Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Neurobiol Dis ; 159: 105483, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34400304

RESUMEN

Mitochondrial diseases are among the most prevalent groups of inherited neurological disorders, affecting up to 1 in 5000 adults. Despite the progress achieved on the identification of gene mutations causing mitochondrial pathologies, they cannot be cured so far. Harlequin mice, a relevant model of mitochondrial pathology due to apoptosis inducing factor depletion, suffer from progressive disappearance of retinal ganglion cells leading to optic neuropathy. In our previous work, we showed that administering adeno-associated virus encompassing the coding sequences for neuroglobin, (a neuroprotective molecule belonging to the globin family) or apoptosis-inducing factor, before neurodegeneration onset, prevented retinal ganglion cell loss and preserved visual function. One of the challenges to develop an effective treatment for optic neuropathies is to consider that by the time patients become aware of their handicap, a large amount of nerve fibers has already disappeared. Gene therapy was performed in Harlequin mice aged between 4 and 5 months with either a neuroglobin or an apoptosis-inducing factor vector to determine whether the increased abundance of either one of these proteins in retinas could preserve visual function at this advanced stage of the disease. We demonstrated that gene therapy, by preserving the connectivity of transduced retinal ganglion cells and optic nerve bioenergetics, results in the enhancement of visual cortex activity, ultimately rescuing visual impairment. This study demonstrates that: (a) An increased abundance of neuroglobin functionally overcomes apoptosis-inducing factor absence in Harlequin mouse retinas at a late stage of neuronal degeneration; (b) The beneficial effect for visual function could be mediated by neuroglobin localization to the mitochondria, thus contributing to the maintenance of the organelle homeostasis.


Asunto(s)
Factor Inductor de la Apoptosis/genética , Complejo IV de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Neuroglobina/genética , Atrofia Óptica/metabolismo , Nervio Óptico/metabolismo , Células Ganglionares de la Retina/metabolismo , Agudeza Visual/genética , Corteza Visual/metabolismo , Animales , Progresión de la Enfermedad , Terapia Genética , Ratones , Atrofia Óptica/patología , Atrofia Óptica/fisiopatología , Nervio Óptico/patología , Nervio Óptico/fisiopatología , Células Ganglionares de la Retina/patología , Corteza Visual/patología , Vías Visuales
2.
Biochim Biophys Acta ; 1863(9): 2322-32, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27321910

RESUMEN

E-prostanoid receptor subtype 2 (EP2) agonists are currently under clinical development as hypotensive agents for the treatment of ocular hypertension. However, the effects of EP2 receptor agonists on trabecular meshwork (TM) alterations leading to primary open-angle glaucoma (POAG) are still unknown. Here, we evaluated whether EP2 receptor activation exhibits protective functions on TM cell death induced by endoplasmic reticulum (ER) stress. We show that the EP2 receptor agonist butaprost protects TM cell death mediated by the ER stress inducer tunicamycin through a cyclic AMP (cAMP)-dependent mechanism, but independent of the classical cAMP sensors, protein kinase A and exchange proteins activated by cAMP. The ER stress-induced intrinsic apoptosis inhibited by the EP2 receptor agonist was correlated with a decreased accumulation of the cellular stress sensor p53. In addition, p53 down-regulation was associated with inhibition of its transcriptional activity, which led to decreased expression of the pro-apoptotic p53-upregulated modulator of apoptosis (PUMA). The stabilization of p53 by nutlin-3a abolished butaprost-mediated cell death protection. In conclusion, we showed that EP2 receptor activation protects against ER stress-dependent mitochondrial apoptosis through down-regulation of p53. The specific inhibition of this pathway could reduce TM alterations observed in POAG patients.


Asunto(s)
Apoptosis , Citoprotección , Regulación hacia Abajo , Estrés del Retículo Endoplásmico , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal , Malla Trabecular/patología , Proteína p53 Supresora de Tumor/metabolismo , Adulto , Alprostadil/análogos & derivados , Alprostadil/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasas/metabolismo , Muerte Celular/efectos de los fármacos , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Citocromos c/metabolismo , Citoprotección/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Choque Térmico/metabolismo , Humanos , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Proteínas Proto-Oncogénicas/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo , Tunicamicina/farmacología , Respuesta de Proteína Desplegada/efectos de los fármacos
3.
Mol Ther ; 22(6): 1096-1109, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24622090

RESUMEN

Neuroglobin (NGB) is considered as an endogenous neuroprotective molecule against stroke, since the protein alleviates the adverse effects of hypoxic and ischemic insults. We previously demonstrated the functional link between NGB and mitochondria since it is required for respiratory chain function. Thus, here, we evaluated the relevance of this effect in the Harlequin (Hq) mouse strain, which exhibits retinal ganglion cell (RGC) loss and optic atrophy due to a respiratory chain complex I (CI) defect. A twofold decrease of NGB amounts was observed in Hq retinas. We constructed a recombinant adeno-associated virus which combines to the mouse NGB open reading frame, its 5' and 3'UTR, for guarantying mRNA stability and translation capacity. The vector was administrated intravitreally to Hq mice and NGB expression was stable for up to 7 months without negative effect on retinal architecture or function. On the contrary, RGCs and their axons were substantially preserved from degeneration; consequently, CI activity in optic nerves was protected conferring improvements in vision. Hence, we established that NGB prevents respiratory chain impairment, therefore, protecting visual function otherwise compromised by mitochondrial energetic failure.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Globinas/genética , Proteínas del Tejido Nervioso/genética , Atrofia Óptica/prevención & control , Atrofia Óptica/terapia , Células Ganglionares de la Retina/metabolismo , Animales , Axones/metabolismo , Axones/patología , Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos/administración & dosificación , Gliosis/patología , Gliosis/prevención & control , Globinas/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuroglobina , Atrofia Óptica/genética , Atrofia Óptica/patología , Células Ganglionares de la Retina/patología
4.
Neuroimage ; 79: 288-94, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23660031

RESUMEN

Amyloid plaques are one of the major microscopic lesions that characterize Alzheimer's disease. Current approaches to detect amyloid plaques by using magnetic resonance imaging (MRI) contrast agents require invasive procedures to penetrate the blood-brain barrier (BBB) and to deliver the contrast agent into the vicinity of amyloid plaques. Here we have developed a new protocol (US-Gd-staining) that enables the detection of amyloid plaques in the brain of an APP/PS1 transgenic mouse model of amyloidosis after intra-venous injection of a non-targeted, clinically approved MRI contrast agent (Gd-DOTA, Dotarem®) by transiently opening the BBB with unfocused ultrasound (1 MHz) and clinically approved microbubbles (Sonovue®, Bracco). This US-Gd-staining protocol can detect amyloid plaques with a short imaging time (32 min) and high in-plane resolution (29 µm). The sensitivity and resolution obtained is at least equal to that provided by MRI protocols using intra-cerebro-ventricular injection of contrast agents, a reference method used to penetrate the BBB. To our knowledge this is the first study to demonstrate the ability of MR imaging to detect amyloid plaques by using a peripheral intra-venous injection of a clinically approved NMR contrast agent.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Compuestos Heterocíclicos/farmacocinética , Imagen por Resonancia Magnética/métodos , Compuestos Organometálicos/farmacocinética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Sonicación/métodos , Animales , Barrera Hematoencefálica/efectos de la radiación , Encéfalo/metabolismo , Encéfalo/patología , Medios de Contraste/farmacocinética , Aumento de la Imagen/métodos , Ratones , Ratones Transgénicos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Coloración y Etiquetado
5.
J Neurosci ; 27(10): 2483-92, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17344386

RESUMEN

Spinocerebellar ataxia 7 (SCA7) is a neurodegenerative disease caused by a polyglutamine (polyQ) expansion in the ataxin 7 (ATXN7) protein, a member of a multiprotein complex involved in histone acetylation. We have created a conditional Drosophila model of SCA7 in which expression of truncated ATXN7 (ATXN7T) with a pathogenic polyQ expansion is induced in neurons in adult flies. In this model, mutant ATXN7T accumulated in neuronal intranuclear inclusions containing ubiquitin, the 19S proteasome subunit, and HSP70 (heat shock protein 70), as in patients. Aggregation was accompanied by a decrease in locomotion and lifespan but limited neuronal death. Disaggregation of the inclusions, when expression of expanded ATXN7T was stopped, correlated with improved locomotor function and increased lifespan, suggesting that the pathology may respond to treatment. Lifespan was then used as a quantitative marker in a candidate gene approach to validate the interest of the model and to identify generic modulators of polyQ toxicity and specific modifiers of SCA7. Several molecular pathways identified in this focused screen (proteasome function, unfolded protein stress, caspase-dependent apoptosis, and histone acetylation) were further studied in primary neuronal cultures. Sodium butyrate, a histone deacetylase inhibitor, improved the survival time of the neurons. This model is therefore a powerful tool for studying SCA7 and for the development of potential therapies for polyQ diseases.


Asunto(s)
Modelos Animales de Enfermedad , Drosophila , Ataxias Espinocerebelosas/genética , Animales , Animales Modificados Genéticamente , Ataxina-7 , Muerte Celular , Células Cultivadas , Discinesias/genética , Glutamina , Humanos , Cuerpos de Inclusión Intranucleares/ultraestructura , Longevidad , Masculino , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Péptidos/genética , Fenotipo , Ratas , Ataxias Espinocerebelosas/metabolismo , Ataxias Espinocerebelosas/patología , Ataxias Espinocerebelosas/fisiopatología , Treonina
6.
Alzheimers Res Ther ; 10(1): 117, 2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30486882

RESUMEN

BACKGROUND: Anti-amyloid ß (Aß) immunotherapy represents a major area of drug development for Alzheimer's disease (AD). However, Aß peptide adopts multiple conformations and the pathological forms to be specifically targeted have not been identified. Aß immunotherapy-related vasogenic edema has also been severely dose limiting for antibodies with effector functions binding vascular amyloid such as bapineuzumab. These two factors might have contributed to the limited efficacy demonstrated so far in clinical studies. METHODS: To address these limitations, we have engineered SAR228810, a humanized monoclonal antibody (mAb) with limited Fc effector functions that binds specifically to soluble protofibrillar and fibrillar forms of Aß peptide and we tested it together with its murine precursor SAR255952 in vitro and in vivo. RESULTS: Unlike gantenerumab and BAN2401, SAR228810 and SAR255952 do not bind to Aß monomers, low molecular weight Aß oligomers or, in human brain sections, to Aß diffuse deposits which are not specific of AD pathology. Both antibodies prevent Aß42 oligomer neurotoxicity in primary neuronal cultures. In vivo, SAR255952, a mouse aglycosylated IgG1, dose-dependently prevented brain amyloid plaque formation and plaque-related inflammation with a minimal active dose of 3 mg/kg/week by the intraperitoneal route. No increase in plasma Aß levels was observed with SAR255952 treatment, in line with its lack of affinity for monomeric Aß. The effects of SAR255952 translated into synaptic functional improvement in ex-vivo hippocampal slices. Brain penetration and decoration of cerebral amyloid plaques was documented in live animals and postmortem. SAR255952 (up to 50 mg/kg/week intravenously) did not increase brain microhemorrhages and/or microscopic changes in meningeal and cerebral arteries in old APPSL mice while 3D6, the murine version of bapineuzumab, did. In immunotolerized mice, the clinical candidate SAR228810 demonstrated the same level of efficacy as the murine SAR255952. CONCLUSION: Based on the improved efficacy/safety profile in non-clinical models of SAR228810, a first-in-man single and multiple dose administration clinical study has been initiated in AD patients.


Asunto(s)
Enfermedad de Alzheimer/terapia , Péptidos beta-Amiloides/inmunología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Encéfalo/inmunología , Inmunoterapia/métodos , Enfermedad de Alzheimer/inmunología , Péptidos beta-Amiloides/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/efectos adversos , Encéfalo/metabolismo , Potenciales Postsinápticos Excitadores/inmunología , Femenino , Hipocampo/inmunología , Hipocampo/fisiopatología , Humanos , Inmunoterapia/efectos adversos , Masculino , Ratones Endogámicos C57BL , Imagen Óptica , Cultivo Primario de Células , Factores de Riesgo
7.
J Neurosci ; 26(5): 1343-54, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16452658

RESUMEN

Locus ceruleus (LC) degeneration and loss of cortical noradrenergic innervation occur early in Alzheimer's disease (AD). Although this has been known for several decades, the contribution of LC degeneration to AD pathogenesis remains unclear. We induced LC degeneration with N-(2-chloroethyl)-N-ethyl-bromo-benzylamine (dsp4) in amyloid precursor protein 23 (APP23) transgenic mice with a low amyloid load. Then 6 months later the LC projection areas showed a robust elevation of glial inflammation along with augmented amyloid plaque deposits. Moreover, neurodegeneration and neuronal loss significantly increased. Importantly, the paraventricular thalamus, a nonprojection area, remained unaffected. Radial arm maze and social partner recognition tests revealed increased memory deficits while high-resolution magnetic resonance imaging-guided micro-positron emission tomography demonstrated reduced cerebral glucose metabolism, disturbed neuronal integrity, and attenuated acetylcholinesterase activity. Nontransgenic mice with LC degeneration were devoid of these alterations. Our data demonstrate that the degeneration of LC affects morphology, metabolism, and function of amyloid plaque-containing higher brain regions in APP23 mice. We postulate that LC degeneration substantially contributes to AD development.


Asunto(s)
Enfermedad de Alzheimer/patología , Precursor de Proteína beta-Amiloide/genética , Locus Coeruleus/patología , Degeneración Nerviosa/patología , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/etiología , Animales , Astrocitos/metabolismo , Encéfalo/diagnóstico por imagen , Trastornos del Conocimiento/etiología , Femenino , Locus Coeruleus/metabolismo , Ratones , Ratones Transgénicos , Microglía/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Norepinefrina/metabolismo , Ácido Peroxinitroso/metabolismo , Placa Amiloide/patología , Tomografía de Emisión de Positrones , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
8.
Brain ; 129(Pt 5): 1194-200, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16481374

RESUMEN

We investigated whether there is neurogenesis in the striatum of aged monkeys, and whether dopamine (DA) depletion induces the genesis of new DA neurons in this structure. Six aged macaques received repeated intraperitoneal injections of bromodeoxyuridine (BrdU) over a 3 week period to label dividing cells. Three macaques were injected in parallel with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to decrease dopaminergic innervation of the striatum. The brains were analysed 3 weeks after the last BrdU injection. In MPTP-treated aged macaques, the number of tyrosine hydroxylase (TH) immunoreactive (ir) striatal neurons increased 2.3-fold compared with controls. These TH-ir striatal cells did not express dopamine beta hydroxylase (DBH) but the dopamine transporter (DAT), suggesting that they are functional DA neurons. They were also negative for calbindin (CB), neuropeptide Y (NPY) and parvalbumin (PV), and a small proportion expressed calretinin (CR). This suggests that these cells stained for TH are interneurons. All these cells also co-expressed glutamic acid decarboxylase (GAD). They thus resemble the small, aspiny, GABAergic interneurons. None of the BrdU-labelled cells in the striatum expressed the neuronal markers neuronal nuclei (NeuN), or GAD or TH, and none of TH-ir cells incorporated BrdU. These data indicate that neurogenesis did not occur in the striatum of aged macaques. The new striatal TH-ir neurons observed after DA depletion was therefore derived from pre-existing GABAergic interneurons. Understanding of the molecular signals mediating this phenotypic shift might help in developing novel and elegant strategies for a cell-based therapy for Parkinson's disease that would avoid many of the drawbacks of cell transplantation.


Asunto(s)
Cuerpo Estriado/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Intoxicación por MPTP/metabolismo , Regeneración Nerviosa , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Bromodesoxiuridina , División Celular , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Dopamina/deficiencia , Glutamato Descarboxilasa/metabolismo , Interneuronas/metabolismo , Interneuronas/patología , Intoxicación por MPTP/patología , Intoxicación por MPTP/fisiopatología , Macaca , Masculino , Fenotipo , Tirosina 3-Monooxigenasa/metabolismo
9.
Mol Ther Methods Clin Dev ; 5: 200-220, 2017 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-28540323

RESUMEN

Mitochondrial dysfunction is responsible for hereditary optic neuropathies. We wished to determine whether preserving mitochondrial bioenergetics could prevent optic neuropathy in a reliable model of glaucoma. DBA/2J mice exhibit elevated intraocular pressure, progressive degeneration of their retinal ganglion cells, and optic neuropathy that resembles glaucoma. We established that glaucoma in these mice is directly associated with mitochondrial dysfunction: respiratory chain activity was compromised in optic nerves 5 months before neuronal loss began, and the amounts of some mitochondrial proteins were reduced in retinas of glaucomatous mice. One of these proteins is neuroglobin, which has a neuroprotective function. Therefore, we investigated whether gene therapy aimed at restoring neuroglobin levels in the retina via ocular administration of an adeno-associated viral vector could reduce neuronal degeneration. The approach of treating 2-month-old mice impeded glaucoma development: few neurons died and respiratory chain activity and visual cortex activity were comparable to those in young, asymptomatic mice. When the treatment was performed in 8-month-old mice, the surviving neurons acquired new morphologic and functional properties, leading to the preservation of visual cortex activity and respiratory chain activity. The beneficial effects of neuroglobin in DBA/2J retinas confirm this protein to be a promising candidate for treating glaucoma.

10.
Neuropsychopharmacology ; 31(6): 1146-57, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16292321

RESUMEN

The generation of new neurons in the hippocampus is a dynamic process regulated by environmental, endocrine, and pharmacological factors. Since enhancement of hippocampal neurogenesis has been associated with learning and memory, and the locus coeruleus-noradrenergic system has been shown to modulate these cognitive functions, we hypothesized that activation of noradrenergic neurotransmission might enhance neurogenesis in the adult hippocampus. To test this hypothesis in vivo, we induced the release of noradrenaline in the hippocampus by blocking presynaptic inhibitory autoreceptors with the selective alpha2-adrenoceptor antagonist dexefaroxan. Confocal microscopy showed that noradrenergic afferents make contact with proliferating and differentiating cells, suggesting a direct noradrenergic influence on neurogenesis. Chronic systemic treatment of rats with dexefaroxan did not affect cell proliferation per se in the dentate gyrus (as monitored by bromodeoxyuridine-labeling), but promoted the long-term survival of newborn neurons by reducing apoptosis. Dexefaroxan treatment also enhanced the number and complexity of the dendritic arborizations of polysialated neural cell adhesion molecule-positive neurons. The trophic effects of dexefaroxan on newborn cells might involve an increase in brain-derived neurotrophic factor, which was upregulated in afferent noradrenergic fiber projection areas and in neurons in the granule cell layer. By promoting the survival of new endogenously formed neurons, dexefaroxan treatment represents a potential therapeutic strategy for maintaining adult neurogenesis in neurodegenerative conditions, such as Alzheimer's disease, that affect the hippocampus.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 2 , Benzopiranos/farmacología , Diferenciación Celular/efectos de los fármacos , Hipocampo/citología , Imidazoles/farmacología , Neuronas/efectos de los fármacos , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Bromodesoxiuridina , Recuento de Células/métodos , Supervivencia Celular/efectos de los fármacos , Cromatografía Líquida de Alta Presión/métodos , Inmunohistoquímica/métodos , Etiquetado Corte-Fin in Situ/métodos , Masculino , Microdiálisis/métodos , Molécula L1 de Adhesión de Célula Nerviosa/metabolismo , Norepinefrina/metabolismo , Ratas , Ratas Sprague-Dawley , Ácidos Siálicos/metabolismo , Factores de Tiempo
11.
Front Aging Neurosci ; 8: 55, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27047372

RESUMEN

Extracellular deposition of ß amyloid plaques is an early event associated to Alzheimer's disease. Here, we have used in vivo gadolinium-stained high resolution (29(∗)29(∗)117 µm(3)) magnetic resonance imaging (MRI) to follow-up in a longitudinal way individual amyloid plaques in APP/PS1 mice and evaluate the efficacy of a new immunotherapy (SAR255952) directed against protofibrillar and fibrillary forms of Aß. APP/PS1 mice were treated for 5 months between the age of 3.5 and 8.5 months. SAR255952 reduced amyloid load in 8.5-months-old animals, but not in 5.5-months animals compared to mice treated with a control antibody (DM4). Histological evaluation confirmed the reduction of amyloid load and revealed a lower density of amyloid plaques in 8.5-months SAR255952-treated animals. The longitudinal follow-up of individual amyloid plaques by MRI revealed that plaques that were visible at 5.5 months were still visible at 8.5 months in both SAR255952 and DM4-treated mice. This suggests that the amyloid load reduction induced by SAR255952 is related to a slowing down in the formation of new plaques rather than to the clearance of already formed plaques.

12.
Invest Ophthalmol Vis Sci ; 57(4): 1816-25, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27082296

RESUMEN

PURPOSE: Prostaglandin F2α analogues are the first-line medication for the treatment of ocular hypertension (OHT), and prostanoid EP2 receptor agonists are under clinical development for this indication. The goal of this study was to investigate the effects of F prostanoid (FP) and EP2 receptor activation on the myofibroblast transition of primary trabecular meshwork (TM) cells, which could be a causal mechanism of TM dysfunction in glaucoma. METHODS: Human primary TM cells were treated with either latanoprost or butaprost and TGF-ß2. Trabecular meshwork contraction was measured in a three-dimensional (3D) TM cell-populated collagen gel (CPCG) model. Expression of α-smooth muscle actin (α-SMA) and phosphorylation of myosin light chain (MLC) were determined by Western blot. Assembly of actin stress fibers and collagen deposition were evaluated by immunocytochemistry. Involvement of p38, extracellular signal-regulated kinase (ERK), and Rho-associated kinase (ROCK) pathways as well as matrix metalloproteinase activation was tested with specific inhibitors. RESULTS: In one source of validated adult TM cells, latanoprost induced cell contraction as observed by CPCG surface reduction and increased actin polymerization, α-SMA expression, and MLC phosphorylation, whereas butaprost inhibited TGF-ß2-induced CPCG contraction, actin polymerization, and MLC phosphorylation. Both agonists inhibited TGF-ß2-dependent collagen deposition. The latanoprost effects were mediated by p38 pathway. CONCLUSIONS: Latanoprost decreased TM collagen accumulation but promoted a contractile phenotype in a source of adult TM cells that could modulate the conventional outflow pathway. In contrast, butaprost attenuated both TM contraction and collagen deposition induced by TGF-ß2, thereby inhibiting myofibroblast transition of TM cells. These results open new perspectives for the management of OHT.


Asunto(s)
Glaucoma/tratamiento farmacológico , Miofibroblastos/metabolismo , Prostaglandinas F Sintéticas/farmacología , ARN/genética , Subtipo EP2 de Receptores de Prostaglandina E/efectos de los fármacos , Receptores de Prostaglandina/efectos de los fármacos , Malla Trabecular/metabolismo , Actinas/biosíntesis , Actinas/genética , Adulto , Alprostadil/análogos & derivados , Alprostadil/farmacología , Animales , Antihipertensivos/farmacología , Western Blotting , Supervivencia Celular , Células Cultivadas , Dinoprost , Glaucoma/genética , Glaucoma/metabolismo , Humanos , Inmunohistoquímica , Latanoprost , Masculino , Miofibroblastos/efectos de los fármacos , Miofibroblastos/patología , Cadenas Ligeras de Miosina/metabolismo , Fármacos Neuroprotectores , Prostaglandinas E Sintéticas , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Prostaglandina/metabolismo , Subtipo EP2 de Receptores de Prostaglandina E/metabolismo , Transducción de Señal/efectos de los fármacos , Malla Trabecular/efectos de los fármacos , Malla Trabecular/patología
13.
Sci Rep ; 6: 20958, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26876372

RESUMEN

Histology is the gold standard to unveil microscopic brain structures and pathological alterations in humans and animal models of disease. However, due to tedious manual interventions, quantification of histopathological markers is classically performed on a few tissue sections, thus restricting measurements to limited portions of the brain. Recently developed 3D microscopic imaging techniques have allowed in-depth study of neuroanatomy. However, quantitative methods are still lacking for whole-brain analysis of cellular and pathological markers. Here, we propose a ready-to-use, automated, and scalable method to thoroughly quantify histopathological markers in 3D in rodent whole brains. It relies on block-face photography, serial histology and 3D-HAPi (Three Dimensional Histology Analysis Pipeline), an open source image analysis software. We illustrate our method in studies involving mouse models of Alzheimer's disease and show that it can be broadly applied to characterize animal models of brain diseases, to evaluate therapeutic interventions, to anatomically correlate cellular and pathological markers throughout the entire brain and to validate in vivo imaging techniques.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico por imagen , Mapeo Encefálico , Encéfalo/diagnóstico por imagen , Imagenología Tridimensional , Enfermedad de Alzheimer/patología , Animales , Encéfalo/patología , Encéfalo/ultraestructura , Modelos Animales de Enfermedad , Humanos , Procesamiento de Imagen Asistido por Computador , Ratones , Programas Informáticos
14.
Brain Res ; 1066(1-2): 196-200, 2005 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-16325783

RESUMEN

Pleiotrophin (PTN), a developmentally-regulated trophic factor, is over-expressed in the striatum of parkinsonian rats. Because striatal PTN can provide trophic support to dopamine neurons, we identified the cellular types containing PTN in the striatum of adult rats. By means of fluorescent double-immunolabeling, we found PTN to co-localize with a neuronal nuclei marker but not with glial fibrillary acidic protein. The number, distribution, and morphology of the PTN-immunolabeled cells suggested that they were interneurons. Further double-immunolabeling studies ruled out PTN localization to calretinin- and parvalbumin-containing interneurons. Instead, approximately 40% of the PTN-immunolabeled neurons contained nitric oxide synthase or somatostatin and approximately 60% expressed the vesicular acetylcholine transporter, supporting that they were GABAergic nitric oxide synthase/somatostatin-containing and cholinergic interneurons. Further work is necessary to determine if PTN from striatal interneurons can provide trophic support to dopamine neurons.


Asunto(s)
Proteínas Portadoras/metabolismo , Citocinas/metabolismo , Interneuronas/metabolismo , Neostriado/metabolismo , Animales , Western Blotting , Calbindina 2 , Femenino , Técnica del Anticuerpo Fluorescente , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inmunoquímica , Inmunohistoquímica , Peso Molecular , Neostriado/citología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Parvalbúminas/metabolismo , Ratas , Ratas Wistar , Proteína G de Unión al Calcio S100/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
15.
PLoS One ; 10(6): e0129518, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26107939

RESUMEN

Myopia is a common ocular disorder generally due to increased axial length of the eye-globe. Its extreme form high myopia (HM) is a multifactorial disease leading to retinal and scleral damage, visual impairment or loss and is an important health issue. Mutations in the endocytic receptor LRP2 gene result in Donnai-Barrow (DBS) and Stickler syndromes, both characterized by HM. To clearly establish the link between Lrp2 and congenital HM we inactivated Lrp2 in the mouse forebrain including the neural retina and the retinal and ciliary pigment epithelia. High resolution in vivo MRI imaging and ophthalmological analyses showed that the adult Lrp2-deficient eyes were 40% longer than the control ones mainly due to an excessive elongation of the vitreal chamber. They had an apparently normal intraocular pressure and developed chorioretinal atrophy and posterior scleral staphyloma features reminiscent of human myopic retinopathy. Immunomorphological and ultrastructural analyses showed that increased eye lengthening was first observed by post-natal day 5 (P5) and that it was accompanied by a rapid decrease of the bipolar, photoreceptor and retinal ganglion cells, and eventually the optic nerve axons. It was followed by scleral thinning and collagen fiber disorganization, essentially in the posterior pole. We conclude that the function of LRP2 in the ocular tissues is necessary for normal eye growth and that the Lrp2-deficient eyes provide a unique tool to further study human HM.


Asunto(s)
Cuerpo Ciliar/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Miopía Degenerativa/genética , Proteínas del Tejido Nervioso/metabolismo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Animales , Axones/metabolismo , Proliferación Celular , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Genotipo , Presión Intraocular , Proteína 2 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Imagen por Resonancia Magnética , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Mutación , Proteínas del Tejido Nervioso/genética , Nervio Óptico/metabolismo , Fenotipo , Prosencéfalo/metabolismo , Retina/embriología , Células Ganglionares de la Retina/metabolismo , Epitelio Pigmentado de la Retina/embriología , Esclerótica/patología
16.
Brain Res Dev Brain Res ; 145(1): 9-17, 2003 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-14519489

RESUMEN

In order to understand the relationship between astrocytes, microglia and injured neurons, we studied the weaver mutant mouse. One of the main characteristics of this mutant is the progressive degeneration of the dopaminergic (DA) nigrostriatal pathway that starts around postnatal day 15 (P15), in the substantia nigra pars compacta (SNpc) and progresses until adult age (P60). In the present paper, we analysed the relationship between astroglial and microglial cells within DA neurons in the nigrostriatal system of homozygous weaver mice, at different postnatal ages corresponding to specific stages of the DA neuronal loss. The activation of astrocytes was found to be an early event in weaver DA denervation, appearing massively at the onset of DA neuronal loss in the SNpc at P15. Astrocytes remained activated in the adult brain even after the slowing down of the neuronal death process. Interestingly, in the ventral tegmental area, where no DA neuronal death could be detected, a profound, permanent astrogliosis was also observed in adult animals. In contrast, an activation of microglial cells was transiently observed in the SNpc but only at the postnatal age when maximal neuronal death was observed (P30). Lastly, in the striatum, where there was a massive loss of DA nerve terminals, neither astrogliosis nor microglial activation was detected. Hence, the reaction of astrocytes and microglial cells to progressive and spontaneous DA neuronal death showed different temporal kinetics, suggesting a different role for these two cell types in the DA neurodegenerative process in the weaver mouse.


Asunto(s)
Astrocitos/fisiología , Muerte Celular/fisiología , Dopamina/metabolismo , Microglía/fisiología , Neuronas/fisiología , Envejecimiento , Animales , Animales Recién Nacidos , Recuento de Células , Proteína Ácida Fibrilar de la Glía/metabolismo , Homocigoto , Inmunohistoquímica , Antígeno de Macrófago-1/metabolismo , Mesencéfalo/anatomía & histología , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Ratones , Ratones Mutantes Neurológicos , Tirosina 3-Monooxigenasa/metabolismo
17.
Neurobiol Aging ; 33(8): 1533-44, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21531045

RESUMEN

Detection of amyloid plaques in the brain by in vivo neuroimaging is a very promising biomarker approach for early diagnosis of Alzheimer's disease (AD) and evaluation of therapeutic efficacy. Here we describe a new method to detect amyloid plaques by in vivo magnetic resonance imaging (MRI) based on the intracerebroventricular injection of a nontargeted gadolinium (Gd)-based contrast agent, which rapidly diffuses throughout the brain and increases the signal and contrast of magnetic resonance (MR) images by shortening the T1 relaxation time. This gain in image sensitivity after in vitro and in vivo Gd staining significantly improves the detection and resolution of individual amyloid plaques in the cortex and hippocampus of AD transgenic mice. The improved image resolution is sensitive enough to demonstrate an age-dependent increase of amyloid plaque load and a good correlation between the amyloid load measured by µMRI and histology. These results provide the first demonstration that nontargeted Gd staining can enhance the detection of amyloid plaques to follow the progression of AD and to evaluate the activity of amyloid-lowering therapeutic strategies in longitudinal studies.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Encéfalo/patología , Gadolinio , Imagen por Resonancia Magnética/métodos , Placa Amiloide/metabolismo , Animales , Medios de Contraste , Femenino , Gadolinio/farmacocinética , Ratones , Ratones Transgénicos , Imagen Molecular/métodos , Coloración y Etiquetado/métodos , Distribución Tisular
18.
Neurobiol Aging ; 33(9): 1995-2005, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22079157

RESUMEN

Alzheimer's disease (AD), the most common age-related neurodegenerative disorder, is characterized by the invariant cerebral accumulation of ß-amyloid peptide. This event occurs early in the disease process. In humans, [18F]-fluoro-2-deoxy-D-glucose ([18F]-FDG) positron emission tomography (PET) is largely used to follow-up in vivo cerebral glucose utilization (CGU) and brain metabolism modifications associated with the Alzheimer's disease pathology. Here, [18F]-FDG positron emission tomography was used to study age-related changes of cerebral glucose utilization under resting conditions in 3-, 6-, and 12-month-old APP(SweLon)/PS1(M146L), a mouse model of amyloidosis. We showed an age-dependent increase of glucose uptake in several brain regions of APP/PS1 mice but not in control animals and a higher [18F]-FDG uptake in the cortex and the hippocampus of 12-month-old APP/PS1 mice as compared with age-matched control mice. We then developed a method of 3-D microscopic autoradiography to evaluate glucose uptake at the level of amyloid plaques and showed an increased glucose uptake close to the plaques rather than in amyloid-free cerebral tissues. These data suggest a macroscopic and microscopic reorganization of glucose uptake in relation to cerebral amyloidosis.


Asunto(s)
Enfermedad de Alzheimer/patología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Circulación Cerebrovascular/fisiología , Glucosa/metabolismo , Enfermedad de Alzheimer/diagnóstico por imagen , Enfermedad de Alzheimer/genética , Precursor de Proteína beta-Amiloide/genética , Análisis de Varianza , Animales , Proteínas de Unión al Calcio/metabolismo , Corteza Cerebral/diagnóstico por imagen , Circulación Cerebrovascular/genética , Modelos Animales de Enfermedad , Fluorodesoxiglucosa F18 , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/metabolismo , Tomografía de Emisión de Positrones , Presenilina-1/genética
19.
Exp Neurol ; 204(2): 733-40, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17306796

RESUMEN

Sepsis is a disease with a high and growing prevalence worldwide. Most studies on sepsis up to date have been focused on reduction of short-term mortality. This study investigates cognitive and neuroanatomical long-term consequences of sepsis in a rat model. Sepsis was induced in male Wistar rats weighing 250-300 g by an i.p. injection of bacterial lipopolysaccharide (LPS, 10 mg/kg). Three months after complete recovery from sepsis, animals showed memory deficits in the radial maze and changes in open field exploratory patterns but unaffected inhibitory avoidance learning. Behavioral findings were matched by sepsis-induced loss of neurons in the hippocampus and the prefrontal cortex on serial sections after NeuN-staining and reduced cholinergic innervation in the parietal cortex measured by immunoradiography of vesicular acetylcholine transporter (VAChT). Together these results suggest that sepsis can induce persistent behavioral and neuroanatomical changes and warrant studies of the neurological long-term consequences of sepsis in humans.


Asunto(s)
Corteza Cerebral/patología , Trastornos del Conocimiento/etiología , Neuronas/patología , Recuperación de la Función/fisiología , Sepsis/patología , Proteínas de Transporte Vesicular/metabolismo , Animales , Modelos Animales de Enfermedad , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Neuronas/efectos de los fármacos , Fosfopiruvato Hidratasa/metabolismo , Polisacáridos , Ratas , Ratas Wistar , Sepsis/inducido químicamente , Sepsis/complicaciones , Factores de Tiempo
20.
J Neurochem ; 102(2): 434-40, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17394553

RESUMEN

One of the few currently approved therapies for Alzheimer's disease (AD) consists in the administration of acetylcholinesterase inhibitors, which enhances the lifetime of the neurotransmitter acetylcholine. Despite numerous studies on the symptomatic effect of acetylcholinesterase inhibitors, there is as yet no direct morphological evidence to indicate that they have a neurorestorative action. We investigated the effect of the acetylcholinesterase inhibitor donepezil administered subcutaneously in a rat model of partial unilateral cortical devascularization that induces a loss of the cortical cholinergic terminal network and a retrograde degeneration of the cholinergic projections that originate in the nucleus basalis. For 6 weeks, lesioned and sham-operated rats received a subcutaneous infusion of donepezil (2 mg/kg/day) or vehicle, delivered by osmotic minipumps implanted 2 weeks before the cortical devascularization. In lesioned rats, donepezil treatment increased the number and the size of vesicular acetylcholine transporter immunoreactive boutons in comparison to vehicle treatment. Donepezil had no observable effect on any of these parameters in sham-operated animals. These results show that donepezil mitigates cholinergic neuronal degeneration in vivo. This suggests a neuroplastic activity of this drug and provides evidence for a potential use of donepezil as a disease modifier in neurodegenerative diseases such as AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Núcleo Basal de Meynert/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Fibras Colinérgicas/efectos de los fármacos , Indanos/farmacología , Piperidinas/farmacología , Acetilcolina/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/fisiopatología , Animales , Núcleo Basal de Meynert/metabolismo , Núcleo Basal de Meynert/fisiopatología , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiopatología , Fibras Colinérgicas/metabolismo , Inhibidores de la Colinesterasa/farmacología , Inhibidores de la Colinesterasa/uso terapéutico , Modelos Animales de Enfermedad , Donepezilo , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/metabolismo , Indanos/uso terapéutico , Masculino , Degeneración Nerviosa/tratamiento farmacológico , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Vías Nerviosas/efectos de los fármacos , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Piperidinas/uso terapéutico , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/metabolismo , Terminales Presinápticos/patología , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/fisiopatología , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA