Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Acta Biomater ; 168: 429-439, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37499727

RESUMEN

Devices capable of recording or stimulating neuronal signals have created new opportunities to understand normal physiology and treat sources of pathology in the brain. However, it is possible that the tissue response to implanted electrodes may influence the nature of the signals detected or stimulated. In this study, we characterized structural and functional changes in deep layer pyramidal neurons surrounding silicon or polyimide-based electrodes implanted in the motor cortex of rats. Devices were captured in 300 µm-thick tissue slices collected at the 1 or 6 week time point post-implantation, and individual neurons were assessed using a combination of whole-cell electrophysiology and 2-photon imaging. We observed disrupted dendritic arbors and a significant reduction in spine densities in neurons surrounding devices. These effects were accompanied by a decrease in the frequency of spontaneous excitatory post-synaptic currents, a reduction in sag amplitude, an increase in spike frequency adaptation, and an increase in filopodia density. We hypothesize that the effects observed in this study may contribute to the signal loss and instability that often accompany chronically implanted electrodes. STATEMENT OF SIGNIFICANCE: Implanted electrodes in the brain can be used to treat sources of pathology and understand normal physiology by recording or stimulating electrical signals generated by local neurons. However, a foreign body response following implantation undermines the performance of these devices. While several studies have investigated the biological mechanisms of device-tissue interactions through histology, transcriptomics, and imaging, our study is the first to directly interrogate effects on the function of neurons surrounding electrodes using single-cell electrophysiology. Additionally, we provide new, detailed assessments of the impacts of electrodes on the dendritic structure and spine morphology of neurons, and we assess effects for both traditional (silicon) and newer polymer electrode materials. These results reveal new potential mechanisms of electrode-tissue interactions.


Asunto(s)
Corteza Motora , Ratas , Animales , Microelectrodos , Corteza Motora/fisiología , Silicio , Neuronas , Células Piramidales , Electrodos Implantados
2.
Artículo en Inglés | MEDLINE | ID: mdl-31962186

RESUMEN

Stress reactivity and glucocorticoid signaling alterations are reported in mouse models of autism spectrum disorder (ASD). BALB/c mice display decreased locomotor activity in the presence of stimulus mice and spend less time exploring enclosed stimulus mice; this mouse strain has been validated as an ASD model. VU0410120, a glycine type 1 transporter (GlyT1) inhibitor, improved sociability in BALB/c mice, consistent with data that NMDA Receptor (NMDAR) activation regulates sociability, and the endogenous tone of NMDAR-mediated neurotransmission is altered in this strain. Effects of a prosocial dose of VU0410120 on conspecific-provoked immobility, and relationships between conspecific-provoked immobility and corticosterone response were explored. VU0410120-treated BALB/c mice showed reduced immobility in the presence of conspecifics and increased the conspecific-provoked corticosterone response. However, the intensity of conspecific-provoked immobility in VU0410120-treated BALB/c mice did not differ as a function of corticosterone response. Expression profiles of 88 glucocorticoid signaling associated genes within frontal cortex and hippocampus were examined. BALB/c mice resistant to prosocial effects of VU0410120 had increased mRNA expression of Ddit4, a negative regulator of mTOR signaling. Dysregulated mTOR signaling activity is a convergent finding in several monogenic syndromic forms of ASD. Prosocial effects of VU0410120 in the BALB/c strain may be related to regulatory influences of NMDAR-activation on mTOR signaling activity. Because corticosterone response is a marker of social stress, the current data suggest that the stressfulness of a social encounter alone may not be the sole determinant of increased immobility in BALB/c mice; this strain may also display an element of social disinterest.


Asunto(s)
Corteza Cerebral/metabolismo , Corticosterona/sangre , Glucocorticoides/biosíntesis , Proteínas de Transporte de Glicina en la Membrana Plasmática/antagonistas & inhibidores , Hipocampo/metabolismo , Inmovilización/fisiología , Animales , Benzamidas/farmacología , Corteza Cerebral/efectos de los fármacos , Expresión Génica , Glucocorticoides/genética , Hipocampo/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Piperidinas/farmacología , Receptores de N-Metil-D-Aspartato/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 39(10): 2168-2191, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31434495

RESUMEN

OBJECTIVE: Endothelial cells (EC) in obese adipose tissue (AT) are exposed to a chronic proinflammatory environment that may induce a mesenchymal-like phenotype and altered function. The objective of this study was to establish whether endothelial-to-mesenchymal transition (EndoMT) is present in human AT in obesity and to investigate the effect of such transition on endothelial function and the endothelial particulate secretome represented by extracellular vesicles (EV). Approach and Results: We identified EndoMT in obese human AT depots by immunohistochemical co-localization of CD31 or vWF and α-SMA (alpha-smooth muscle actin). We showed that AT EC exposed in vitro to TGF-ß (tumor growth factor-ß), TNF-α (tumor necrosis factor-α), and IFN-γ (interferon-γ) undergo EndoMT with progressive loss of endothelial markers. The phenotypic change results in failure to maintain a tight barrier in culture, increased migration, and reduced angiogenesis. EndoMT also reduced mitochondrial oxidative phosphorylation and glycolytic capacity of EC. EVs produced by EC that underwent EndoMT dramatically reduced angiogenic capacity of the recipient naïve ECs without affecting their migration or proliferation. Proteomic analysis of EV produced by EC in the proinflammatory conditions showed presence of several pro-inflammatory and immune proteins along with an enrichment in angiogenic receptors. CONCLUSIONS: We demonstrated the presence of EndoMT in human AT in obesity. EndoMT in vitro resulted in production of EV that transferred some of the functional and metabolic features to recipient naïve EC. This result suggests that functional and molecular features of EC that underwent EndoMT in vivo can be disseminated in a paracrine or endocrine fashion and may induce endothelial dysfunction in distant vascular beds.


Asunto(s)
Tejido Adiposo/irrigación sanguínea , Transición Epitelial-Mesenquimal/genética , Neovascularización Patológica/genética , Obesidad/genética , Factor de Crecimiento Transformador beta1/farmacología , Tejido Adiposo/metabolismo , Análisis de Varianza , Biomarcadores/metabolismo , Estudios de Casos y Controles , Movimiento Celular/genética , Proliferación Celular/genética , Células Cultivadas , Células Endoteliales/metabolismo , Transición Epitelial-Mesenquimal/efectos de los fármacos , Citometría de Flujo/métodos , Humanos , Obesidad/fisiopatología , Proteómica/métodos
4.
J Vis Exp ; (137)2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30080200

RESUMEN

Obesity is accompanied by an extensive remodeling of adipose tissue primarily via adipocyte hypertrophy. Extreme adipocyte growth results in a poor response to insulin, local hypoxia, and inflammation. By stimulating the differentiation of functional white adipocytes from progenitors, radical hypertrophy of the adipocyte population can be prevented and, consequently, the metabolic health of adipose tissue can be improved along with a reduction of inflammation. Also, by stimulating a differentiation of beige/brown adipocytes, the total body energy expenditure can be increased, resulting in weight loss. This approach could prevent the development of obesity co-morbidities such as type 2 diabetes and cardiovascular disease. This paper describes the isolation, expansion, and differentiation of white and beige adipocytes from a subset of human adipose tissue endothelial cells that co-express the CD31 and CD34 markers. The method is relatively cheap and is not labor-intensive. It requires access to human adipose tissue and the subcutaneous depot is suitable for sampling. For this protocol, fresh adipose tissue samples from morbidly obese subjects [body mass index (BMI) >35] are collected during bariatric surgery procedures. Using a sequential immunoseparation from the stromal vascular fraction, enough cells are produced from as little as 2-3 g of fat. These cells can be expanded in culture over 10-14 days, can be cryopreserved, and retain their adipogenic properties with passaging up to passage 5-6. The cells are treated for 14 days with an adipogenic cocktail using a combination of human insulin and the PPARγ agonist-rosiglitazone. This methodology can be used for obtaining proof of concept experiments on molecular mechanisms that drive adipogenic responses in adipose endothelial cells, or for screening new drugs that can enhance the adipogenic response directed either towards white or beige/brown adipocyte differentiation. Using small subcutaneous biopsies, this methodology can be used to screen out non-responder subjects for clinical trials aimed to stimulate beige/brown and white adipocytes for the treatment of obesity and co-morbidities.


Asunto(s)
Antígenos CD34/metabolismo , Células Endoteliales/metabolismo , Obesidad Mórbida/fisiopatología , Epiplón/metabolismo , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Grasa Subcutánea/metabolismo , Animales , Diferenciación Celular , Humanos , Masculino , Grasa Subcutánea/patología
5.
Artículo en Inglés | MEDLINE | ID: mdl-29605541

RESUMEN

The 12-lipoxygenase (12LO) pathway is a promising target to reduce islet dysfunction, adipose tissue (AT) inflammation and insulin resistance. Optimal pre-clinical models for the investigation of selective12LO inhibitors in this context have not yet been identified. The objective of this study was to characterize the time course of 12LO isoform expression and metabolite production in pancreatic islets and AT of C57BLKS/J-db/db obese diabetic mouse in a pre-diabetic state in order to establish a suitable therapeutic window for intervention with selective lipoxygenase inhibitors. Mice have 2 major 12LO isoforms -the leukocyte type (12/15LO) and the platelet type (p12LO) and both are expressed in islets and AT. We found a sharp increase in protein expression of 12/15LO in the pancreatic islets of 10-week old db-/- mice compared to 8- week old counterparts. Immunohistochemistry showed that the increase in islet 12/15LO parallels a decline in islet number. Analysis of 12- and 15-hydroperoxytetraeicosanoid acids (HETE)s showed a 2-3 fold increase especially in 12(S)-HETE that mirrored the increase in 12/15LO expression in islets. Analysis of AT and stromal vascular fraction (SVF) showed a significant increase of platelet 12LO gene expression along with 12- and 15- HETEs. The data demonstrate that the db/db mouse is a suitable model for investigation of 12/15LO inhibitors in the development of inflammatory mediated type 2 diabetes, with a narrow window of therapeutic intervention prior to 8 weeks of age.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Diabetes Mellitus Tipo 2/enzimología , Células Secretoras de Insulina/enzimología , Inhibidores de la Lipooxigenasa/farmacología , Estado Prediabético/enzimología , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/patología , Activación Enzimática/efectos de los fármacos , Células Secretoras de Insulina/patología , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Obesos , Estado Prediabético/tratamiento farmacológico , Estado Prediabético/patología
6.
Mediators Inflamm ; 2017: 5420718, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28400678

RESUMEN

Visceral adipose tissue (AT) inflammation is linked to the complications of obesity, including insulin resistance (IR) and type 2 diabetes. Recent data from our lab showed that germline deficiency in STAT4 reduces inflammation and improves IR in obese mice. The objective of this study was to determine the contribution of selective STAT4 deficiency in subsets of hematopoietic cells to IR and AT inflammation. To determine the contribution of hematopoietic lineage, we sublethally irradiated Stat4-/-C57Bl6 mice and reconstituted them with bone marrow cells (BMC) from Stat4+/+C57Bl6 congenic donors. We also established the contribution of selective STAT4 deficiency in CD4+ or CD8+ T cells using adoptive transfer in Rag1-/- mice. All mice received a HFD for 15 weeks (n = 7-12 mice/group). BMC that expressed STAT4 induced increases in glucose intolerance and IR compared to STAT4-deficient cells. Also, AT inflammation was increased and the numbers of CD8+ cells infiltrating AT were higher in mice with STAT4 expressing BMC. Studies in Rag1-/- mice further confirmed the prominent role of CD8+ cells expressing STAT4 in insulin resistance and AT and islet inflammation. Collectively our results show specific and dominant contribution of STAT4 in the hematopoietic compartment to metabolic health and inflammation in diet-induced obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Inflamación/metabolismo , Factor de Transcripción STAT4/metabolismo , Adipocitos/metabolismo , Animales , Western Blotting , Citometría de Flujo , Hematopoyesis/genética , Hematopoyesis/fisiología , Inflamación/genética , Insulina/metabolismo , Resistencia a la Insulina/genética , Resistencia a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Transcripción STAT4/genética
7.
J Clin Endocrinol Metab ; 99(9): E1713-20, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24955608

RESUMEN

CONTEXT: Visceral adipose tissue (VAT) is a key contributor to chronic inflammation in obesity. The 12/15-lipoxygenase pathway (ALOX) is present in adipose tissue (AT) and leads to inflammatory cascades that are causal for the onset of insulin resistance in rodent models of obesity. OBJECTIVE: The pathophysiology of the ALOX 12/15 pathway in human AT is unknown. We characterized the ALOX pathway in different AT depots in obese humans with or without type 2 diabetes (T2D). DESIGN: This study includes a cross-sectional cohort of 46 morbidly obese (body mass index >39 kg/m(2)) nondiabetic (n = 25) and T2D (n = 21) subjects. SETTING: This study was conducted at Eastern Virginia Medical School (Norfolk, Virginia) in collaboration with Sentara Metabolic and Weight Loss Surgery Center (Sentara Medical Group, Norfolk, Virginia). PATIENTS: Twenty-five obese (body mass index 44.8 ± 4.4 kg/m(2)) nondiabetic (hemoglobin A1c 5.83% ± 0.27%) and 21 obese (43.4 ± 4.1 kg/m(2)) and T2D (hemoglobin A1c 7.66% ± 1.22%) subjects were included in the study. The subjects were age matched and both groups had a bias toward female gender. MAIN OUTCOMES AND MEASURES: Expression of ALOX isoforms along with fatty acid substrates and downstream lipid metabolites were measured. Correlations with depot-specific inflammatory markers were also established. RESULTS: ALOX 12 expression and its metabolite 12(S)-hydroxyeicosatetraenoic acid were significantly increased in the VAT of T2D subjects. ALOX 15A was exclusively expressed in VAT in both groups. ALOX 12 expression positively correlated with expression of inflammatory genes IL-6, IL-12a, CXCL10, and lipocalin-2. CONCLUSIONS: ALOX 12 may have a critical role in regulation of inflammation in VAT in obesity and T2D. Selective ALOX 12 inhibitors may constitute a new approach to limit AT inflammation in human obesity.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato 15-Lipooxigenasa/metabolismo , Diabetes Mellitus Tipo 2/enzimología , Grasa Intraabdominal/enzimología , Obesidad Mórbida/enzimología , Adolescente , Adulto , Anciano , Araquidonato 12-Lipooxigenasa/genética , Araquidonato 15-Lipooxigenasa/genética , Biomarcadores/metabolismo , Estudios Transversales , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Ácidos Grasos/metabolismo , Femenino , Regulación Enzimológica de la Expresión Génica/inmunología , Hemoglobina Glucada/metabolismo , Humanos , Inflamación/enzimología , Inflamación/genética , Inflamación/inmunología , Grasa Intraabdominal/inmunología , Metabolismo de los Lípidos/genética , Metabolismo de los Lípidos/inmunología , Masculino , Persona de Mediana Edad , Obesidad Mórbida/genética , Obesidad Mórbida/inmunología , Adulto Joven
8.
Diabetes ; 62(12): 4109-21, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23939393

RESUMEN

Signal transducer and activator of transcription (STAT) 4 is one of the seven members of the STAT family. STAT4 has a prominent role in mediating interleukin-12-induced T-helper cell type 1 lineage differentiation. T cells are key players in the maintenance of adipose tissue (AT) inflammation. The role of STAT4 in obesity and AT inflammation is unknown. We sought to determine the role of STAT4 in AT inflammation in obesity-induced insulin resistance. We studied STAT4-null mice on the C57Bl6/J background. We have found that STAT4(-/-)C57Bl6/J mice develop high-fat diet-induced obesity (DIO) similar to wild-type controls, but that they have significantly improved insulin sensitivity and better glucose tolerance. Using flow cytometry and real-time PCR, we show that STAT4(-/-) mice with DIO produce significantly reduced numbers of inflammatory cytokines and chemokines in adipocytes, have reduced numbers of CD8(+) cells, and display increased alternative (M2) macrophage polarization. CD8(+) cells, but not CD4(+) cells, from STAT4(-/-) mice displayed reduced in vitro migration. Also, we found that adipocyte inflammation is reduced and insulin signaling is improved in STAT4(-/-) mice with DIO. We have identified STAT4 as a key contributor to insulin resistance and AT inflammation in DIO. Targeting STAT4 activation could be a novel approach to reducing AT inflammation and insulin resistance in obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina/genética , Obesidad/metabolismo , Factor de Transcripción STAT4/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa , Inflamación/genética , Inflamación/patología , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/patología , Factor de Transcripción STAT4/genética
9.
J Biol Chem ; 275(19): 14700-7, 2000 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-10799558

RESUMEN

To identify genes that are transcriptionally activated when human macrophages accumulate excess lipids, we employed the mRNA differential display technique using RNA isolated from human monocyte-macrophages incubated in the absence or presence of acetylated low density lipoprotein and sterols (cholesterol and 25-hydroxycholesterol). These studies identified a mRNA whose levels were highly induced in lipid-loaded macrophages. The mRNA encoded the human White protein, a member of the ATP-binding cassette (ABC) transporter superfamily of proteins. The mRNA levels of ABC8, the murine homolog of the human white gene, were also induced when a murine macrophage cell line, RAW264.7, was incubated with acetylated low density lipoprotein and sterols. Additional studies demonstrated that white/ABC8 mRNA levels were induced by specific oxysterols that included 25-, 20(S)-, and 22(R)-hydroxycholesterol, and by a retinoid X receptor-specific ligand. Furthermore, the oxysterol-mediated induction of ABC8 expression in mouse peritoneal macrophages was dependent on the presence of the nuclear oxysterol receptors, liver X receptors (LXRs). Macrophages derived from mice lacking both LXRalpha and LXRbeta failed to up-regulate the expression of ABC8 following incubation with 22(R)-hydroxycholesterol. Oxysterol-dependent induction of white/ABC8 mRNA was blocked by actinomycin D but not by cycloheximide treatment of cells. We conclude that the white and ABC8 genes are primary response genes that are transcriptionally activated by specific oxysterols and that this induction is mediated by the LXR subfamily of nuclear hormone receptors. These data strongly support the hypothesis that white/ABC8 has a role in cellular sterol homeostasis.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Macrófagos/metabolismo , Isoformas de Proteínas/genética , ARN Mensajero/genética , Esteroles/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Proteínas de Unión al ADN , Regulación de la Expresión Génica , Homeostasis , Humanos , Receptores X del Hígado , Ratones , Ratones Noqueados , Receptores Nucleares Huérfanos , ARN Mensajero/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Transcripción Genética/efectos de los fármacos
10.
J Biol Chem ; 275(22): 16543-9, 2000 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-10748047

RESUMEN

The synthesis of 7alpha-hydroxylated bile acids from oxysterols requires an oxysterol 7alpha-hydroxylase encoded by the Cyp7b1 locus. As expected, mice deficient in this enzyme have elevated plasma and tissue levels of 25- and 27-hydroxycholesterol; however, levels of another major oxysterol, 24-hydroxycholesterol, are not increased in these mice, suggesting the presence of another oxysterol 7alpha-hydroxylase. Here, we describe the cloning and characterization of murine and human cDNAs and genes that encode a second oxysterol 7alpha-hydroxylase. The genes contain 12 exons and are located on chromosome 6 in the human (CYP39A1 locus) and in a syntenic position on chromosome 17 in the mouse (Cyp39a1 locus). CYP39A1 is a microsomal cytochrome P450 enzyme that has preference for 24-hydroxycholesterol and is expressed in the liver. The levels of hepatic CYP39A1 mRNA do not change in response to dietary cholesterol, bile acids, or a bile acid-binding resin, unlike those encoding other sterol 7alpha-hydroxylases. Hepatic CYP39A1 expression is sexually dimorphic (female > male), which is opposite that of CYP7B1 (male > female). We conclude that oxysterol 7alpha-hydroxylases with different substrate specificities exist in mice and humans and that sexually dimorphic expression patterns of these enzymes in the mouse may underlie differences in bile acid metabolism between the sexes.


Asunto(s)
Sistema Enzimático del Citocromo P-450/genética , Hidroxicolesteroles/metabolismo , Esteroide Hidroxilasas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células CHO , Clonación Molecular , Cricetinae , Sistema Enzimático del Citocromo P-450/metabolismo , ADN Complementario , Femenino , Cromatografía de Gases y Espectrometría de Masas , Humanos , Masculino , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido , Esteroide Hidroxilasas/metabolismo
11.
Theriogenology ; 41(5): 1045-52, 1994.
Artículo en Inglés | MEDLINE | ID: mdl-16727457

RESUMEN

Thirty crossbred bulls, 12 to 13 mo of age, were used to examine the relationship of testosterone and progesterone concentrations and testosterone: progesterone ratio to measurements of testicular function. Bulls were allotted to 1 of 2 groups based on scrotal circumferences (SC) as follows: the Small SC (n=20) group had scrotal circumference less than 28 cm while the Large SC (n=10) group had scrotal circumference greater than 28 cm. All bulls were administered GnRH (100 mug, im), and blood was obtained immediately prior to injection (t=0), 30 min after injection (t=30) and 2 to 3 h after injection (t=150). Serum was assayed for concentrations of testosterone and progesterone. Semen was evaluated for the percentage of morphologically normal spermatozoa. Testicular parenchyma was sectioned and stained, and 300 cross sections per testis of seminiferous tubules were examined under a light microscope and classified as either active (spermatocytes and spermatids present) or inactive (no spermatocytes or spermatids present). Although progesterone concentrations varied widely (range: 21 pg/ml to 1070 pg/ml), repeated measurements from individual bulls were highly correlated (r(2)=0.74) and did not change significantly (P > 0.1) in response to GnRH treatment. Small SC bulls had a higher percentage of inactive seminiferous tubules (P < 0.001) and a lower percentage morphologically normal spermatozoa (P < 0.001) than Large SC bulls, but no differences in testosterone or progesterone concentrations or in the ratio of testosterone: progesterone were detected. Mean serum testosterone concentration increased (P < 0.0001) by 30 min after GnRH treatment and continued to increase (P < 0.0001) through t=150 but did not differ (p > 0.1) between groups. Normal testosterone secretion in response to GnRH injection suggested that no biochemical lesions in the testosterone production pathway were present in bulls with very small scrotal circumference.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA