Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Neurobiol ; 56(3): 1852-1869, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29951942

RESUMEN

Hypothermia is currently the only approved therapy for global cerebral ischemia (GCI) after cardiac arrest; however, it unfortunately has multiple adverse effects. As a noninvasive procedure, photobiomodulation (PBM) therapy has emerged as a potential novel treatment for brain injury. PBM involves the use of low-level laser light therapy to influence cell behavior. In this study, we evaluated the therapeutic effects of PBM treatment with an 808-nm diode laser initiated 6 h after GCI. It was noted that PBM dose-dependently protected against GCI-induced neuronal death in the vulnerable hippocampal CA1 subregion. Functional assessments demonstrated that PBM markedly preserved both short-term (a week) and long-term (6 months) spatial learning and memory function following GCI. Further mechanistic studies revealed that PBM post-treatment (a) preserved healthy mitochondrial dynamics and suppressed substantial mitochondrial fragmentation of CA1 neurons, by reducing the detrimental Drp1 GTPase activity and its interactions with adaptor proteins Mff and Fis1 and by balancing mitochondrial targeting fission and fusion protein levels; (b) reduced mitochondrial oxidative damage and excessive mitophagy and restored mitochondrial overall health status and preserved mitochondrial function; and (c) suppressed mitochondria-dependent apoptosome formation/caspase-3/9 apoptosis-processing activities. Additionally, we validated, in an in vitro ischemia model, that cytochrome c oxidase served as a key PBM target for mitochondrial function preservation and neuroprotection. Our findings suggest that PBM serves as a promising therapeutic strategy for the functional recovery after GCI, with mechanisms involving PBM's preservation on mitochondrial dynamics and functions and the inhibition of delayed apoptotic neuronal death in GCI.


Asunto(s)
Isquemia Encefálica/radioterapia , Muerte Celular/efectos de la radiación , Hipocampo/efectos de la radiación , Terapia por Luz de Baja Intensidad , Mitocondrias/efectos de la radiación , Dinámicas Mitocondriales/efectos de la radiación , Animales , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto/efectos de la radiación , Mitocondrias/metabolismo , Neuronas/metabolismo , Neuronas/efectos de la radiación , Ratas , Ratas Sprague-Dawley
2.
J Mol Neurosci ; 65(4): 514-526, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30032397

RESUMEN

Photobiomodulation (PBM) has been demonstrated as a neuroprotective strategy, but its effect on perinatal hypoxic-ischemic encephalopathy is still unknown. The current study was designed to shed light on the potential beneficial effect of PBM on neonatal brain injury induced by hypoxia ischemia (HI) in a rat model. Postnatal rats were subjected to hypoxic-ischemic insult, followed by a 7-day PBM treatment via a continuous wave diode laser with a wavelength of 808 nm. We demonstrated that PBM treatment significantly reduced HI-induced brain lesion in both the cortex and hippocampal CA1 subregions. Molecular studies indicated that PBM treatment profoundly restored mitochondrial dynamics by suppressing HI-induced mitochondrial fragmentation. Further investigation of mitochondrial function revealed that PBM treatment remarkably attenuated mitochondrial membrane collapse, accompanied with enhanced ATP synthesis in neonatal HI rats. In addition, PBM treatment led to robust inhibition of oxidative damage, manifested by significant reduction in the productions of 4-HNE, P-H2AX (S139), malondialdehyde (MDA), as well as protein carbonyls. Finally, PBM treatment suppressed the activation of mitochondria-dependent neuronal apoptosis in HI rats, as evidenced by decreased pro-apoptotic cascade 3/9 and TUNEL-positive neurons. Taken together, our findings demonstrated that PBM treatment contributed to a robust neuroprotection via the attenuation of mitochondrial dysfunction, oxidative stress, and final neuronal apoptosis in the neonatal HI brain.


Asunto(s)
Hipoxia-Isquemia Encefálica/terapia , Terapia por Luz de Baja Intensidad/métodos , Animales , Apoptosis , Corteza Cerebral/metabolismo , Femenino , Hipocampo/metabolismo , Masculino , Mitocondrias/metabolismo , Estrés Oxidativo , Carbonilación Proteica , Ratas
3.
Exp Neurol ; 299(Pt A): 86-96, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29056360

RESUMEN

Recent work has indicated that photobiomodulation (PBM) may beneficially alter the pathological status of several neurological disorders, although the mechanism currently remains unclear. The current study was designed to investigate the beneficial effect of PBM on behavioral deficits and neurogenesis in a photothrombotic (PT) model of ischemic stroke in rats. From day 1 to day 7 after the establishment of PT model, 2-minute daily PBM (CW, 808nm, 350mW/cm2, total 294J at scalp level) was applied on the infarct injury area (1.8mm anterior to the bregma and 2.5mm lateral from the midline). Rats received intraperitoneal injections of 5-bromodeoxyuridine (BrdU) twice daily (50mg/kg) from day 2 to 8 post-stoke, and samples were collected at day 14. We demonstrated that PBM significantly attenuated behavioral deficits and infarct volume induced by PT stroke. Further investigation displayed that PBM remarkably enhanced neurogenesis and synaptogenesis, as evidenced by immunostaining of BrdU, Ki67, DCX, MAP2, spinophilin, and synaptophysin. Mechanistic studies suggested beneficial effects of PBM were accompanied by robust suppression of reactive gliosis and the production of pro-inflammatory cytokines. On the contrary, the release of anti-inflammatory cytokines, cytochrome c oxidase activity and ATP production in peri-infarct regions were elevated following PBM treatment. Intriguingly, PBM could effectively switch an M1 microglial phenotype to an anti-inflammatory M2 phenotype. Our novel findings indicated that PBM is capable of promoting neurogenesis after ischemic stroke. The underlying mechanisms may rely on: 1) promotion of proliferation and differentiation of internal neuroprogenitor cells in the peri-infarct zone; 2) improvement of the neuronal microenvironment by altering inflammatory status and promoting mitochondrial function. These findings provide strong support for the promising therapeutic effect of PBM on neuronal repair following ischemic stroke.


Asunto(s)
Terapia por Luz de Baja Intensidad/métodos , Células-Madre Neurales , Neurogénesis , Accidente Cerebrovascular/terapia , Animales , Conducta Animal , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Diferenciación Celular , Proliferación Celular , Microambiente Celular , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/patología , Citocinas/metabolismo , Proteína Doblecortina , Complejo IV de Transporte de Electrones/metabolismo , Masculino , Microglía/patología , Mitocondrias/metabolismo , Ratas , Ratas Sprague-Dawley , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/psicología , Sinapsis/efectos de los fármacos
4.
Mol Neurobiol ; 54(6): 4551-4559, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-27379735

RESUMEN

Major depressive disorder (MDD) is one of the leading forms of psychiatric disorders, characterized by aversion to mobility, neurotransmitter deficiency, and energy metabolic decline. Low-level laser therapy (LLLT) has been investigated in a variety of neurodegenerative disorders associated with mitochondrial dysfunction and functional impairments. The goal of this study was to examine the effect of LLLT on depression-like behaviors and to explore the potential mechanism by detecting mitochondrial function following LLLT. Depression models in space restriction mice and Abelson helper integration site-1 (Ahi1) knockout (KO) mice were employed in this work. Our results revealed that LLLT effectively improved depression-like behaviors, in the two depression mice models, by decreasing immobility duration in behavioral despair tests. In addition, ATP biosynthesis and the level of mitochondrial complex IV expression and activity were significantly elevated in prefrontal cortex (PFC) following LLLT. Intriguingly, LLLT has no effects on ATP content and mitochondrial complex I-IV levels in other tested brain regions, hippocampus and hypothalamus. As a whole, these findings shed light on a novel strategy of transcranial LLLT on depression improvement by ameliorating neurotransmitter abnormalities and promoting mitochondrial function in PFC. The present work provides concrete groundwork for further investigation of LLLT for depression treatment.


Asunto(s)
Conducta Animal/efectos de la radiación , Depresión/terapia , Terapia por Luz de Baja Intensidad , Proteínas Adaptadoras del Transporte Vesicular , Adenosina Trifosfato/biosíntesis , Animales , Depresión/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Ratones Endogámicos ICR , Ratones Noqueados , Mitocondrias/metabolismo , Neurotransmisores/metabolismo , Fenotipo , Corteza Prefrontal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Restricción Física , Estrés Psicológico/complicaciones
5.
Neurobiol Aging ; 49: 165-182, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815990

RESUMEN

Beta amyloid (Aß) is well accepted to play a central role in the pathogenesis of Alzheimer's disease (AD). The present work evaluated the therapeutic effects of low-level laser irradiation (LLI) on Aß-induced neurotoxicity in rat hippocampus. Aß 1-42 was injected bilaterally to the hippocampus CA1 region of adult male rats, and 2-minute daily LLI treatment was applied transcranially after Aß injection for 5 consecutive days. LLI treatment suppressed Aß-induced hippocampal neurodegeneration and long-term spatial and recognition memory impairments. Molecular studies revealed that LLI treatment: (1) restored mitochondrial dynamics, by altering fission and fusion protein levels thereby suppressing Aß-induced extensive fragmentation; (2) suppressed Aß-induced collapse of mitochondrial membrane potential; (3) reduced oxidized mitochondrial DNA and excessive mitophagy; (4) facilitated mitochondrial homeostasis via modulation of the Bcl-2-associated X protein/B-cell lymphoma 2 ratio and of mitochondrial antioxidant expression; (5) promoted cytochrome c oxidase activity and adenosine triphosphate synthesis; (6) suppressed Aß-induced glucose-6-phosphate dehydrogenase and nicotinamide adenine dinucleotide phosphate oxidase activity; (7) enhanced the total antioxidant capacity of hippocampal CA1 neurons, whereas reduced the oxidative damage; and (8) suppressed Aß-induced reactive gliosis, inflammation, and tau hyperphosphorylation. Although development of AD treatments has focused on reducing cerebral Aß levels, by the time the clinical diagnosis of AD or mild cognitive impairment is made, the brain is likely to have already been exposed to years of elevated Aß levels with dire consequences for multiple cellular pathways. By alleviating a broad spectrum of Aß-induced pathology that includes mitochondrial dysfunction, oxidative stress, neuroinflammation, neuronal apoptosis, and tau pathology, LLI could represent a new promising therapeutic strategy for AD.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/radioterapia , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Región CA1 Hipocampal/metabolismo , Terapia por Luz de Baja Intensidad , Dinámicas Mitocondriales , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Enfermedad de Alzheimer/psicología , Animales , Apoptosis , Región CA1 Hipocampal/patología , Inflamación , Láseres de Semiconductores/uso terapéutico , Terapia por Luz de Baja Intensidad/métodos , Masculino , Neuronas/patología , Estrés Oxidativo , Ratas Sprague-Dawley , Reconocimiento en Psicología , Memoria Espacial , Tauopatías/etiología , Tauopatías/radioterapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA