Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
CNS Neurosci Ther ; 30(2): e14574, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38421088

RESUMEN

RATIONALE: Numerous epidemiological studies have reported a link between low testosterone levels and an increased risk of cerebrovascular disease in men. However, there is ongoing controversy surrounding testosterone replacement therapy due to potential side effects. PBMT has been demonstrated to improve cerebrovascular function and promote testosterone synthesis in peripheral tissues. Despite this, the molecular mechanisms that could connect PBMT with testosterone and vascular function in the brain of photothrombosis (PT)-induced stroke rats remain largely unknown. METHODS: We measured behavioral performance, cerebral blood flow (CBF), vascular permeability, and the expression of vascular-associated and apoptotic proteins in PT-induced stroke rats treated with flutamide and seven consecutive days of PBM treatment (350 mW, 808 nM, 2 min/day). To gain further insights into the mechanism of PBM on testosterone synthesis, we used testosterone synthesis inhibitors to study their effects on bEND.3 cells. RESULTS: We showed that PT stroke caused a decrease in cerebrovascular testosterone concentration, which was significantly increased by 7-day PBMT (808 nm, 350 mW/cm2 , 42 J/cm2 ). Furthermore, PBMT significantly increased cerebral blood flow (CBF) and the expression of vascular-associated proteins, while inhibiting vascular permeability and reducing endothelial cell apoptosis. This ultimately mitigated behavioral deficits in PT stroke rats. Notably, treatment with the androgen receptor antagonist flutamide reversed the beneficial effects of PBMT. Cellular experiments confirmed that PBMT inhibited cell apoptosis and increased vascular-associated protein expression in brain endothelial cell line (bEnd.3) subjected to oxygen-glucose deprivation (OGD). However, these effects were inhibited by flutamide. Moreover, mechanistic studies revealed that PBMT-induced testosterone synthesis in bEnd.3 cells was partly mediated by 17ß-hydroxysteroid dehydrogenase 5 (17ß-HSD5). CONCLUSIONS: Our study provides evidence that PBMT attenuates cerebrovascular injury and behavioral deficits associated with testosterone/AR following ischemic stroke. Our findings suggest that PBMT may be a promising alternative approach for managing cerebrovascular diseases.


Asunto(s)
Terapia por Luz de Baja Intensidad , Accidente Cerebrovascular , Humanos , Masculino , Ratas , Ratones , Animales , Testosterona/metabolismo , Andrógenos/metabolismo , Receptores Androgénicos/metabolismo , Células Endoteliales/metabolismo , Flutamida/farmacología , Flutamida/uso terapéutico , Flutamida/metabolismo , Accidente Cerebrovascular/terapia
2.
Theranostics ; 11(3): 1269-1294, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33391534

RESUMEN

Neonatal hypoxic-ischemic (HI) injury is a severe complication often leading to neonatal death and long-term neurobehavioral deficits in children. Currently, the only treatment option available for neonatal HI injury is therapeutic hypothermia. However, the necessary specialized equipment, possible adverse side effects, and limited effectiveness of this therapy creates an urgent need for the development of new HI treatment methods. Photobiomodulation (PBM) has been shown to be neuroprotective against multiple brain disorders in animal models, as well as limited human studies. However, the effects of PBM treatment on neonatal HI injury remain unclear. Methods: Two-minutes PBM (808 nm continuous wave laser, 8 mW/cm2 on neonatal brain) was applied three times weekly on the abdomen of pregnant rats from gestation day 1 (GD1) to GD21. After neonatal right common carotid artery ligation, cortex- and hippocampus-related behavioral deficits due to HI insult were measured using a battery of behavioral tests. The effects of HI insult and PBM pretreatment on infarct size; synaptic, dendritic, and white matter damage; neuronal degeneration; apoptosis; mitochondrial function; mitochondrial fragmentation; oxidative stress; and gliosis were then assessed. Results: Prenatal PBM treatment significantly improved the survival rate of neonatal rats and decreased infarct size after HI insult. Behavioral tests revealed that prenatal PBM treatment significantly alleviated cortex-related motor deficits and hippocampus-related memory and learning dysfunction. In addition, mitochondrial function and integrity were protected in HI animals treated with PBM. Additional studies revealed that prenatal PBM treatment significantly alleviated HI-induced neuroinflammation, oxidative stress, and myeloid cell/astrocyte activation. Conclusion: Prenatal PBM treatment exerts neuroprotective effects on neonatal HI rats. Underlying mechanisms for this neuroprotection may include preservation of mitochondrial function, reduction of inflammation, and decreased oxidative stress. Our findings support the possible use of PBM treatment in high-risk pregnancies to alleviate or prevent HI-induced brain injury in the perinatal period.


Asunto(s)
Hipoxia-Isquemia Encefálica/radioterapia , Hipoxia/radioterapia , Isquemia/radioterapia , Animales , Animales Recién Nacidos , Apoptosis/efectos de la radiación , Astrocitos/efectos de la radiación , Corteza Cerebral/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Hipocampo/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Masculino , Mitocondrias/efectos de la radiación , Neuronas/efectos de la radiación , Fármacos Neuroprotectores/uso terapéutico , Estrés Oxidativo/efectos de la radiación , Embarazo , Ratas , Ratas Sprague-Dawley
3.
J Biophotonics ; 13(2): e201960117, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31657525

RESUMEN

Repeated traumatic brain injury, leads to cumulative neuronal injury and neurological impairments. There are currently no effective treatments to prevent these consequences. Growing interest is building in the use of transcranial photobiomodulation (PBM) therapy to treat traumatic brain injury. Here, we examined PBM in a repeated closed head injury (rCHI) rat model. Rats were administered a total of three closed head injuries, with each injury separated by 5 days. PBM treatment was initiated 2 hours after the first injury and administered daily for a total of 15 days. We found that PBM-treated rCHI rats had a significant reduction in motor ability, anxiety and cognitive deficits compared to CHI group. PBM group showed an increase of synaptic proteins and surviving neurons, along with a reduction in reactive gliosis and neuronal injury. These findings highlight the complexity of gliosis and neuronal injury following rCHI and suggest that PBM may be a viable treatment option to mitigate these effects and their detrimental consequences.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Disfunción Cognitiva , Traumatismos Cerrados de la Cabeza , Terapia por Luz de Baja Intensidad , Animales , Lesiones Traumáticas del Encéfalo/terapia , Neuronas , Ratas
4.
J Biophotonics ; 12(6): e201800359, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30652418

RESUMEN

Neonatal hypoxia-ischemia (HI) injury caused by oxygen deprivation is the most common cause of mortality and severe neurologic deficits in neonates. The present work evaluated the preventative effect of photobiomodulation (PBM) preconditioning, and its underlying mechanism of action on brain damage in an HI model in neonatal rats. According to the optimal time response of ATP levels in brain samples removed from normal rats, a PBM preconditioning (PBM-P) regimen (808 nm CW laser, 1 cm2 spot, 100 mW/cm2 , 12 J/cm2 ) was delivered to the scalp 6 hours before HI. PBM-P significantly attenuated cognitive impairment, volume shrinkage in the brain, neuron loss, dendritic and synaptic injury after HI. Further mechanistic investigation found that PBM-P could restore HI-induced mitochondrial dynamics and inhibit mitochondrial fragmentation, followed by a robust suppression of cytochrome c release, and prevention of neuronal apoptosis by inhibition of caspase activation. Our work suggests that PBM-P can attenuate HI-induced brain injury by maintaining mitochondrial dynamics and inhibiting the mitochondrial apoptotic pathway.


Asunto(s)
Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/prevención & control , Hipoxia-Isquemia Encefálica/complicaciones , Terapia por Luz de Baja Intensidad , Animales , Animales Recién Nacidos , Apoptosis/efectos de la radiación , Conducta Animal/efectos de la radiación , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Citocromos c/metabolismo , Células Dendríticas/patología , Células Dendríticas/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Masculino , Dinámicas Mitocondriales/efectos de la radiación , Neuronas/patología , Neuronas/efectos de la radiación , Ratas , Ratas Sprague-Dawley , Sinapsis/patología , Sinapsis/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA