Masculino , Ratones , Ratones Noqueados , ARN Interferente Pequeño/genética , Valores de Referencia , Técnicas de Cultivo de Tejidos , Receptor fas/efectos de los fármacos
6.
Am J Bot ; 103(11): 1872-1879, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27864266

RESUMEN

PREMISE OF STUDY: Deciduous woody species invest considerable resources in the growth of new foliage and distal stems. This new growth is at risk for mechanical damage from high winds and storms. Pawpaw has large leaves borne distally on thin twigs. Following a storm, pawpaw branches sometimes exhibit a persistent "flipped" orientation, slowly returning upright over 24 h. We investigated biomechanical properties of pawpaw twigs, comparing them to co-occurring species with similarly high leaf areas and loads, which do not exhibit this "flipping". Our goal was to determine biomechanical and structural properties in these species and how variation in form might relate to functional differences. METHODS: We measured flexural stiffness, torsional stiffness, and viscoelastic creep in pawpaw and co-occurring trees Liriodendron tulipifera and Carya cordiformis. We also recorded twig/foliage reconfiguration in high winds. We stained thin cross sections of distal twigs and recorded images using fluorescent light microscopy. KEY RESULTS: Flexural and torsional stiffness increased with twig radius in pawpaw and tulip tree, although torsional stiffness increased more slowly in pawpaw. Pawpaw had a high ratio of flexural to torsional stiffness (EI/GJ) across a range of twig radii and significant viscoelastic creep compared with the other species. CONCLUSIONS: Biomechanical data showed that pawpaw twigs were "twistier" than the comparison species, which were shown previously to alleviate drag-induced damage by reorienting petioles and leaves. Pawpaw has an unusual strategy of low torsional stiffness in twigs, allowing for reorientation of the entire distal appendage, likely minimizing drag-induced damage in storms.


Asunto(s)
Asimina/crecimiento & desarrollo , Asimina/anatomía & histología , Asimina/fisiología , Fenómenos Biomecánicos , Hojas de la Planta/anatomía & histología , Hojas de la Planta/crecimiento & desarrollo , Hojas de la Planta/fisiología , Tallos de la Planta/anatomía & histología , Tallos de la Planta/crecimiento & desarrollo , Tallos de la Planta/fisiología , Especificidad de la Especie , Árboles , Viento , Madera
7.
J Immunol ; 192(8): 3837-46, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24623132

RESUMEN

Macrophages play a fundamental role in innate immunity and the pathogenesis of silicosis. Phagocytosis of silica particles is associated with the generation of reactive oxygen species (ROS), secretion of cytokines, such as TNF, and cell death that contribute to silica-induced lung disease. In macrophages, ROS production is executed primarily by activation of the NADPH oxidase (Phox) and by generation of mitochondrial ROS (mtROS); however, the relative contribution is unclear, and the effects on macrophage function and fate are unknown. In this study, we used primary human and mouse macrophages (C57BL/6, BALB/c, and p47(phox-/-)) and macrophage cell lines (RAW 264.7 and IC21) to investigate the contribution of Phox and mtROS to silica-induced lung injury. We demonstrate that reduced p47(phox) expression in IC21 macrophages is linked to enhanced mtROS generation, cardiolipin oxidation, and accumulation of cardiolipin hydrolysis products, culminating in cell death. mtROS production is also observed in p47(phox-/-) macrophages, and p47(phox-/-) mice exhibit increased inflammation and fibrosis in the lung following silica exposure. Silica induces interaction between TNFR1 and Phox in RAW 264.7 macrophages. Moreover, TNFR1 expression in mitochondria decreased mtROS production and increased RAW 264.7 macrophage survival to silica. These results identify TNFR1/Phox interaction as a key event in the pathogenesis of silicosis that prevents mtROS formation and reduces macrophage apoptosis.


Asunto(s)
Mitocondrias/metabolismo , NADPH Oxidasas/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Silicosis/metabolismo , Animales , Muerte Celular , Línea Celular , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Lesión Pulmonar/etiología , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , NADPH Oxidasas/genética , Unión Proteica , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Dióxido de Silicio/efectos adversos , Dióxido de Silicio/metabolismo , Silicosis/genética
8.
Am J Respir Crit Care Med ; 189(2): 214-22, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24160862

RESUMEN

The median survival of patients with idiopathic pulmonary fibrosis (IPF) continues to be approximately 3 years from the time of diagnosis, underscoring the lack of effective medical therapies for this disease. In the United States alone, approximately 40,000 patients die of this disease annually. In November 2012, the NHLBI held a workshop aimed at coordinating research efforts and accelerating the development of IPF therapies. Basic, translational, and clinical researchers gathered with representatives from the NHLBI, patient advocacy groups, pharmaceutical companies, and the U.S. Food and Drug Administration to review the current state of IPF research and identify priority areas, opportunities for collaborations, and directions for future research. The workshop was organized into groups that were tasked with assessing and making recommendations to promote progress in one of the following six critical areas of research: (1) biology of alveolar epithelial injury and aberrant repair; (2) role of extracellular matrix; (3) preclinical modeling; (4) role of inflammation and immunity; (5) genetic, epigenetic, and environmental determinants; (6) translation of discoveries into diagnostics and therapeutics. The workshop recommendations provide a basis for directing future research and strategic planning by scientific, professional, and patient communities and the NHLBI.


Asunto(s)
Fibrosis Pulmonar Idiopática , Animales , Investigación Biomédica/tendencias , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Predisposición Genética a la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/diagnóstico , Fibrosis Pulmonar Idiopática/fisiopatología , Fibrosis Pulmonar Idiopática/terapia , Inflamación/inmunología , Ratones , Alveolos Pulmonares/patología , Mucosa Respiratoria/patología
9.
Am J Respir Cell Mol Biol ; 50(4): 825-37, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24325577

RESUMEN

Idiopathic pulmonary fibrosis (IPF) is a relentless, fibrotic parenchymal lung disease in which alternatively programmed macrophages produce profibrotic molecules that promote myofibroblast survival and collagen synthesis. Effective therapies to treat patients with IPF are lacking, and conventional therapy may be harmful. We tested the hypothesis that therapeutic lung delivery of the proinflammatory cytokine tumor necrosis factor (TNF)-α into wild-type fibrotic mice would reduce the profibrotic milieu and accelerate the resolution of established pulmonary fibrosis. Fibrosis was assessed in bleomycin-instilled wild-type and TNF-α(-/-) mice by measuring hydroxyproline levels, static compliance, and Masson's trichrome staining. Macrophage infiltration and programming status was assessed by flow cytometry of enzymatically digested lung and in situ immunostaining. Pulmonary delivery of TNF-α to wild-type mice with established pulmonary fibrosis was found to reduce their fibrotic burden, to improve lung function and architecture, and to reduce the number and programming status of profibrotic alternatively programmed macrophages. In contrast, fibrosis and alternative macrophage programming were prolonged in bleomycin-instilled TNF-α(-/-) mice. To address the role of the reduced numbers of alternatively programmed macrophages in the TNF-α-induced resolution of established pulmonary fibrosis, we conditionally depleted macrophages in MAFIA (MAcrophage Fas-Induced Apoptosis) mice. Conditional macrophage depletion phenocopied the resolution of established pulmonary fibrosis observed after therapeutic TNF-α delivery. Taken together, our results show for the first time that TNF-α is involved in the resolution of established pulmonary fibrosis via a mechanism involving reduced numbers and programming status of profibrotic macrophages. We speculate that pulmonary delivery of TNF-α or augmenting its signaling pathway represent a novel therapeutic strategy to resolve established pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Pulmón/efectos de los fármacos , Macrófagos Alveolares/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Animales , Bleomicina , Células Cultivadas , Modelos Animales de Enfermedad , Hidroxiprolina/metabolismo , Fibrosis Pulmonar Idiopática/genética , Fibrosis Pulmonar Idiopática/metabolismo , Fibrosis Pulmonar Idiopática/patología , Fibrosis Pulmonar Idiopática/fisiopatología , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Rendimiento Pulmonar , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptor de Factor Estimulante de Colonias de Macrófagos/genética , Recuperación de la Función , Inducción de Remisión , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Factores de Tiempo , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Receptor fas/genética , Receptor fas/metabolismo
10.
Am J Respir Crit Care Med ; 188(3): 370-5, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23713908

RESUMEN

The National Heart, Lung, and Blood Institute (NHLBI) of the National Institutes of Health convened the Cell Therapy for Lung Disease Working Group on November 13-14, 2012, to review and formulate recommendations for future research directions. The workshop brought together investigators studying basic mechanisms and the roles of cell therapy in preclinical models of lung injury and pulmonary vascular disease, with clinical trial experts in cell therapy for cardiovascular diseases and experts from the NHLBI Production Assistance for Cell Therapy program. The purpose of the workshop was to discuss the current status of basic investigations in lung cell therapy, to identify some of the scientific gaps in current knowledge regarding the potential roles and mechanisms of cell therapy in the treatment of lung diseases, and to develop recommendations to the NHLBI and the research community on scientific priorities and practical steps that would lead to first-in-human trials of lung cell therapy.


Asunto(s)
Investigación Biomédica/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedades Pulmonares/terapia , National Heart, Lung, and Blood Institute (U.S.) , Humanos , Estados Unidos
11.
Am J Respir Cell Mol Biol ; 49(2): 306-15, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23590297

RESUMEN

In the mouse lung, Escherichia coli LPS can decrease surfactant protein-B (SFTPB) mRNA and protein concentrations. LPS also regulates the expression, synthesis, and concentrations of a variety of gene and metabolic products that inhibit SFTPB gene expression. The purpose of the present study was to determine whether LPS acts directly or indirectly on pulmonary epithelial cells to trigger signaling pathways that inhibit SFTPB expression, and whether the transcription factor CCAAT/enhancer binding protein (C/EBP)-ß (CEBPB) is a downstream inhibitory effector. To investigate the mechanism of SFTPB repression, the human pulmonary epithelial cell lines NCI-H441 (H441) and NCI-H820 (H820) and the mouse macrophage-like cell line RAW264.7 were treated with LPS. Whereas LPS did not decrease SFTPB transcripts in H441 or H820 cells, the conditioned medium of LPS-treated RAW264.7 cells decreased SFTPB transcripts in H441 and H820 cells, and inhibited SFTPB promoter activity in H441 cells. In the presence of neutralizing anti-tumor necrosis factor (TNF) antibodies, the conditioned medium of LPS-treated RAW264.7 cells did not inhibit SFTPB promoter activity. In H441 cells treated with recombinant TNF protein, SFTPB transcripts decreased, whereas CEBPB transcripts increased and the transient coexpression of CEBPB decreased SFTPB promoter activity. Further, CEBPB short, interfering RNA increased basal SFTPB transcripts and countered the decrease of SFTPB transcripts by TNF. Together, these findings suggest that macrophages participate in the repression of SFTPB expression by LPS, and that macrophage-released cytokines (including TNF) regulate the transcription factor CEBPB, which can function as a downstream transcriptional repressor of SFTPB gene expression in pulmonary epithelial cells.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Lipopolisacáridos/toxicidad , Macrófagos Alveolares/metabolismo , Proteína B Asociada a Surfactante Pulmonar/biosíntesis , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Macrófagos Alveolares/citología , Ratones , Regiones Promotoras Genéticas , Mucosa Respiratoria/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
12.
Stem Cells ; 30(5): 975-87, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22367737

RESUMEN

Large scale expansion of human mesenchymal stem cells (MSCs) is routinely performed for clinical therapy. In contrast, developing protocols for large scale expansion of primary mouse MSCs has been more difficult due to unique aspects of rodent biology. Currently, established methods to isolate mouse MSCs select for rapidly dividing subpopulations that emerge from bone marrow cultures following long-term (months) expansion in atmospheric oxygen. Herein, we demonstrate that exposure to atmospheric oxygen rapidly induced p53, TOP2A, and BCL2-associated X protein (BAX) expression and mitochondrial reactive oxygen species (ROS) generation in primary mouse MSCs resulting in oxidative stress, reduced cell viability, and inhibition of cell proliferation. Alternatively, procurement and culture in 5% oxygen supported more prolific expansion of the CD45(-ve) /CD44(+ve) cell fraction in marrow, produced increased MSC yields following immunodepletion, and supported sustained MSC growth resulting in a 2,300-fold increase in cumulative cell yield by fourth passage. MSCs cultured in 5% oxygen also exhibited enhanced trilineage differentiation. The oxygen-induced stress response was dependent upon p53 since siRNA-mediated knockdown of p53 in wild-type cells or exposure of p53(-/-) MSCs to atmospheric oxygen failed to induce ROS generation, reduce viability, or arrest cell growth. These data indicate that long-term culture expansion of mouse MSCs in atmospheric oxygen selects for clones with absent or impaired p53 function, which allows cells to escape oxygen-induced growth inhibition. In contrast, expansion in 5% oxygen generates large numbers of primary mouse MSCs that retain sensitivity to atmospheric oxygen, and therefore a functional p53 protein, even after long-term expansion in vitro.


Asunto(s)
Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Oxígeno/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Células de la Médula Ósea/citología , Células Cultivadas , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Proteínas de Unión a Poli-ADP-Ribosa , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo
13.
J Occup Environ Hyg ; 10(12): 685-93, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24195535

RESUMEN

In this study a serial multi-cyclone sampling array capable of simultaneously sampling particles of multiple size fractions, from an occupational environment, for use in in vivo and in vitro toxicity studies and physical/chemical characterization, was developed and tested. This method is an improvement over current methods used to size-segregate occupational aerosols for characterization, due to its simplicity and its ability to collect sufficient masses of nano- and ultrafine sized particles for analysis. This method was evaluated in a chamber providing a uniform atmosphere of dust concentrations using crystalline silica particles. The multi-cyclone sampling array was used to segregate crystalline silica particles into four size fractions, from a chamber concentration of 10 mg/m(3). The size distributions of the particles collected at each stage were confirmed, in the air, before and after each cyclone stage. Once collected, the particle size distribution of each size fraction was measured using light scattering techniques to further confirm the size distributions. As a final confirmation, scanning electron microscopy was used to collect images of each size fraction. The results presented here, using multiple measurement techniques, show that this multi-cyclone system was able to successfully collect distinct size-segregated particles at sufficient masses to perform toxicological evaluations and physical/chemical characterization.


Asunto(s)
Aerosoles/análisis , Monitoreo del Ambiente/instrumentación , Exposición Profesional/análisis , Aerosoles/química , Monitoreo del Ambiente/métodos , Microscopía de Fuerza Atómica , Tamaño de la Partícula , Dióxido de Silicio/análisis , Dióxido de Silicio/química
14.
JCI Insight ; 8(3)2023 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-36752201

RESUMEN

Patients with progressive fibrosing interstitial lung diseases (PF-ILDs) carry a poor prognosis and have limited therapeutic options. A hallmark feature is fibroblast resistance to apoptosis, leading to their persistence, accumulation, and excessive deposition of extracellular matrix. A complex balance of the B cell lymphoma 2 (BCL-2) protein family controlling the intrinsic pathway of apoptosis and fibroblast reliance on antiapoptotic proteins has been hypothesized to contribute to this resistant phenotype. Examination of lung tissue from patients with PF-ILD (idiopathic pulmonary fibrosis and silicosis) and mice with PF-ILD (repetitive bleomycin and silicosis) showed increased expression of antiapoptotic BCL-2 family members in α-smooth muscle actin-positive fibroblasts, suggesting that fibroblasts from fibrotic lungs may exhibit increased susceptibility to inhibition of antiapoptotic BCL-2 family members BCL-2, BCL-XL, and BCL-W with the BH3 mimetic ABT-263. We used 2 murine models of PF-ILD to test the efficacy of ABT-263 in reversing established persistent pulmonary fibrosis. Treatment with ABT-263 induced fibroblast apoptosis, decreased fibroblast numbers, and reduced lung collagen levels, radiographic disease, and histologically evident fibrosis. Our studies provide insight into how fibroblasts gain resistance to apoptosis and become sensitive to the therapeutic inhibition of antiapoptotic proteins. By targeting profibrotic fibroblasts, ABT-263 offers a promising therapeutic option for PF-ILDs.


Asunto(s)
Fibrosis Pulmonar Idiopática , Enfermedades Pulmonares Intersticiales , Silicosis , Ratones , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Fibrosis Pulmonar Idiopática/patología , Apoptosis/genética , Enfermedades Pulmonares Intersticiales/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Fibroblastos/metabolismo , Silicosis/metabolismo
15.
Sci Adv ; 9(45): eadi2387, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37948519

RESUMEN

Mesenchymal stem/stromal cells (MSCs) have been evaluated in >1500 clinical trials, but outcomes remain suboptimal because of knowledge gaps in quality attributes that confer potency. We show that TWIST1 directly represses TSG6 expression that TWIST1 and TSG6 are inversely correlated across bone marrow-derived MSC (BM-MSC) donor cohorts and predict interdonor differences in their proangiogenic, anti-inflammatory, and immune suppressive activity in vitro and in sterile inflammation and autoimmune type 1 diabetes preclinical models. Transcript profiling of TWIST1HiTSG6Low versus TWISTLowTSG6Hi BM-MSCs revealed previously unidentified roles for TWIST1/TSG6 in regulating cellular oxidative stress and TGF-ß2 in modulating TSG6 expression and anti-inflammatory activity. TWIST1 and TSG6 levels also correlate to donor stature and predict differences in iPSC-derived MSC quality attributes. These results validate TWIST1 and TSG6 as biomarkers that predict interdonor differences in potency across laboratories and assay platforms, thereby providing a means to manufacture MSC products tailored to specific diseases.


Asunto(s)
Células Madre Mesenquimatosas , Humanos , Antiinflamatorios/farmacología , Biomarcadores/metabolismo , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Factores Inmunológicos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
16.
Front Immunol ; 13: 936167, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341426

RESUMEN

In the lungs, macrophages constitute the first line of defense against pathogens and foreign bodies and play a fundamental role in maintaining tissue homeostasis. Activated macrophages show altered immunometabolism and metabolic changes governing immune effector mechanisms, such as cytokine secretion characterizing their classic (M1) or alternative (M2) activation. Lipopolysaccharide (LPS)-stimulated macrophages demonstrate enhanced glycolysis, blocked succinate dehydrogenase (SDH), and increased secretion of interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α). Glycolysis suppression using 2 deoxyglucose in LPS-stimulated macrophages inhibits IL-1ß secretion, but not TNF-α, indicating metabolic pathway specificity that determines cytokine production. In contrast to LPS, the nature of the immunometabolic responses induced by non-organic particles, such as silica, in macrophages, its contribution to cytokine specification, and disease pathogenesis are not well understood. Silica-stimulated macrophages activate pattern recognition receptors (PRRs) and NLRP3 inflammasome and release IL-1ß, TNF-α, and interferons, which are the key mediators of silicosis pathogenesis. In contrast to bacteria, silica particles cannot be degraded, and the persistent macrophage activation results in an increased NADPH oxidase (Phox) activation and mitochondrial reactive oxygen species (ROS) production, ultimately leading to macrophage death and release of silica particles that perpetuate inflammation. In this manuscript, we reviewed the effects of silica on macrophage mitochondrial respiration and central carbon metabolism determining cytokine specification responsible for the sustained inflammatory responses in the lungs.


Asunto(s)
Lipopolisacáridos , Dióxido de Silicio , Lipopolisacáridos/farmacología , Lipopolisacáridos/metabolismo , Dióxido de Silicio/farmacología , Macrófagos , Activación de Macrófagos , Citocinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
17.
Adv Sci (Weinh) ; 9(35): e2204760, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36310116

RESUMEN

Polymer dielectrics are essential for advanced electrical and electronic power systems due to their ultrafast charge-discharge rate. However, a long-standing challenge is to maintain their dielectric performance at high temperatures. Here, a layered barium titanate/polyamideimide nanocomposite reinforced with rationally designed interfaces is reported for high-temperature high-energy-density dielectrics. Nanocoatings composed of 2D montmorillonite nanosheets with anisotropic conductivities are interposed at two kinds of macroscopic interfaces: 1) the interfaces between adjacent layers in the nanocomposites (inside) and 2) the interfaces between the surface of the nanocomposite and the electrode (outside). By revealing the charge transport behavior with Kelvin probe force microscope, surface potential decay, and finite element simulation, it is demonstrated that the outside nanocoatings are observed to diminish charge injection from the electrode, while the inside nanocoatings can suppress the kinetic energy of hot carriers by redirecting their transport. In this interface-reinforced nanocomposite, an ultrahigh energy density of 2.48 J cm-3 , as well as a remarkable charge-discharge efficiency >80%, is achieved at 200 °C, six times higher than that of the nanocomposite without interfacial nanocoatings. This research unveils a novel approach for the structural design of polymer nanocomposites based on engineered interfaces to achieve high-efficient and high-temperature capacitive energy storage.

18.
In Vivo ; 35(6): 3053-3066, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34697137

RESUMEN

BACKGROUND/AIM: The role of senescence and bone marrow-derived cells in silica-induced pulmonary fibrosis is unknown. MATERIALS AND METHODS: C57BL/6HNsd, p16+/LUC, and tdTOMp16+ mice were intratracheally injected with 200 mg/kg crystalline silica or irradiated (20 Gy) to the thoracic cavity and followed for the development of lung fibrosis. RESULTS: The p16+/LUC mice demonstrated senescence by day 7 after silica exposure. C57BL/6 mice exposed to silica demonstrated upregulation of p16, p21, and tyrosine kinase Fgr by day 7, whereas thoracic irradiation induced p21 and Fgr by day 50 and p16 by day 110. Silica exposed GFP+ bone marrow chimeric C57BL/6 mice demonstrated senescent cells and gfp+/Fgr+ monocyte/macrophages in the lungs on day 21. The Fgr inhibitor TL02-59 abrogated monocyte/macrophages recruitment in in vitro transwell experiments. CONCLUSION: Both silica and radiation exposure induce senescence and upregulate tyrosine kinase Fgr for the recruitment of bone marrow-derived monocyte/macrophages and the development of pulmonary fibrosis.


Asunto(s)
Fibrosis Pulmonar , Dióxido de Silicio , Animales , Médula Ósea , Senescencia Celular , Pulmón , Macrófagos , Ratones , Ratones Endogámicos C57BL , Monocitos , Fibrosis Pulmonar/inducido químicamente , Dióxido de Silicio/toxicidad
20.
Proc Natl Acad Sci U S A ; 104(26): 11002-7, 2007 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-17569781

RESUMEN

Mesenchymal stem cells (MSCs) have been exploited as cellular vectors to treat a wide array of diseases but the mechanisms responsible for their therapeutic effect remain indeterminate. Previously, we reported that MSCs inhibit bleomycin (BLM)-induced inflammation and fibrosis within the lungs of mice. Interrogation of the MSC transcriptome identified interleukin 1 receptor antagonist (IL1RN) as a potential mediator of this effect. Fractionation studies indicated that MSCs are the principal source of IL1RN in murine bone marrow and that its expression is restricted to a unique subpopulation of cells. Moreover, MSC-conditioned media was shown to block proliferation of an IL-1alpha-dependent T cell line and inhibit production of TNF-alpha by activated macrophages in vitro. Studies conducted in mice revealed that MSC administration was more effective than recombinant IL1RN delivered via adenoviral infection or osmotic pumps in inhibiting BLM-induced increases in TNF-alpha, IL-1alpha, and IL1RN mRNA in lung, IL1RN protein in bronchoalveolar lavage (BAL) fluid, and trafficking of lymphocytes and neutrophils into the lung. Therefore, MSCs protect lung tissue from BLM-induced injury by blocking TNF-alpha and IL-1, two fundamental proinflammatory cytokines in lung. Identification of IL1RN-expressing human MSC subpopulations may provide a novel cellular vector for treating chronic inflammatory diseases in humans.


Asunto(s)
Fibrosis , Inflamación , Proteína Antagonista del Receptor de Interleucina 1/fisiología , Enfermedades Pulmonares/patología , Células Madre Mesenquimatosas/fisiología , Animales , Médula Ósea , Proteína Antagonista del Receptor de Interleucina 1/biosíntesis , Interleucina-1alfa/antagonistas & inhibidores , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA