Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Glia ; 72(3): 546-567, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37987116

RESUMEN

Although brain scars in adults have been extensively studied, there is less data available regarding scar formation during the neonatal period, and the involvement of peripheral immune cells in this process remains unexplored in neonates. Using a murine model of neonatal hypoxic-ischemic encephalopathy (HIE) and confocal microscopy, we characterized the scarring process and examined the recruitment of peripheral immune cells to cortical and hippocampal scars for up to 1 year post-insult. Regional differences in scar formation were observed, including the presence of reticular fibrotic networks in the cortex and perivascular fibrosis in the hippocampus. We identified chemokines with chronically elevated levels in both regions and demonstrated, through a parabiosis-based strategy, the recruitment of lymphocytes, neutrophils, and monocyte-derived macrophages to the scars several weeks after the neonatal insult. After 1 year, however, neutrophils and lymphocytes were absent from the scars. Our data indicate that peripheral immune cells are transient components of HIE-induced brain scars, opening up new possibilities for late therapeutic interventions.


Asunto(s)
Cicatriz , Hipoxia-Isquemia Encefálica , Adulto , Animales , Humanos , Ratones , Cicatriz/patología , Encéfalo/patología , Macrófagos , Hipoxia-Isquemia Encefálica/patología
2.
Stroke ; 53(2): 586-594, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34794335

RESUMEN

BACKGROUND AND PURPOSE: Despite the advances in treating neonatal hypoxic-ischemic encephalopathy (HIE) with induced hypothermia, the rates of severe disability are still high among survivors. Preclinical studies have indicated that cell therapies with hematopoietic stem/progenitor cells could improve neurological outcomes in HIE. In this study, we investigated whether the administration of AMD3100, a CXCR4 antagonist that mobilizes hematopoietic stem/progenitor cells into the circulation, has therapeutic effects in HIE. METHODS: P10 Wistar rats of both sexes were subjected to right common carotid artery occlusion or sham procedure, and then were exposed to hypoxia for 120 minutes. Two subcutaneous injections of AMD3100 or vehicle were given on the third and fourth day after HIE. We first assessed the interindividual variability in brain atrophy after experimental HIE and vehicle treatment in a small cohort of rats. Based on this exploratory analysis, we designed and conducted an experiment to test the efficacy of AMD3100. Brain atrophy on day 21 after HIE was defined as the primary end point. Secondary efficacy end points were cognitive (T-water maze) and motor function (rotarod) on days 17 and 18 after HIE, respectively. RESULTS: AMD3100 did not decrease the brain atrophy in animals of either sex. Cognitive impairments were not observed in the T-water maze, but male hypoxic-ischemic animals exhibited motor coordination deficits on the rotarod, which were not improved by AMD3100. A separate analysis combining data from animals of both sexes also revealed no evidence of the effectiveness of AMD3100 treatment. CONCLUSIONS: These results indicate that the subacute treatment with AMD3100 does not improve structural and functional outcomes in a rat HIE model.


Asunto(s)
Bencilaminas/uso terapéutico , Ciclamas/uso terapéutico , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Receptores CXCR4/antagonistas & inhibidores , Animales , Animales Recién Nacidos , Atrofia , Bencilaminas/administración & dosificación , Encéfalo/patología , Disfunción Cognitiva/psicología , Ciclamas/administración & dosificación , Determinación de Punto Final , Femenino , Masculino , Aprendizaje por Laberinto , Embarazo , Desempeño Psicomotor/efectos de los fármacos , Ratas , Ratas Wistar , Caracteres Sexuales , Insuficiencia del Tratamiento
3.
Stroke ; 52(5): 1788-1797, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33827248

RESUMEN

Background and Purpose: Heme is a red blood cell component released in the brain parenchyma following intracerebral hemorrhage. However, the study of the pathophysiological mechanisms triggered by heme in the brain is hampered by the lack of well-established in vivo models of intracerebral heme injection. This study aims to optimize and characterize a protocol of intrastriatal heme injection in mice, with a focus on the induction of lipid peroxidation, neuroinflammation and, ultimately, sensorimotor deficits. We also evaluated the involvement of NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3), an inflammasome sensor, in the behavior deficits induced by heme in this model. Methods: Mice were injected with heme in the striatum for the evaluation of neuroinflammation and brain damage through histological and biochemical techniques. Immunoblot was used to evaluate the expression of proteins involved in heme/iron metabolism and antioxidant responses and the activation of the MAPK (mitogen-activated protein kinase) signaling pathway. For the assessment of neurological function, we followed-up heme-injected mice for 2 weeks using the rotarod, elevated body swing, and cylinder tests. Mice injected with the vehicle (sham), or autologous blood were used as controls. Results: Heme induced lipid peroxidation and inflammation in the brain. Moreover, heme increased the expression of HO-1 (heme oxygenase-1), ferritin, p62, and superoxide dismutase 2, and activated the MAPK signaling pathway promoting pro-IL (interleukin)-1ß production and its cleavage to the active form. Heme-injected mice exhibited signs of brain damage and reactive astrogliosis around the injection site. Behavior deficits were observed after heme or autologous blood injection in comparison to sham-operated controls. In addition, behavior deficits and IL-1ß production were reduced in Nlrp3 knockout mice in comparison to wild-type mice. Conclusions: Our results show that intracerebral heme injection induces neuroinflammation, and neurological deficits, in an NLRP3-dependent manner, suggesting that this is a feasible model to evaluate the role of heme in neurological disorders.


Asunto(s)
Conducta Animal/efectos de los fármacos , Cuerpo Estriado/efectos de los fármacos , Hemo/administración & dosificación , Peroxidación de Lípido/efectos de los fármacos , Enfermedades Neuroinflamatorias/metabolismo , Animales , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedades Neuroinflamatorias/patología
4.
J Neurochem ; 158(3): 694-709, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34081777

RESUMEN

Gangliosides are glycosphingolipids abundantly expressed in the vertebrate nervous system, and are classified into a-, b-, or c-series according to the number of sialic acid residues. The enzyme GD3 synthase converts GM3 (an a-series ganglioside) into GD3, a b-series ganglioside highly expressed in the developing and adult retina. The present study evaluated the visual system of GD3 synthase knockout mice (GD3s-/- ), morphologically and functionally. The absence of b- series gangliosides in the retinas of knockout animals was confirmed by mass spectrometry imaging, which also indicated an accumulation of a-series gangliosides, such as GM3. Retinal ganglion cell (RGC) density was significantly reduced in GD3s-/- mice, with a similar reduction in the number of axons in the optic nerve. Knockout animals also showed a 15% reduction in the number of photoreceptor nuclei, but no difference in the bipolar cells. The area occupied by GFAP-positive glial cells was smaller in GD3s-/- retinas, but the number of microglial cells/macrophages did not change. In addition to the morphological alterations, a 30% reduction in light responsiveness was detected through quantification of pS6-expressing RGC, an indicator of neural activity. Furthermore, electroretinography (ERG) indicated a significant reduction in RGC and photoreceptor electrical activity in GD3s-/- mice, as indicated by scotopic ERG and pattern ERG (PERG) amplitudes. Finally, evaluation of the optomotor response demonstrated that GD3s-/- mice have reduced visual acuity and contrast sensitivity. These results suggest that b-series gangliosides play a critical role in regulating the structure and function of the mouse visual system.


Asunto(s)
Sensibilidad de Contraste/fisiología , Eliminación de Gen , Retina/enzimología , Sialiltransferasas/deficiencia , Sialiltransferasas/genética , Agudeza Visual/fisiología , Animales , Electrorretinografía/métodos , Femenino , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Estimulación Luminosa/métodos
5.
Int J Mol Sci ; 22(14)2021 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-34299066

RESUMEN

Stem cell therapy is a promising alternative approach to heart diseases. The most prevalent source of multipotent stem cells, usually called somatic or adult stem cells (mesenchymal stromal/stem cells, MSCs) used in clinical trials is bone marrow (BM-MSCs), adipose tissue (AT-MSCs), umbilical cord (UC-MSCs) and placenta. Therapeutic use of MSCs in cardiovascular diseases is based on the benefits in reducing cardiac fibrosis and inflammation that compose the cardiac remodeling responsible for the maintenance of normal function, something which may end up causing progressive and irreversible dysfunction. Many factors lead to cardiac fibrosis and failure, and an effective therapy is lacking to reverse or attenuate this condition. Different approaches have been shown to be promising in surpassing the poor survival of transplanted cells in cardiac tissue to provide cardioprotection and prevent cardiac remodeling. This review includes the description of pre-clinical and clinical investigation of the therapeutic potential of MSCs in improving ventricular dysfunction consequent to diverse cardiac diseases.


Asunto(s)
Fibrosis/terapia , Cardiopatías/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/citología , Animales , Humanos
6.
Gene Ther ; 27(1-2): 27-39, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31243393

RESUMEN

After an injury, axons in the central nervous system do not regenerate over large distances and permanently lose their connections to the brain. Two promising approaches to correct this condition are cell and gene therapies. In the present work, we evaluated the neuroprotective and neuroregenerative potential of pigment epithelium-derived factor (PEDF) gene therapy alone and combined with human mesenchymal stem cell (hMSC) therapy after optic nerve injury by analysis of retinal ganglion cell survival and axonal outgrowth. Overexpression of PEDF by intravitreal delivery of AAV2 vector significantly increased Tuj1-positive cells survival and modulated FGF-2, IL-1ß, Iba-1, and GFAP immunostaining in the ganglion cell layer (GCL) at 4 weeks after optic nerve crush, although it could not promote axonal outgrowth. The combination of AAV2.PEDF and hMSC therapy showed a higher number of Tuj1-positive cells and a pronounced axonal outgrowth than unimodal therapy after optic nerve crush. In summary, our results highlight a synergistic effect of combined gene and cell therapy relevant for future therapeutic interventions regarding optic nerve injury.


Asunto(s)
Proteínas del Ojo/farmacología , Factores de Crecimiento Nervioso/farmacología , Traumatismos del Nervio Óptico/terapia , Células Ganglionares de la Retina/efectos de los fármacos , Serpinas/farmacología , Animales , Axones/fisiología , Línea Celular Tumoral , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Modelos Animales de Enfermedad , Proteínas del Ojo/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/metabolismo , Compresión Nerviosa , Factores de Crecimiento Nervioso/metabolismo , Regeneración Nerviosa , Neuroprotección , Nervio Óptico , Ratas Wistar , Retina , Células Ganglionares de la Retina/metabolismo , Serpinas/metabolismo
7.
Exp Eye Res ; 200: 108212, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32910940

RESUMEN

PURPOSE: Cellular therapy with mesenchymal stem cells (MSC) is emerging as an effective option to treat optic neuropathies. In models of retinal degeneration, MSC injected in the vitreous body protects injured retinal ganglion cells and stimulate their regeneration, however the mechanism is still unknown. Considering the immunomodulating proprieties of MSC and the controversial role of microglial contribution on retinal regeneration, we developed an in vitro co-culture model to analyze the effect of MSC on retinal microglia population. METHODS: We used whole adult rat retinal explants in co-culture with human Wharton's jelly mesenchymal stem cells (hMSC) separated by a transwell membrane and analyzed hMSC effect on both retinal ganglion cells (RGCs) and retinal microglia. RESULTS: hMSC in co-culture protected RGCs after 3 days in vitro by paracrine signaling. In addition, hMSC reduced microglia population and inhibited the pro-inflammatory phenotype of the remaining microglia. CONCLUSIONS: Using a co-culture model, we demonstrated the paracrine effect of hMSC on RGC survival after injury concomitant with a reduction of microglial population. Paracrine signaling of hMSC also changed microglia phenotype and the expression of antiinflammatory factors in the retina. Our results are consistent with a detrimental effect of microglia on RGC survival and regeneration after injury.


Asunto(s)
Células Madre Mesenquimatosas/citología , Microglía/patología , Regeneración Nerviosa , Comunicación Paracrina/fisiología , Degeneración Retiniana/diagnóstico , Células Ganglionares de la Retina/patología , Animales , Supervivencia Celular , Tratamiento Basado en Trasplante de Células y Tejidos , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Masculino , Microglía/metabolismo , Fenotipo , Ratas , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo
8.
J Biol Chem ; 293(6): 1957-1975, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29284679

RESUMEN

Alzheimer's disease (AD) is a disabling and highly prevalent neurodegenerative condition, for which there are no effective therapies. Soluble oligomers of the amyloid-ß peptide (AßOs) are thought to be proximal neurotoxins involved in early neuronal oxidative stress and synapse damage, ultimately leading to neurodegeneration and memory impairment in AD. The aim of the current study was to evaluate the neuroprotective potential of mesenchymal stem cells (MSCs) against the deleterious impact of AßOs on hippocampal neurons. To this end, we established transwell cocultures of rat hippocampal neurons and MSCs. We show that MSCs and MSC-derived extracellular vesicles protect neurons against AßO-induced oxidative stress and synapse damage, revealed by loss of pre- and postsynaptic markers. Protection by MSCs entails three complementary mechanisms: 1) internalization and degradation of AßOs; 2) release of extracellular vesicles containing active catalase; and 3) selective secretion of interleukin-6, interleukin-10, and vascular endothelial growth factor to the medium. Results support the notion that MSCs may represent a promising alternative for cell-based therapies in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Vesículas Extracelulares/metabolismo , Hipocampo/citología , Células Madre Mesenquimatosas/citología , Neuronas/metabolismo , Estrés Oxidativo , Sinapsis/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/química , Animales , Células Cultivadas , Técnicas de Cocultivo , Vesículas Extracelulares/genética , Hipocampo/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Neuronas/citología , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/metabolismo
9.
J Neuroinflammation ; 16(1): 218, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31727149

RESUMEN

BACKGROUND: The development of new therapeutic strategies to treat amyotrophic lateral sclerosis (ALS) is of utmost importance. The use of cyclic nitroxides such as tempol may provide neuroprotection and improve lifespan. We investigated whether tempol (50 mg/kg) presents therapeutic potential in SOD1G93A transgenic mice. METHODS: Tempol treatment began at the asymptomatic phase of the disease (10th week) and was administered every other day until week 14, after which it was administered twice a week until the final stage of the disease. The animals were sacrificed at week 14 (initial stage of symptoms-ISS) and at the end stage (ES) of the disease. The lumbar spinal cord of the animals was dissected and processed for use in the following techniques: Nissl staining to evaluate neuronal survival; immunohistochemistry to evaluate astrogliosis and microgliosis (ISS and ES); qRT-PCR to evaluate the expression of neurotrophic factors and pro-inflammatory cytokines (ISS); and transmission electron microscopy to evaluate the alpha-motoneurons (ES). Behavioral analyses considering the survival of animals, bodyweight loss, and Rotarod motor performance test started on week 10 and were performed every 3 days until the end-stage of the disease. RESULTS: The results revealed that treatment with tempol promoted greater neuronal survival (23%) at ISS compared to untreated animals, which was maintained until ES. The intense reactivity of astrocytes and microglia observed in vehicle animals was reduced in the lumbar spinal cords of the animals treated with tempol. In addition, the groups treated with tempol showed reduced expression of proinflammatory cytokines (IL1ß and TNFα) and a three-fold decrease in the expression of TGFß1 at ISS compared with the group treated with vehicle. CONCLUSIONS: Altogether, our results indicate that treatment with tempol has beneficial effects, delaying the onset of the disease by enhancing neuronal survival and decreasing glial cell reactivity during ALS progression in SOD1G93A mice.


Asunto(s)
Esclerosis Amiotrófica Lateral/fisiopatología , Óxidos N-Cíclicos/uso terapéutico , Inflamación/tratamiento farmacológico , Destreza Motora/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Médula Espinal/efectos de los fármacos , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Supervivencia Celular/efectos de los fármacos , Óxidos N-Cíclicos/farmacología , Modelos Animales de Enfermedad , Femenino , Inflamación/metabolismo , Inflamación/patología , Interleucina-1beta/metabolismo , Masculino , Ratones , Neuronas Motoras/efectos de los fármacos , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Destreza Motora/fisiología , Fármacos Neuroprotectores/farmacología , Prueba de Desempeño de Rotación con Aceleración Constante , Marcadores de Spin , Médula Espinal/metabolismo , Médula Espinal/patología , Superóxido Dismutasa-1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
10.
Mol Biol Rep ; 46(1): 191-198, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30421128

RESUMEN

Cell transplantation offers a promising approach in many neurological disorders. Neural stem (NS) cells are potential candidates for cell therapy. The ability to track the grafted cells in the host tissue will refine this therapy. Superparamagnetic iron oxide nanoparticles (SPION) have been suggested as a feasible method, but there is no consensus about its safety. Here we investigated the feasibility of label NS cells with SPION and track by MRI after transplantation into mouse striatum with SPION cells and its therapeutic effects by grafting the cells into mouse striatum. We demonstrated that SPION-labeled NS cells display normal patterns of cellular processes including proliferation, migration, differentiation and neurosphere formation. Transmission electron microscopy reveals SPION in the cytoplasm of the cells, which was confirmed by microanalysis. Neurons and astrocytes generated from SPION-labeled NS cells were able to carry nanoparticles after 7 days under differentiation. SPION-labeled NS cells transplanted into striatum of mice were detected by magnetic resonance imaging (MRI) and microscopy 51 days later. In agreement with others reports, we demonstrated that NS cells are able to incorporate SPION in vitro without altering the stemness, and can survive and be tracked by MRI after they have been grafted into mice striatum.


Asunto(s)
Rastreo Celular/métodos , Nanopartículas de Magnetita/química , Células-Madre Neurales/fisiología , Animales , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Compuestos Férricos/metabolismo , Hierro/metabolismo , Imagen por Resonancia Magnética/métodos , Ratones , Microscopía Electrónica de Transmisión/métodos , Células-Madre Neurales/citología , Neuronas/fisiología
11.
Mol Biol Rep ; 45(6): 1827-1837, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30145640

RESUMEN

Despite the progress on development of new culture media, in vitro-produced embryos still display lower quality when compared to the in vivo-produced counterparts. Coculture has been reconsidered as an alternative to improve embryo quality. Mesenchymal stem cells (MSC) and murine embryonic fibroblasts (MEF) have been extensively used as feeder layers due to their capacity to release growth factors. In the present study we investigated the effect of these feeder layers in oocyte maturation and/or embryo development under in vitro conditions. Oocytes were matured in control (CTRL) conditions or in coculture with MSC or MEF. In vitro fertilization and embryo culture until fourth day were performed in CTRL condition for all groups. Embryos from fourth day on were then cultured until the eighth day in CTRL or in coculture system. No significant differences for metaphase II stage and apoptosis in oocytes were found among the groups. There was also no difference among the groups when we evaluated blastocyst formation on the seventh and eighth day, with exception of a higher hatched blastocyst rate in the group maturated and cultivated in CTRL condition when compared to the group matured and cocultured with MSC. Also no difference was observed in the number of cells in the whole embryos, in the inner cell mass, in the trophoblast and at apoptotic stage on the eighth day. We conclude that coculture with MSC or MEF during maturation and/or embryo development do not enhance the in vitro production of bovine embryos.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Desarrollo Embrionario/fisiología , Técnicas de Maduración In Vitro de los Oocitos/métodos , Animales , Blastocisto/citología , Blastocisto/efectos de los fármacos , Bovinos/embriología , Técnicas de Cocultivo , Embrión de Mamíferos/fisiología , Femenino , Fertilización In Vitro/métodos , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones/embriología , Oocitos/citología , Oocitos/efectos de los fármacos
12.
Crit Care ; 22(1): 249, 2018 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-30290827

RESUMEN

BACKGROUND: Ischemic stroke causes brain inflammation, which we postulate may result in lung damage. Several studies have focused on stroke-induced immunosuppression and lung infection; however, the possibility that strokes may trigger lung inflammation has been overlooked. We hypothesized that even focal ischemic stroke might induce acute systemic and pulmonary inflammation, thus altering respiratory parameters, lung tissue integrity, and alveolar macrophage behavior. METHODS: Forty-eight Wistar rats were randomly assigned to ischemic stroke (Stroke) or sham surgery (Sham). Lung function, histology, and inflammation in the lung, brain, bronchoalveolar lavage fluid (BALF), and circulating plasma were evaluated at 24 h. In vitro, alveolar macrophages from naïve rats (unstimulated) were exposed to serum or BALF from Sham or Stroke animals to elucidate possible mechanisms underlying alterations in alveolar macrophage phagocytic capability. Alveolar macrophages and epithelial and endothelial cells of Sham and Stroke animals were also isolated for evaluation of mRNA expression of interleukin (IL)-6 and tumor necrosis factor (TNF)-α. RESULTS: Twenty-four hours following ischemic stroke, the tidal volume, expiratory time, and mean inspiratory flow were increased. Compared to Sham animals, the respiratory rate and duty cycle during spontaneous breathing were reduced, but this did not affect lung mechanics during mechanical ventilation. Lungs from Stroke animals showed clear evidence of increased diffuse alveolar damage, pulmonary edema, and inflammation markers. This was associated with an increase in ultrastructural damage, as evidenced by injury to type 2 pneumocytes and endothelial cells, cellular infiltration, and enlarged basement membrane thickness. Protein levels of proinflammatory mediators were documented in the lung, brain, and plasma (TNF-α and IL-6) and in BALF (TNF-α). The phagocytic ability of macrophages was significantly reduced. Unstimulated macrophages isolated from naïve rats only upregulated expression of TNF-α and IL-6 following exposure to serum from Stroke rats. Exposure to BALF from Stroke or Sham animals did not change alveolar macrophage behavior, or gene expression of TNF-α and IL-6. IL-6 expression was increased in macrophages and endothelial cells from Stroke animals. CONCLUSIONS: In rats, focal ischemic stroke is associated with brain-lung crosstalk, leading to increased pulmonary damage and inflammation, as well as reduced alveolar macrophage phagocytic capability, which seems to be promoted by systemic inflammation.


Asunto(s)
Lesión Pulmonar/etiología , Macrófagos Alveolares/patología , Fagocitos/patología , Accidente Cerebrovascular/complicaciones , Animales , Isquemia Encefálica/complicaciones , Isquemia Encefálica/fisiopatología , Modelos Animales de Enfermedad , Terapia de Inmunosupresión/efectos adversos , Interleucina-6/análisis , Interleucina-6/sangre , Lesión Pulmonar/sangre , Lesión Pulmonar/patología , Imagen por Resonancia Magnética/métodos , Imagen por Resonancia Magnética/veterinaria , ARN Mensajero/análisis , ARN Mensajero/sangre , Ratas , Ratas Wistar/inmunología , Ratas Wistar/metabolismo , Estadísticas no Paramétricas , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/fisiopatología , Factor de Necrosis Tumoral alfa/análisis , Factor de Necrosis Tumoral alfa/sangre
13.
Int J Mol Sci ; 18(1)2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28067793

RESUMEN

Nerve growth factor (NGF) is suggested to be neuroprotective after nerve injury; however, retinal ganglion cells (RGC) degenerate following optic-nerve crush (ONC), even in the presence of increased levels of endogenous NGF. To further investigate this apparently paradoxical condition, a time-course study was performed to evaluate the effects of unilateral ONC on NGF expression and signaling in the adult retina. Visually evoked potential and immunofluorescence staining were used to assess axonal damage and RGC loss. The levels of NGF, proNGF, p75NTR, TrkA and GFAP and the activation of several intracellular pathways were analyzed at 1, 3, 7 and 14 days after crush (dac) by ELISA/Western Blot and PathScan intracellular signaling array. The progressive RGC loss and nerve impairment featured an early and sustained activation of apoptotic pathways; and GFAP and p75NTR enhancement. In contrast, ONC-induced reduction of TrkA, and increased proNGF were observed only at 7 and 14 dac. We propose that proNGF and p75NTR contribute to exacerbate retinal degeneration by further stimulating apoptosis during the second week after injury, and thus hamper the neuroprotective effect of the endogenous NGF. These findings might aid in identifying effective treatment windows for NGF-based strategies to counteract retinal and/or optic-nerve degeneration.


Asunto(s)
Factor de Crecimiento Nervioso/metabolismo , Traumatismos del Nervio Óptico/complicaciones , Degeneración Retiniana/metabolismo , Células Ganglionares de la Retina/metabolismo , Transducción de Señal , Animales , Apoptosis , Western Blotting , Potenciales Evocados Visuales/fisiología , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Microscopía Fluorescente , Compresión Nerviosa , Factores de Crecimiento Nervioso/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Precursores de Proteínas/metabolismo , Ratas , Ratas Long-Evans , Receptor trkA/metabolismo , Receptores de Factor de Crecimiento Nervioso/metabolismo , Retina/metabolismo , Retina/fisiopatología , Degeneración Retiniana/etiología , Degeneración Retiniana/fisiopatología , Factores de Tiempo
14.
Cell Biol Int ; 39(1): 121-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25045067

RESUMEN

Embryonic stem cells (ES cells) express a transient and heterogeneous pattern of molecules, which suggests a notable mechanism to control self-renewal avoid the differentiation into germ layers. We show that 9-O-acetyl GD3 (9OacGD3), a highly expressed b-series ganglioside in neural stem (NS) cells, is expressed in undifferentiated mouse ES cells in a heterogeneous fashion. After sorting, undifferentiated 9OacGD3(+) ES cell population had higher levels of nestin and Sox2 mRNA than the 9OacGD3(-) cells. Even with elevated expression of these neural transcription factors, 9OacGD3(+) cells did not give rise to more neural progenitors than 9OacGD3(-) cells. Expression of 9OacGD3 was recovered from 9OacGD3(-) cell population, demonstrating that expression of this ganglioside in mouse embryonic stem cells is transient, and does not reflect cell fate. Our findings show that the ganglioside 9OacGD3 is expressed heterogeneously and transiently in ES cells, and this expression corresponds to higher levels of Sox2 and Nestin transcripts.


Asunto(s)
Células Madre Embrionarias/metabolismo , Gangliósidos/genética , Regulación del Desarrollo de la Expresión Génica , Animales , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Gangliósidos/metabolismo , Ratones , Nestina/genética , Nestina/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , ARN Mensajero/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
15.
Neural Regen Res ; 18(1): 81-86, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35799513

RESUMEN

Gangliosides, sialic acid-containing sphingolipids, are major constituents of neuronal membranes. According to the number of sialic acids and the structure of the oligosaccharide chain, gangliosides can be classified as simple or complex and grouped in different ganglio-series. Hundreds of gangliosides have been identified in vertebrate cells, with different expression patterns during development and related to several physiological processes, especially in the nervous system. While GD3 and its O-acetylated form, 9acGD3, are highly expressed in early developmental stages, GM1, GD1a, GD1b, and GT1b are the most abundant ganglioside species in the mature nervous system. Mutations in enzymes involved in ganglioside metabolism can lead to the accumulation of specific species, a condition termed gangliosidosis and usually marked by severe neurological impairment. Changes in ganglioside levels have also been described in several neurodegenerative diseases, such as Alzheimer's and Parkinson's. In this review, we summarized recent information about the roles of GD3, 9acGD3, GM1, GD1a, GD1b, GT1b, and other ganglioside species in nervous system development and regeneration, as well as clinical trials evaluating possible therapeutic applications of these molecules.

16.
Neural Regen Res ; 18(8): 1645-1651, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36751774

RESUMEN

Alzheimer's disease is a severe, highly disabling neurodegenerative disease, clinically characterized by a progressive decline in cognitive functions, and is the most common form of dementia in the elderly. For decades, the search for disease-modifying therapies has focused on the two main Alzheimer's disease histopathological hallmarks, seeking to prevent, mitigate, or clear the formation of extracellular aggregates of ß-amyloid peptide and intracellular neurofibrillary tangles of tau protein, although without clinical success. Mesenchymal stem cell-based therapy has emerged as a promising alternative for the treatment of Alzheimer's disease, especially because it also targets other crucial players in the pathogenesis of the disease, such as neuroinflammation, synaptic dysfunction/loss, oxidative stress, and impaired neurogenesis. Herein, we review current knowledge of the therapeutic potential of mesenchymal stem cells and their extracellular vesicles for Alzheimer's disease, discussing the most recent findings in both preclinical and clinical trials as well as how advanced technologies have helped to overcome some limitations and contributed to stimulate the development of more effective treatments.

17.
Biomedicines ; 11(12)2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38137470

RESUMEN

Chronic pain presents a major challenge in contemporary medicine, given the limited effectiveness and numerous adverse effects linked to available treatments. Recognizing the potential of the cholinergic pathway as a therapeutic target, the present work evaluates the antinociceptive activity of a combination of Cris-104, a novel α4ß2* receptor agonist, and donepezil, a central anticholinesterase agent. Isobolographic analysis revealed that equimolar combination was approximately 10 times more potent than theoretically calculated equipotent additive dose. Administration of Cris-104 and donepezil combination (3 µmol/kg) successfully reversed hyperalgesia and mechanical allodynia observed in rats subjected to spinal nerve ligation (SNL). The combination also modulated neuroinflammation by reducing astrocyte activation, evident in the decreased expression of glial fibrillary acidic protein (GFAP) in the spinal cord. The observed synergism in combining a nicotinic receptor agonist with an anticholinesterase agent underscores its potential for treating chronic pain. This alternative therapeutic distinct advantage, including dose reduction and high selectivity for the receptor, contribute to a more favorable profile with minimized adverse effects.

18.
Pediatr Res ; 71(4 Pt 2): 464-73, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22430382

RESUMEN

Despite recent advances in the treatment of neonatal hypoxic-ischemic encephalopathy (HIE) using therapeutic hypothermia, at least 30% of the cooled infants will die or have moderate/severe neurological disability. Umbilical cord blood cells (UCBCs), which are readily available at birth, have been shown to reduce sensorimotor and/or cognitive impairments in several models of brain damage, representing a promising option for the treatment of neurological diseases. In this review, we discuss recent preclinical studies that assessed the effects of UCBC transplantation in the Rice-Vannucci animal model of HIE. We also review the possible cell types and mechanisms involved in the therapeutic effect of UCBC transplantation, including neuroprotection, immunomodulation, and stimulation of neural plasticity and regeneration. In addition, we discuss how neuroimaging methods, such as bioluminescence imaging, nuclear-medicine imaging, or magnetic resonance imaging, could be used to evaluate the biodistribution of UCBCs in both preclinical and clinical studies.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Sangre Fetal/citología , Hipoxia-Isquemia Encefálica/terapia , Enfermedades del Recién Nacido/terapia , Neuroimagen/métodos , Pediatría/métodos , Animales , Humanos , Recién Nacido , Leucocitos Mononucleares , Pediatría/tendencias , Ratas
19.
Future Sci OA ; 8(4): FSO793, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35369279

RESUMEN

Aim: Intracerebral hemorrhage (ICH) has limited therapeutic options. We have shown that an intravenous injection of human umbilical cord-derived mesenchymal stromal cells (hUC-MSC) 24 h after an ICH in rats reduced the residual hematoma volume after a moderate hemorrhage but was inefficient in severe ICH. Here, we investigated whether a treatment in the hyperacute phase would be more effective in severe ICH. Materials & methods: Wistar rats were randomly selected to receive an intravenous injection of hUC-MSC or the vehicle 1 h after a severe ICH. Results: The hyperacute treatment with hUC-MSC did not affect the 22-day survival rate, the motor function or the residual hematoma volume. Conclusion: These results indicate the need for optimization of hUC-MSC-based therapies for severe ICH.

20.
Cells ; 11(2)2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-35053356

RESUMEN

The incidence and prevalence of diabetes mellitus (DM) are increasing worldwide, and the resulting cardiac complications are the leading cause of death. Among these complications is diabetes-induced cardiomyopathy (DCM), which is the consequence of a pro-inflammatory condition, oxidative stress and fibrosis caused by hyperglycemia. Cardiac remodeling will lead to an imbalance in cell survival and death, which can promote cardiac dysfunction. Since the conventional treatment of DM generally does not address the prevention of cardiac remodeling, it is important to develop new alternatives for the treatment of cardiovascular complications induced by DM. Thus, therapy with mesenchymal stem cells has been shown to be a promising approach for the prevention of DCM because of their anti-apoptotic, anti-fibrotic and anti-inflammatory effects, which could improve cardiac function in patients with DM.


Asunto(s)
Cardiomiopatías Diabéticas/terapia , Trasplante de Células Madre Mesenquimatosas , Animales , Ensayos Clínicos como Asunto , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/fisiopatología , Epigénesis Genética , Humanos , Modelos Biológicos , Remodelación Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA