Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Cell Mol Neurobiol ; 38(4): 901-917, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29177613

RESUMEN

Omega-3 and omega-6 polyunsaturated fatty acids (PUFAs) are necessary for functional cell integrity. Preconditioning (PC), as we define it, is an acquired protection or resilience by a cell, tissue, or organ to a lethal stimulus enabled by a previous sublethal stressor or stimulus. In this study, we provide evidence that the omega-3 fatty acid docosahexaenoic acid (DHA) and its derivatives, the docosanoids 17-hydroxy docosahexaenoic acid (17-HDHA) and neuroprotectin D1 (NPD1), facilitate cell survival in both in vitro and in vivo models of retinal PC. We also demonstrate that PC requires the enzyme 15-lipoxygenase-1 (15-LOX-1), which synthesizes 17-HDHA and NPD1, and that this is specific to docosanoid signaling despite the concomitant release of the omega-6 arachidonic acid and eicosanoid synthesis. These findings advocate that DHA and docosanoids are protective enablers of PC in photoreceptor and retinal pigment epithelial cells.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Fármacos Neuroprotectores/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Animales , Araquidonato 15-Lipooxigenasa/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Retina/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
2.
Exp Eye Res ; 92(2): 155-60, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21035444

RESUMEN

Spectral Domain Optical Coherence Tomography (SD-OCT) applied to the mouse retina has been limited due to inherent movement artifacts and lack of resolution. Recently, SD-OCT scans from a commercially available imaging system have yielded retinal thickness values comparable to histology. However, these measurements are based on single point analysis of images. Here we report that using the Spectralis HRA + OCT Spectral Domain OCT and Fluorescein Angiography system (Heidelberg Engineering, Heidelberg, Germany), retinal thickness of linear expanses from SD-OCT data can be accurately assessed. This is possible by the development of a Spectralis-compatible ImageJ plug-in that imports 8-bit SLO and 32-bit OCT B-scan images, retaining scale and segmentation data and enabling analysis and 3D reconstruction. Moreover, mouse retinal layer thickness values obtained with this plug-in exhibit a high correlation to thickness measurements from histology of the same retinas. Thus, use of this ImageJ plug-in results in reliable quantification of long retinal expanses from in vivo SD-OCT images.


Asunto(s)
Células Fotorreceptoras de Vertebrados/citología , Epitelio Pigmentado de la Retina/citología , Tomografía de Coherencia Óptica , Animales , Antropometría , Biometría/métodos , Angiografía con Fluoresceína , Masculino , Ratones , Ratones Endogámicos BALB C
3.
Exp Eye Res ; 93(5): 636-48, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21854772

RESUMEN

Retinal pathologies common to human eye diseases, including abnormal retinal pigment epithelial (RPE) cells, drusen-like accumulation, photoreceptor atrophy, and choroidal neovascularization, have been reported in the Ccl2/Cx3cr1-deficient mouse. The Ccl2 gene encodes the pro-inflammatory chemokine CCL2 (MCP-1), which is responsible for chemotactic recruitment of monocyte-derived macrophages to sites of inflammation. The Cx3cr1 gene encodes the fractalkine receptor, CX3CR1, and is required for accumulation of monocytes and microglia recruited via CCL2. Chemokine-mediated inflammation is implicated in retinal degenerative diseases such as diabetic retinopathy, age-related macular degeneration, retinitis pigmentosa, and uveoretinitis, and proper chemokine signaling from the RPE, Müller glia, and astrocytes is necessary to regulate leukocyte trafficking. Therefore, this mouse, possessing aberrant chemokine signaling coupled with retinal degenerative pathologies, presents an ideal opportunity to investigate the effect of altered signaling on retinal homeostasis and photoreceptor degeneration. Since this mouse is a recent development, more data covering the onset, location, and progression rate of pathologies is needed. In the present study we establish these parameters and show two photoreceptor cell death processes. Our observations of decreased glutamine synthetase and increased glial fibrillary acidic protein suggest that Müller cells respond very early within regions where lesions are forming. Finally, we suggest that retinal angiomatous proliferation contributes to pathological angiogenesis in this Ccl2/Cx3cr1-deficient mouse.


Asunto(s)
Quimiocina CCL2/fisiología , Modelos Animales de Enfermedad , Células Fotorreceptoras de Vertebrados/patología , Receptores de Quimiocina/fisiología , Degeneración Retiniana/patología , Tomografía de Coherencia Óptica , Animales , Western Blotting , Receptor 1 de Quimiocinas CX3C , Progresión de la Enfermedad , Angiografía con Fluoresceína , Proteína Ácida Fibrilar de la Glía , Gliosis/metabolismo , Gliosis/patología , Glutamato-Amoníaco Ligasa/metabolismo , Imagenología Tridimensional , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas del Tejido Nervioso/metabolismo , Oftalmoscopía , Degeneración Retiniana/metabolismo
4.
Mol Vis ; 16: 320-9, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20216940

RESUMEN

PURPOSE: To examine the effects of neuroprotectin D1 (NPD1), a stereospecific derivative of docosahexaenoic acid, on choroidal neovascularization (CNV) in a laser-induced mouse model. Specifically, this was assessed by clinically grading laser-induced lesions, measuring leakage area, and volumetrically quantifying vascular endothelial cell proliferation. METHODS: C57Bl/6 mice were treated with vehicle control or NPD1, and choroidal neovascularization was induced by laser rupture of Bruch's membrane; treatment was administered throughout the first week of recovery. One and two weeks after CNV induction, fundus fluorescein angiography was performed. Angiograms were clinically graded to assess leakage severity, while leakage area was measured by image analysis of angiograms. Proliferation of vascular endothelial cells was evaluated volumetrically by three-dimensional laser confocal immunofluorescent microscopy of cytoskeletal, nuclear, and endothelial cell markers. RESULTS: At seven days after CNV induction, NPD1-treated mice had 60% fewer clinically relevant lesions than controls, dropping to 80% fewer by 14 days. NPD1 mice exhibited 25% smaller leakage area than controls at 7 days and 44% smaller area at 14 days. Volumetric immunofluorescence revealed 46% less vascular endothelial cell volume in 7-day NPD1-treated mice than in 7-day controls, and by 14 days NPD1 treatment was 68% lower than controls. Furthermore, comparison of 7- and 14-day volumes of NPD1-treated mice revealed a 50% reduction at 14 days. CONCLUSIONS: NPD1 significantly inhibits choroidal neovascularization. There are at least two possible mechanisms that could explain the neuroprotective action of NPD1. Ultimately, nuclear factor-kappaB could be inhibited with a reduction in cyclooxygenase-2 (COX-2) to reduce vascular endothelial growth factor (VEGF) expression, and/or activation of the resolution phase of the inflammatory response/survival pathways could be upregulated. Moreover, NPD1 continues to be effective after treatment is concluded, suggesting sustained protection and highlighting the potential applicability of this lipid mediator in preventing or ameliorating endothelial cell growth in pathoangiogenesis.


Asunto(s)
Neovascularización Coroidal/prevención & control , Ácidos Docosahexaenoicos/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Tamaño de la Célula/efectos de los fármacos , Neovascularización Coroidal/inducido químicamente , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Fluoresceína/metabolismo , Angiografía con Fluoresceína , Fondo de Ojo , Rayos Láser , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Retina/efectos de los fármacos , Retina/patología
5.
Brain Circ ; 6(4): 260-268, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33506149

RESUMEN

OBJECTIVE: Acute ischemic stroke triggers complex neurovascular, neuroinflammatory, and synaptic alterations. This study explores whether blocking pro-inflammatory platelet-activating factor receptor (PAF-R) plus selected docosanoids after middle cerebral artery occlusion (MCAo) would lead to neurological recovery. The following small molecules were investigated: (a) LAU-0901, a PAF-R antagonist that blocks pro-inflammatory signaling; and (b) derivatives of docosahexaenoic acid (DHA), neuroprotectin D1 (NPD1), and aspirin-triggered NPD1 (AT-NPD1), which activates cell survival pathways and are exert potent anti-inflammatory activity in the brain. MATERIALS AND METHODS: Sprague-Dawley rats received 2 h MCAo and LAU-0901 (30 or 60 mg/kg, 2 h after stroke), NPD1, and AT-NPD1 (333 µg/kg), DHA (5 mg/kg), and their combination were administered intravenous at 3 h after stroke. Behavior testing and ex vivo magnetic resonance imaging were conducted on day 3 or 14 to assess lesion characteristics and lipidomic analysis on day 1. Series 1 (LAU-0901 + NPD1, 14d), Series 2 (LAU-0901 + AT-NPD1, 3d), and Series 3 (LAU-0901 + DHA, 1d). RESULTS: All combinatory groups improved behavior compared to NPD1, AT-NPD1, or DHA treatments alone. Total lesion volumes were reduced with LAU-0901 + NPD1 by 62% and LAU-0901 + AT-NPD1 by 90% treatments versus vehicle groups. LAU-0901 and LAU-0901 + DHA increased the production of vasoactive lipid mediators (prostaglandins: PGE2, PGF2- α, 6-keto-PGF1- α, and PGD2) as well an inflammatory regulating mediator hydroxyoctadecadienoic acid. In contrast, LAU-0901 and LAU-0901 + DHA decreased the production of 12-hydroxyeicosatetraenoic acid, a pro-inflammatory mediator. CONCLUSION: Combination therapy with LAU-0901 and selected docosanoids is more effective than the single therapy, affording synergistic neuroprotection, with restored pro-homeostatic lipid mediators and improved neurological recovery. Altogether, our findings support the combinatory therapy as the basis for future therapeutics for ischemic stroke.

6.
Sci Rep ; 7(1): 12770, 2017 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-28986580

RESUMEN

To assess the metabolically beneficial effects of fenugreek (Trigonella foenum-graecum), C57BL/6J mice were fed a low- or high-fat diet for 16 weeks with or without 2% (w/w) fenugreek supplementation. Body weight, body composition, energy expenditure, food intake, and insulin/glucose tolerance were measured regularly, and tissues were collected for histological and biochemical analysis after 16 weeks of diet exposure. Fenugreek did not alter body weight, fat mass, or food intake in either group, but did transiently improve glucose tolerance in high fat-fed mice. Fenugreek also significantly improved high-density lipoprotein to low-density lipoprotein ratios in high fat-fed mice without affecting circulating total cholesterol, triglycerides, or glycerol levels. Fenugreek decreased hepatic expression of fatty acid-binding protein 4 and increased subcutaneous inguinal adipose tissue expression of adiponectin, but did not prevent hepatic steatosis. Notably, fenugreek was not as effective at improving glucose tolerance as was four days of voluntary wheel running. Overall, our results demonstrate that fenugreek promotes metabolic resiliency via significant and selected effects on glucose regulation, hyperlipidemia, and adipose pathology; but may not be as effective as behavioral modifications at preventing the adverse metabolic consequences of a high fat diet.


Asunto(s)
Biomarcadores/metabolismo , Dieta Alta en Grasa , Suplementos Dietéticos , Conducta Alimentaria , Salud , Metabolismo , Trigonella/química , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Adiposidad , Animales , Glucemia/metabolismo , Peso Corporal , Epidídimo/metabolismo , Ácido Graso Sintasas/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Ácidos Grasos/metabolismo , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/patología , Inflamación/patología , Insulina/sangre , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Lipoproteínas HDL/sangre , Lipoproteínas LDL/sangre , Masculino , Ratones Endogámicos C57BL , Condicionamiento Físico Animal , Triglicéridos/sangre
7.
Nat Commun ; 6: 6228, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25736573

RESUMEN

The identification of pathways necessary for photoreceptor and retinal pigment epithelium (RPE) function is critical to uncover therapies for blindness. Here we report the discovery of adiponectin receptor 1 (AdipoR1) as a regulator of these cells' functions. Docosahexaenoic acid (DHA) is avidly retained in photoreceptors, while mechanisms controlling DHA uptake and retention are unknown. Thus, we demonstrate that AdipoR1 ablation results in DHA reduction. In situ hybridization reveals photoreceptor and RPE cell AdipoR1 expression, blunted in AdipoR1(-/-) mice. We also find decreased photoreceptor-specific phosphatidylcholine containing very long-chain polyunsaturated fatty acids and severely attenuated electroretinograms. These changes precede progressive photoreceptor degeneration in AdipoR1(-/-) mice. RPE-rich eyecup cultures from AdipoR1(-/-) reveal impaired DHA uptake. AdipoR1 overexpression in RPE cells enhances DHA uptake, whereas AdipoR1 silencing has the opposite effect. These results establish AdipoR1 as a regulatory switch of DHA uptake, retention, conservation and elongation in photoreceptors and RPE, thus preserving photoreceptor cell integrity.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Células Fotorreceptoras de Vertebrados/fisiología , Receptores de Adiponectina/metabolismo , Epitelio Pigmentado de la Retina/fisiología , Animales , Electrorretinografía , Hibridación in Situ , Ratones , Ratones Noqueados , Fosfatidilcolinas/metabolismo , Células Fotorreceptoras de Vertebrados/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
8.
Mol Neurobiol ; 44(2): 216-22, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21918832

RESUMEN

The harmony and function of the complex brain circuits and synapses are sustained mainly by excitatory and inhibitory neurotransmission, neurotrophins, gene regulation, and factors, many of which are incompletely understood. A common feature of brain circuit components, such as dendrites, synaptic membranes, and other membranes of the nervous system, is that they are richly endowed in docosahexaenoic acid (DHA), the main member of the omega-3 essential fatty acid family. DHA is avidly retained and concentrated in the nervous system and known to play a role in neuroprotection, memory, and vision. Only recently has it become apparent why the surprisingly rapid increases in free (unesterified) DHA pool size take place at the onset of seizures or brain injury. This phenomenon began to be clarified by the discovery of neuroprotectin D1 (NPD1), the first-uncovered bioactive docosanoid formed from free DHA through 15-lipoxygenase-1 (15-LOX-1). NPD1 synthesis includes, as agonists, oxidative stress and neurotrophins. The evolving concept is that DHA-derived docosanoids set in motion endogenous signaling to sustain homeostatic synaptic and circuit integrity. NPD1 is anti-inflammatory, displays inflammatory resolving activities, and induces cell survival, which is in contrast to the pro-inflammatory actions of the many of omega-6 fatty acid family members. We highlight here studies relevant to the ability of DHA to sustain neuronal function and protect synapses and circuits in the context of DHA signalolipidomics. DHA signalolipidomics comprises the integration of the cellular/tissue mechanism of DHA uptake, its distribution among cellular compartments, the organization and function of membrane domains containing DHA phospholipids, and the precise cellular and molecular events revealed by the uncovering of signaling pathways regulated by docosanoids endowed with prohomeostatic and cell survival bioactivity. Therefore, this approach offers emerging targets for prevention, pharmaceutical intervention, and clinical translation involving DHA-mediated signaling.


Asunto(s)
Ácidos Docosahexaenoicos/metabolismo , Ácidos Grasos Omega-3/metabolismo , Homeostasis , Red Nerviosa/fisiología , Transducción de Señal/fisiología , Sinapsis/fisiología , Apoptosis/fisiología , Supervivencia Celular , Humanos , Neuronas/citología , Neuronas/metabolismo , Convulsiones/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA