Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 223, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702815

RESUMEN

Cardiac muscle targeting is a notoriously difficult task. Although various nanoparticle (NP) and adeno-associated viral (AAV) strategies with heart tissue tropism have been developed, their performance remains suboptimal. Significant off-target accumulation of i.v.-delivered pharmacotherapies has thwarted development of disease-modifying cardiac treatments, such as gene transfer and gene editing, that may address both rare and highly prevalent cardiomyopathies and their complications. Here, we present an intriguing discovery: cargo-less, safe poly (lactic-co-glycolic acid) particles that drastically improve heart delivery of AAVs and NPs. Our lead formulation is referred to as ePL (enhancer polymer). We show that ePL increases selectivity of AAVs and virus-like NPs (VLNPs) to the heart and de-targets them from the liver. Serotypes known to have high (AAVrh.74) and low (AAV1) heart tissue tropisms were tested with and without ePL. We demonstrate up to an order of magnitude increase in heart-to-liver accumulation ratios in ePL-injected mice. We also show that ePL exhibits AAV/NP-independent mechanisms of action, increasing glucose uptake in the heart, increasing cardiac protein glycosylation, reducing AAV neutralizing antibodies, and delaying blood clearance of AAV/NPs. Current approaches utilizing AAVs or NPs are fraught with challenges related to the low transduction of cardiomyocytes and life-threatening immune responses; our study introduces an exciting possibility to direct these modalities to the heart at reduced i.v. doses and, thus, has an unprecedented impact on drug delivery and gene therapy. Based on our current data, the ePL system is potentially compatible with any therapeutic modality, opening a possibility of cardiac targeting with numerous pharmacological approaches.


Asunto(s)
Dependovirus , Vectores Genéticos , Miocardio , Nanopartículas , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Dependovirus/genética , Animales , Nanopartículas/química , Ratones , Miocardio/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química , Humanos , Ratones Endogámicos C57BL , Corazón , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Hígado/metabolismo , Tropismo Viral , Células HEK293
3.
J Cell Physiol ; 236(7): 5339-5351, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33432609

RESUMEN

Iron overload (IO) is a common yet underappreciated finding in metabolic syndrome (MetS) patients. With the prevalence of MetS continuing to rise, it is imperative to further elucidate cellular mechanisms leading to metabolic dysfunction. Adiponectin has many beneficial effects and is a therapeutic target for the treatment of MetS and cardiovascular diseases. IO positively correlates with reduced circulating adiponectin levels yet the impact of IO on adiponectin action is unknown. Here, we established a model of IO in L6 skeletal muscle cells and found that IO-induced adiponectin resistance. This was shown via reduced p38 mitogen-activated protein kinase phosphorylation in response to the small molecule adiponectin receptor (AdipoR) agonist, AdipoRon, in presence of IO. This correlated with reduced messenger RNA and protein levels of AdipoR1 and its facilitative signaling binding partner, APPL1. IO caused phosphorylation, nuclear extrusion, and thus inhibition of FOXO1, a known transcription factor regulating AdipoR1 expression. The antioxidant N-acetyl cystine attenuated the production of reactive oxygen species (ROS) by IO, and blunted its effect on FOXO1 phosphorylation and removal from the nucleus, as well as subsequent adiponectin resistance. In conclusion, our study identifies a ROS/FOXO1/AdipoR1 axis as a cause of skeletal muscle adiponectin resistance in response to IO. This new knowledge provides insight into a cellular mechanism with potential relevance to disease pathophysiology in MetS patients with IO.


Asunto(s)
Adiponectina/metabolismo , Proteína Forkhead Box O1/metabolismo , Sobrecarga de Hierro/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular , Síndrome Metabólico/metabolismo , Ratas , Receptores de Adiponectina/metabolismo
4.
Sci Rep ; 10(1): 14348, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32873817

RESUMEN

Chronic exposure to particulate matter < 2.5µ (PM2.5) has been linked to cardiopulmonary disease. Tissue-resident (TR) alveolar macrophages (AΦ) are long-lived, self-renew and critical to the health impact of inhalational insults. There is an inadequate understanding of the impact of PM2.5 exposure on the nature/time course of transcriptional responses, self-renewal of AΦ, and the contribution from bone marrow (BM) to this population. Accordingly, we exposed chimeric (CD45.2/CD45.1) mice to concentrated PM2.5 or filtered air (FA) to evaluate the impact on these end-points. PM2.5 exposure for 4-weeks induced an influx of BM-derived monocytes into the lungs with no contribution to the overall TR-AΦ pool. Chronic (32-weeks) PM2.5 exposure on the other hand while associated with increased recruitment of BM-derived monocytes and their incorporation into the AΦ population, resulted in enhanced apoptosis and decreased proliferation of TR-AΦ. RNA-seq analysis of isolated TR-AΦ and BM-AΦ from 4- and 32-weeks exposed mice revealed a unique time-dependent pattern of differentially expressed genes. PM2.5 exposure resulted in altered histological changes in the lungs, a reduced alveolar fraction which corresponded to protracted lung inflammation. Our findings suggest a time-dependent entrainment of BM-derived monocytes into the AΦ population of PM2.5 exposed mice, that together with enhanced apoptosis of TR-AΦ and reorganization of transcriptional responses, could collectively contribute to the perpetuation of chronic inflammation.


Asunto(s)
Contaminación del Aire/efectos adversos , Células de la Médula Ósea/citología , Exposición por Inhalación/efectos adversos , Macrófagos Alveolares/inmunología , Monocitos/inmunología , Neumonía/inmunología , Contaminantes Atmosféricos/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Material Particulado/efectos adversos
6.
Sci Rep ; 10(1): 13786, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32796856

RESUMEN

Biodegradable materials, including the widely used poly (lactic-co-glycolic acid) (PLGA) nanoparticles contained in slow-release drug formulations, scaffolds and implants, are ubiquitous in modern biomedicine and are considered inert or capable of being metabolized through intermediates such as lactate. However, in the presence of metabolic stress, such as in obesity, the resulting degradation products may play a detrimental role, which is still not well understood. We evaluated the effect of intravenously-administered PLGA nanoparticles on the gut-liver axis under conditions of caloric excess in C57BL/6 mice. Our results show that PLGA nanoparticles accumulate and cause gut acidification in the cecum, accompanied by significant changes in the microbiome, with a marked decrease of Firmicutes and Bacteroidetes. This was associated with transcriptomic reprogramming in the liver, with a downregulation of mitochondrial function, and an increase in key enzymatic, inflammation and cell activation pathways. No changes were observed in systemic inflammation. Metagenome analysis coupled with publicly available microarray data suggested a mechanism of impaired PLGA degradation and intestinal acidification confirming an important enterohepatic axis of metabolite-microbiome interaction resulting in maintenance of metabolic homeostasis. Thus, our results have important implications for the investigation of PLGA use in metabolically-compromised clinical and experimental settings.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Hígado/efectos de los fármacos , Obesidad/genética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/administración & dosificación , Transcriptoma/efectos de los fármacos , Administración Intravenosa , Animales , Bacteroidetes/genética , Bacteroidetes/fisiología , Materiales Biocompatibles/administración & dosificación , Materiales Biocompatibles/química , Ciego/química , Ciego/efectos de los fármacos , Ciego/microbiología , Modelos Animales de Enfermedad , Firmicutes/genética , Firmicutes/fisiología , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/fisiología , Humanos , Concentración de Iones de Hidrógeno , Hígado/metabolismo , Ratones Endogámicos C57BL , Nanopartículas/administración & dosificación , Nanopartículas/química , Obesidad/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico/química
7.
Toxicol Sci ; 169(1): 95-107, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30812033

RESUMEN

Fine ambient particulate matter (PM2.5) is able to induce sympathetic activation and inflammation in the brain. However, direct evidence demonstrating an essential role of sympathetic activation in PM2.5-associated disease progression is lacking. We assess the contribution of α2B-adrenergic receptor (Adra2b) in air pollution-associated hypertension and behavioral changes in this study. Wild-type mice and Adra2b-transgenic mice overexpressing Adra2b in the brain (Adra2bTg) were exposed to concentrated PM2.5 or filtered air for 3 months via a versatile aerosol concentrator exposure system. Mice were fed with a high salt diet (4.0% NaCl) for 1 week at week 11 of exposure to induce blood pressure elevation. Intra-arterial blood pressure was monitored by radio-telemetry and behavior changes were assessed by open field, light-dark, and prepulse inhibition tests. PM2.5 exposure increased Adra2b in the brain of wild-type mice. Adra2b overexpression enhanced the anxiety-like behavior and high salt diet-induced blood pressure elevation in response to air pollution but not filtered air exposure. Adra2b overexpression induced upregulation of inflammatory genes such as TLR2, TLR4, and IL-6 in the brain exposed to PM2.5. In addition, there were increased frequencies of activated effector T cells and increased expression of oxidative stress-related genes, such as SOD1, NQO1, Nrf2, and Gclm in Adra2bTg mice compared with wild-type mice. Our results provide new evidence of distinct behavioral changes consistent with anxiety and blood pressure elevation in response to high salt intake and air pollution exposure, highlighting the importance of centrally expressed Adra2b in the vulnerability to air pollution exposure.


Asunto(s)
Conducta Animal/efectos de los fármacos , Presión Sanguínea/efectos de los fármacos , Encéfalo/efectos de los fármacos , Hipertensión/inducido químicamente , Material Particulado/toxicidad , Receptores Adrenérgicos alfa 2/metabolismo , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Regulación de la Expresión Génica , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Mediadores de Inflamación/metabolismo , Exposición por Inhalación/efectos adversos , Locomoción/efectos de los fármacos , Masculino , Ratones Transgénicos , Estrés Oxidativo/efectos de los fármacos , Inhibición Prepulso/efectos de los fármacos , Receptores Adrenérgicos alfa 2/genética , Medición de Riesgo , Cloruro de Sodio Dietético/efectos adversos , Regulación hacia Arriba
8.
J Mol Endocrinol ; 59(4): 339-350, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28954814

RESUMEN

Skeletal muscle insulin resistance is known to play an important role in the pathogenesis of diabetes, and one potential causative cellular mechanism is endoplasmic reticulum (ER) stress. Adiponectin mediates anti-diabetic effects via direct metabolic actions and by improving insulin sensitivity, and we recently demonstrated an important role in stimulation of autophagy by adiponectin. However, there is limited knowledge on crosstalk between autophagy and ER stress in skeletal muscle and in particular how they are regulated by adiponectin. Here, we utilized the model of high insulin/glucose (HIHG)-induced insulin resistance, determined by measuring Akt phosphorylation (T308 and S473) and glucose uptake in L6 skeletal muscle cells. HIHG reduced autophagic flux measured by LC3 and p62 Western blotting and tandem fluorescent RFP/GFP-LC3 immunofluorescence (IF). HIHG also induced ER stress assessed by thioflavin T/KDEL IF, pIRE1, pPERK, peIF2α and ATF6 Western blotting and induction of a GRP78-mCherry reporter. Induction of autophagy by adiponectin or rapamycin attenuated HIHG-induced ER stress and improved insulin sensitivity. The functional significance of enhanced autophagy was validated by demonstrating a lack of improved insulin sensitivity in response to adiponectin in autophagy-deficient cells generated by overexpression of dominant negative mutant of Atg5. In summary, adiponectin-induced autophagy in skeletal muscle cells alleviated HIHG-induced ER stress and insulin resistance.


Asunto(s)
Adiponectina/metabolismo , Autofagia , Resistencia a la Insulina , Insulina/metabolismo , Animales , Autofagia/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glucosa/metabolismo , Humanos , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Sirolimus/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
9.
PLoS One ; 11(11): e0165962, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27812198

RESUMEN

Obesity in humans and mice is typified by an activated macrophage phenotype in the visceral adipose tissue (VAT) leading to increased macrophage-mediated inflammation. microRNAs (miRNAs) play an important role in regulating inflammatory pathways in macrophages, and in this study we compared miRNA expression in the VAT of insulin resistant morbidly obese humans to a non-obese cohort with normal glucose tolerance. miR-223-3p was found to be significantly upregulated in the whole omental tissue RNA of 12 human subjects, as were 8 additional miRNAs. We then confirmed that miR-223 upregulation was specific to the stromal vascular cells of human VAT, and found that miR-223 levels were unchanged in adipocytes and circulating monocytes of the non-obese and obese. miR-223 ablation increased basal / unstimulated TLR4 and STAT3 expression and LPS-stimulated TLR4, STAT3, and NOS2 expression in primary macrophages. Conversely, miR-223 mimics decreased TLR4 expression in primary macrophage, at the same time it negatively regulated FBXW7 expression, a well described suppressor of Toll-like receptor 4 (TLR4) signaling. We concluded that the abundance of miR-223 in macrophages significantly modulates macrophage phenotype / activation state and response to stimuli via effects on the TLR4/FBXW7 axis.


Asunto(s)
Grasa Intraabdominal/metabolismo , Macrófagos/inmunología , MicroARNs/genética , Obesidad/genética , Obesidad/inmunología , Regulación hacia Arriba , Adulto , Animales , Proteínas de Ciclo Celular/metabolismo , Estudios de Cohortes , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Células HeLa , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Resistencia a la Insulina , Activación de Macrófagos , Masculino , Ratones , Persona de Mediana Edad , Óxido Nítrico Sintasa de Tipo II/metabolismo , Obesidad/patología , Fenotipo , Receptor Toll-Like 4/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
10.
Int J Cardiol ; 216: 32-42, 2016 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-27140334

RESUMEN

BACKGROUND: The cardiac remodelling process in advanced heart failure due to pressure overload has not been clearly defined but likely involves mechanisms of cardiac fibrosis and cardiomyocyte hypertrophy. The aim of this study was to examine pressure overload (PO)-induced cardiac remodelling processes and their reversibility after unloading in both humans with heart failure and a mouse model of PO induced by aortic constriction. METHODS & RESULTS: Speckle tracking echocardiography showed PO-induced cardiac dysfunction in mice was reversible after removal of aortic constriction to unload. Masson's Trichrome staining suggested that PO-induced myocardial fibrosis was reversible, however detailed analysis of 3-dimensional collagen architecture by scanning electron microscopy demonstrated that matrix remodelling was not completely normalised as a disorganised network of thin collagen fibres was evident. Analysis of human left ventricular biopsy samples from HF patients revealed increased presence of large collagen fibres which were greatly reduced in paired samples from the same individuals after unloading by left ventricular assist device implantation. Again, an extensive network of small collagen fibres was still clearly seen to closely surround cardiomyocytes after unloading. Other features of PO-induced remodelling including increased myofibroblast content, cardiomyocyte disarray and hypertrophy were largely reversed upon unloading in both humans and mouse model. Previous work in humans demonstrated that receptors for adiponectin, an important mediator of cardiac fibrosis and hypertrophy, decreased in heart failure patients and returned to normal after unloading. Here we provide novel data showing a similar trend for adiponectin receptor adaptor protein APPL1, but not APPL2 isoform. CONCLUSIONS: LV unloading diminishes PO-induced cardiac remodelling and improves function. These findings add new insights into the cardiac remodelling process, and provide novel targets for future pharmacologic therapies.


Asunto(s)
Estenosis de la Válvula Aórtica/complicaciones , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Colágeno/ultraestructura , Insuficiencia Cardíaca/patología , Ventrículos Cardíacos/diagnóstico por imagen , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Cardiomiopatía Hipertrófica/etiología , Cardiomiopatía Hipertrófica/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Ventrículos Cardíacos/metabolismo , Humanos , Masculino , Ratones , Microscopía Electrónica de Rastreo , Remodelación Ventricular
11.
J Immunol ; 190(9): 4795-804, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23554310

RESUMEN

Mucosal inflammation in conditions ranging from infective acute enteritis or colitis to inflammatory bowel disease is accompanied by alteration in serotonin (5-hydroxytryptamine [5-HT]) content in the gut. Recently, we have identified an important role of 5-HT in the pathogenesis of experimental colitis. 5-HT type 7 (5-HT7) receptor is one of the most recently identified members of the 5-HT receptor family, and dendritic cells express this receptor. In this study, we investigated the effect of blocking 5-HT7 receptor signaling in experimental colitis with a view to develop an improved therapeutic strategy in intestinal inflammatory disorders. Colitis was induced with dextran sulfate sodium (DSS) or dinitrobenzene sulfonic acid (DNBS) in mice treated with selective 5-HT7 receptor antagonist SB-269970, as well as in mice lacking 5-HT7 receptor (5-HT7(-/-)) and irradiated wild-type mice reconstituted with bone marrow cells harvested from 5-HT7(-/-) mice. Inhibition of 5-HT7 receptor signaling with SB-269970 ameliorated both acute and chronic colitis induced by DSS. Treatment with SB-269970 resulted in lower clinical disease, histological damage, and proinflammatory cytokine levels compared with vehicle-treated mice post-DSS. Colitis severity was significantly lower in 5-HT7(-/-) mice and in mice reconstituted with bone marrow cells from 5-HT7(-/-) mice compared with control mice after DSS colitis. 5-HT7(-/-) mice also had significantly reduced DNBS-induced colitis. These observations provide us with novel information on the critical role of the 5-HT7 receptor in immune response and inflammation in the gut, and highlight the potential benefit of targeting this receptor to alleviate the severity of intestinal inflammatory disorders such as inflammatory bowel disease.


Asunto(s)
Inflamación/inmunología , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Receptores de Serotonina/inmunología , Receptores de Serotonina/metabolismo , Animales , Bencenosulfonatos/farmacología , Colitis/inducido químicamente , Colitis/inmunología , Colitis/metabolismo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Inflamación/inducido químicamente , Intestinos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/inmunología , FN-kappa B/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA