Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Arch Gerontol Geriatr ; 106: 104874, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36470179

RESUMEN

Exosomes released from different cell types of the central nervous system play an essential role in the pathogenesis of Alzheimer's disease (AD). In this study, we aimed to create an animal model by injecting exosomes that carry AD markers into the brain to shed light on the mechanism behind Alzheimer's pathology. Exosomes obtained from mouse Neuro2A, to which Aß toxicity model applied, were used as a mediator to build an AD phenotype. For this purpose, exosomes were administered into hippocampal CA3 region of mice with different ages. Firstly, the possible role of exosomes on brain volume was analyzed. Then, neurons and astrocytes were evaluated for survival. In addition, the progenitor cells' differentiation capacity was investigated via BrdU staining. AKT signaling pathway components were examined to detect the molecular mechanisms behind the exosomal function. We found different responses in different age groups. Expression of APP upregulated only in young animals upon delivery of Aß-exosomes. Interestingly, young animals represented increased numbers of neurons in the hippocampus, and neurogenesis was found to be restricted after Aß-Ex injections. However, in relation to exosome administration, the glial intensity increased in aged animals. Lastly, phosphorylation of survival kinase AKT was downregulated due to the presence of Aß in both young and old animals. The findings reveal that the exosomes from an in vitro Aß toxicity model may induce different responses in an age-dependent manner. This study is the first to report the relationship between exosomal function and aging by evaluating the key molecules.


Asunto(s)
Enfermedad de Alzheimer , Exosomas , Animales , Ratones , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/toxicidad , Péptidos beta-Amiloides/metabolismo , Modelos Animales de Enfermedad , Exosomas/metabolismo , Exosomas/patología , Neuronas/metabolismo , Neuronas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo
2.
Mol Neurobiol ; 59(1): 574-589, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34735672

RESUMEN

Phosphodiesterase 10A (PDE10A) hydrolyzes adenosine 3',5'-cyclic monophosphate (cAMP) and guanosine 3',5'-cyclic monophosphate (cGMP). It is highly expressed in the striatum. Recent evidence implied that PDE10A may be involved in the inflammatory processes following injury, such as ischemic stroke. Its role in ischemic injury was unknown. Herein, we exposed mice to 90 or 30-min middle cerebral artery occlusion, followed by the delivery of the highly selective PDE10A inhibitor TAK-063 (0.3 mg/kg or 3 mg/kg) immediately after reperfusion. Animals were sacrificed after 24 or 72 h, respectively. Both TAK-063 doses enhanced neurological function, reduced infarct volume, increased neuronal survival, reduced brain edema, and increased blood-brain barrier integrity, alongside cerebral microcirculation improvements. Post-ischemic neuroprotection was associated with increased phosphorylation (i.e., activation) of pro-survival Akt, Erk-1/2, GSK-3α/ß and anti-apoptotic Bcl-xL abundance, decreased phosphorylation of pro-survival mTOR, and HIF-1α, MMP-9 and pro-apoptotic Bax abundance. Interestingly, PDE10A inhibition reduced inflammatory cytokines/chemokines, including IFN-γ and TNF-α, analyzed by planar surface immunoassay. In addition, liquid chromatography-tandem mass spectrometry revealed 40 proteins were significantly altered by TAK-063. Our study established PDE10A as a target for ischemic stroke therapy.


Asunto(s)
Edema Encefálico/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Inhibidores de Fosfodiesterasa/uso terapéutico , Hidrolasas Diéster Fosfóricas/metabolismo , Animales , Edema Encefálico/metabolismo , Modelos Animales de Enfermedad , Accidente Cerebrovascular Isquémico/metabolismo , Ratones , Microcirculación/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Inhibidores de Fosfodiesterasa/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirazoles/farmacología , Pirazoles/uso terapéutico , Piridazinas/farmacología , Piridazinas/uso terapéutico , Transducción de Señal/efectos de los fármacos
3.
Behav Brain Res ; 392: 112719, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32479849

RESUMEN

Apart from its well-established therapeutic activity on bipolar disorder and depression, lithium exerts neuroprotective activity upon neurodegenerative disorders, such as traumatic brain injury (TBI). However, the cellular signaling mechanisms mediating lithium's neuroprotective activity and long-term dose- and time-dependent effects on close and remote proximity are largely unknown. Herein, we tested prophylactic and acute effects of lithium (2 mmol/kg) after cold- induced TBI. In both conditions, treatments with lithium resulted in reduced infarct volume and apoptosis. Its acute treatment resulted in the increase of Akt, ERK-1/2 and GSK-3 α/ß phosphoylations. Interestingly, its prophylactic treatment instead resulted in decreased phosphorylations of Akt, ERK-1/2, p38, JNK-1 moderately and GSK-3 α/ß significantly. Then, we tested subacute (35-day follow-up) role of low (0.2 mmol/kg) and high dose (2 mmol/kg) lithium and revealed that high dose lithium group was the most mobile so the least depressed in the tail suspension test. Anxiety level was assessed by light-dark test, all groups' anxiety levels were decreased with time, but lithium had no effect on anxiety like behavior. When subacute effects of injury and drug treatment were evaluated on the defined brain regions, infarct volume was decreased in the high dose lithium group significantly. In contrast to other brain regions, hippocampal atrophies were observed in both lithium treatment groups, which were significant in the low dose lithium group in both hemispheres, which was associated with the reduced cell proliferation and neurogenesis. Our data demonstrate that lithium treatment protects neurons from TBI. However, long term particularly low-dose lithium causes hippocampal atrophy and decreased neurogenesis.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Litio/farmacología , Animales , Apoptosis/efectos de los fármacos , Encéfalo/efectos de los fármacos , Lesiones Encefálicas/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3/metabolismo , Litio/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Ratones , Ratones Endogámicos BALB C , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo
4.
Exp Neurol ; 331: 113364, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32454038

RESUMEN

Owing to its potent longterm neuroprotective and neurorestorative properties, glial cell line-derived neurotrophic factor (GDNF) is currently studied in neurodegenerative disease clinical trials. However, little is known about the longterm effect of GDNF on neurological recovery, brain remodeling and neuroplasticity in the post-acute phase of ischemic stroke. In a comprehensive set of experiments, we examined the effects of lentiviral GDNF administration after ischemic stroke. GDNF reduced neurological deficits, neuronal injury, blood-brain barrier permeability in the acute phase in mice. As compared with control, enhanced motor-coordination and spontaneous locomotor activity were noted in GDNF-treated mice, which were associated with increased microvascular remodeling, increased neurogenesis and reduced glial scar formation in the peri-infarct tissue. We observed reduced brain atrophy and increased plasticity of contralesional pyramidal tract axons that crossed the midline in order to innervate denervated neurons in the ipsilesional red and facial nuclei. Contralesional axonal plasticity by GDNF was associated with decreased abundance of the axonal growth inhibitors brevican and versican in contralesional and ipsilesional brain tissue, reduced abundance of the growth repulsive guidance molecule ephrin b1 in contralesional brain tissue, increased abundance of the midline growth repulsive protein Slit1 in contralesional brain tissue and reduced abundance of Slit1's receptor Robo2 in ipsilesional brain tissue. These data indicate that GDNF potently induces longterm neurological recovery, peri-infarct brain remodeling and contralesional neuroplasticity, which are associated with the fine-tuned regulation of axonal growth inhibitors and guidance molecules that facilitate the growth of contralesional corticofugal axons in the direction to the ipsilesional hemisphere.


Asunto(s)
Factor Neurotrófico Derivado de la Línea Celular Glial/administración & dosificación , Accidente Cerebrovascular Isquémico/patología , Neurogénesis/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Recuperación de la Función/efectos de los fármacos , Animales , Lentivirus , Masculino , Ratones , Ratones Endogámicos C57BL
5.
Behav Brain Res ; 379: 112338, 2020 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-31733311

RESUMEN

BACKGROUND: Newborn hypoxia ischemia (HI) is one of the most prevalent cases in the emergency and can result from fetal hypoxia during delivery. In HI, restricted blood supply to the fetal brain may cause epilepsy or mental disorders. METHODS: In the present study, seven-day-old pups were subjected HI and treated with different normobaric oxygen (NBO) concentrations (21%, 70% or 100%). In the acute phase, we analyzed infarct area, disseminate neuronal injury and surviving neurons. In addition, we studied the regulation of PTEN and MMP-9 proteins which were suggested to be activated by HI in the ischemic tissue. Moreover, long-term effects of NBO treatments were evaluated with open field, rotarod and Barnes maze tests. We also examined axonal plasticity with EGFP-AAV injection. RESULTS: Here, we demonstrate that hyperoxic NBO concentration causes an increase in cellular survival and a decrease in the number of apoptotic cells, meanwhile inhibiting the proteins involved in cellular death mechanisms. Moreover, we found that hyperoxia decreases anxiety, promotes motor coordination and improve spatial learning and memory. Notably that axonal sprouting was promoted by hyperoxia. CONCLUSION: Our data suggest that NBO is a promising approach for the treatment of newborn HI, which encourage proof-of-concept studies in newborn.


Asunto(s)
Hipoxia-Isquemia Encefálica/terapia , Enfermedades del Recién Nacido/terapia , Actividad Motora/fisiología , Plasticidad Neuronal/fisiología , Terapia por Inhalación de Oxígeno , Recuperación de la Función/fisiología , Aprendizaje Espacial/fisiología , Animales , Animales Recién Nacidos , Conducta Animal/fisiología , Supervivencia Celular/fisiología , Modelos Animales de Enfermedad , Humanos , Hipoxia-Isquemia Encefálica/metabolismo , Recién Nacido , Metaloproteinasa 9 de la Matriz/metabolismo , Neuronas/fisiología , Terapia por Inhalación de Oxígeno/métodos , Fosfohidrolasa PTEN/metabolismo , Ratas , Ratas Sprague-Dawley
6.
Mol Biol Rep ; 46(1): 241-250, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30406889

RESUMEN

Traumatic brain injury (TBI) is the leading cause of mortality and morbidity in young adults and children in the industrialized countries; however, there are presently no FDA approved therapies. TBI results in oxidative stress due to the overproduction of reactive oxygen species and overwhelming of the endogenous antioxidant mechanisms. Recently, it has been reported that antioxidants including phytochemicals have a protective role against oxidative damage and inflammation after TBI. To analyze the effects of a naturally occurring antioxidant molecule, allyl isothiocyanate (AITC), on the nuclear factor erythroid 2-related factor 2 (Nrf2) and nuclear factor kappa B (NF-κB) signaling pathways in TBI, a cryogenic injury model was induced in mice. Here, we showed that AITC administered immediately after the injury significantly decreased infarct volume and blood-brain barrier (BBB) permeability. Protein levels of proinflammatory cytokines interleukin-1ß (IL1ß) and interleukin-6 (IL6), glial fibrillary acidic protein (GFAP) and NF-κB were decreased, while Nrf2, growth-associated protein 43 (GAP43) and neural cell adhesion molecule levels were increased with AITC when compared with vehicle control. Our results demonstrated that the antioxidant molecule AITC, when applied immediately after TBI, provided beneficial effects on inflammatory processes while improving infarct volume and BBB permeability. Increased levels of plasticity markers, as well as an antioxidant gene regulator, Nrf2, by AITC, suggest that future studies are warranted to assess the protective activities of dietary or medicinal AITC in clinical studies.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Isotiocianatos/farmacología , Animales , Antioxidantes/farmacología , Lesiones Encefálicas/tratamiento farmacológico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Hemo-Oxigenasa 1/efectos de los fármacos , Inflamación/tratamiento farmacológico , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/efectos de los fármacos , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Isotiocianatos/metabolismo , Masculino , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Factor 2 Relacionado con NF-E2/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
7.
CNS Neurol Disord Drug Targets ; 17(6): 439-447, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29714150

RESUMEN

BACKGROUND: Cinnamon polyphenol extract is a traditional spice commonly used in different areas of the world for the treatment of different disease conditions which are associated with inflammation and oxidative stress. Despite many preclinical studies showing the anti-oxidative and antiinflammatory effects of cinnamon, the underlying mechanisms in signaling pathways via which cinnamon protects the brain after brain trauma remained largely unknown. However, there is still no preclinical study delineating the possible molecular mechanism of neuroprotective effects cinnamon polyphenol extract in Traumatic Brain Injury (TBI). The primary aim of the current study was to test the hypothesis that cinnamon polyphenol extract administration would improve the histopathological outcomes and exert neuroprotective activity through its antioxidative and anti-inflammatory properties following TBI. METHODS: To investigate the effects of cinnamon, we induced brain injury using a cold trauma model in male mice that were treated with cinnamon polyphenol extract (10 mg/kg) or vehicle via intraperitoneal administration just after TBI. Mice were divided into two groups: TBI+vehicle group and TBI+ cinnamon polyphenol extract group. Brain samples were collected 24 h later for analysis. RESULTS: We have shown that cinnamon polyphenol extract effectively reduced infarct and edema formation which were associated with significant alterations in inflammatory and oxidative parameters, including nuclear factor-κB, interleukin 1-beta, interleukin 6, nuclear factor erythroid 2-related factor 2, glial fibrillary acidic protein, neural cell adhesion molecule, malondialdehyde, superoxide dismutase, catalase and glutathione peroxidase. CONCLUSION: Our results identify an important neuroprotective role of cinnamon polyphenol extract in TBI which is mediated by its capability to suppress the inflammation and oxidative injury. Further, specially designed experimental studies to understand the molecular cross-talk between signaling pathways would provide valuable evidence for the therapeutic role of cinnamon in TBI and other TBI related conditions.


Asunto(s)
Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Cinnamomum zeylanicum/química , Fármacos Neuroprotectores/uso terapéutico , Extractos Vegetales/uso terapéutico , Animales , Edema Encefálico/etiología , Edema Encefálico/prevención & control , Infarto Encefálico/etiología , Infarto Encefálico/prevención & control , Lesiones Traumáticas del Encéfalo/complicaciones , Catalasa/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Molécula 1 de Adhesión Intercelular/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Superóxido Dismutasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...