Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Biophotonics ; 17(2): e202300343, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37909411

RESUMEN

Repeated closed head injury (rCHI) is one of the most common brain injuries. Although extensive studies have focused on how to treat rCHI-induced brain injury and reduce the possibility of developing memory deficits, the prevention of rCHI-induced anxiety has received little research attention. The current study was designed to assess the effects of photobiomodulation (PBM) therapy in preventing anxiety following rCHI. The rCHI disease model was constructed by administering three repeated closed-head injuries within an interval 5 days. 2-min daily PBM therapy using an 808 nm continuous wave laser at 350 mW/cm2 on the scalp was implemented for 20 days. We found that PBM significantly ameliorated rCHII-induced anxiety-like behaviors, neuronal apoptosis, neuronal injury, promotes astrocyte/microglial polarization to anti-inflammatory phenotype, preserves mitochondrial fusion-related protein MFN2, attenuates the elevated mitochondrial fission-related protein DRP1, and mitigates neuronal senescence. We concluded that PBM therapy possesses great potential in preventing anxiety following rCHI.


Asunto(s)
Traumatismos Cerrados de la Cabeza , Terapia por Luz de Baja Intensidad , Humanos , Apoptosis , Neuronas , Ansiedad/etiología , Ansiedad/prevención & control
2.
Neuroscience ; 522: 57-68, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37164305

RESUMEN

Repeated mild traumatic brain injury (rTBI), one of the most common forms of traumatic brain injury, is a worldwide severe public health concern. rTBI induces cumulative neuronal injury, neurological dysfunction, and cognitive deficits. Although there are clinical treatment methods, there is still an urgent need to develop preventive approaches for susceptible populations. Using a repeated closed head injury (rCHI) rat model, we interrogate the effect of sub-lethal hyperthermia preconditioning (SHP) on rCHI-induced neuronal injury and behavioral changes. Our study applied the repeated weight-drop model to induce the rCHI. According to the changes of heat shock protein 70 (HSP 70) in the cortex and hippocampus following a single SHP treatment in normal rats, the SHP was delivered to the rats 18 h before rCHI. We found that HSP significantly alleviated rCHI-induced anxiety-like behaviors and impairments in motor abilities and spatial memory. SHP exerts significant neuroprotection against rCHI-induced neuronal damage, apoptosis, and neuroinflammation. Our findings support the potential use of SHP as a preventative approach for alleviating rCHI-induced brain damage.


Asunto(s)
Conmoción Encefálica , Lesiones Encefálicas , Traumatismos Cerrados de la Cabeza , Hipertermia Inducida , Fármacos Neuroprotectores , Ratas , Animales , Fármacos Neuroprotectores/farmacología , Modelos Animales de Enfermedad
3.
J Alzheimers Dis ; 83(4): 1415-1429, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34219711

RESUMEN

BACKGROUND: Anxious-depressive-like behavior has been recognized as an early endophenotype in Alzheimer's disease (AD). Recent studies support early treatment of anxious-depressive-like behavior as a potential target to alleviate memory loss and reduce the risk of developing dementia. We hypothesize that photobiomodulation (PBM) could be an effective method to alleviate depression and anxiety at the early stage of AD pathogenesis. OBJECTIVE: To analyze the effect of PBM treatment on anxious-depressive-like behavior at the early stage of AD. METHODS: Using a novel transgenic AD rat model, animals were divided into wild-type, AD+sham PBM, and AD+PBM groups. Two-minute daily PBM (irradiance: 25 mW/cm2 and fluence: 3 J/cm2 at the cortical level) was applied transcranially to the brain of AD animals from 2 months of age to 10 months of age. After completing PBM treatment at 10 months of age, behavioral tests were performed to measure learning, memory, and anxious-depressive-like behavior. Neuronal apoptosis, neuronal degeneration, neuronal damage, mitochondrial function, neuroinflammation, and oxidative stress were measured to test the effects of PBM on AD animals. RESULTS: Behavioral tests showed that: 1) no spatial memory deficits were detected in TgF344 rats at 10 months of age; 2) PBM alleviated anxious-depressive-like behavior in TgF344 rats; 3) PBM attenuated neuronal damage, degeneration, and apoptosis; and 4) PBM suppresses neuroinflammation and oxidative stress. CONCLUSION: Our findings support our hypothesis that PBM could be an effective method to alleviate depression and anxiety during the early stage of AD development. The mechanism underlying these beneficial effects may be due to the improvement of mitochondria function and integrity and the inhibition of neuroinflammation and oxidative stress.


Asunto(s)
Ansiedad/prevención & control , Depresión/prevención & control , Terapia por Luz de Baja Intensidad , Ratas Transgénicas , Enfermedad de Alzheimer/radioterapia , Animales , Apoptosis , Modelos Animales de Enfermedad , Humanos , Masculino , Aprendizaje por Laberinto , Mitocondrias/efectos de la radiación , Neuronas/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Ratas
4.
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
5.
Transl Neurodegener ; 9(1): 19, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32475349

RESUMEN

Mitochondrial dysfunction plays a central role in the formation of neuroinflammation and oxidative stress, which are important factors contributing to the development of brain disease. Ample evidence suggests mitochondria are a promising target for neuroprotection. Recently, methods targeting mitochondria have been considered as potential approaches for treatment of brain disease through the inhibition of inflammation and oxidative injury. This review will discuss two widely studied approaches for the improvement of brain mitochondrial respiration, methylene blue (MB) and photobiomodulation (PBM). MB is a widely studied drug with potential beneficial effects in animal models of brain disease, as well as limited human studies. Similarly, PBM is a non-invasive treatment that promotes energy production and reduces both oxidative stress and inflammation, and has garnered increasing attention in recent years. MB and PBM have similar beneficial effects on mitochondrial function, oxidative damage, inflammation, and subsequent behavioral symptoms. However, the mechanisms underlying the energy enhancing, antioxidant, and anti-inflammatory effects of MB and PBM differ. This review will focus on mitochondrial dysfunction in several different brain diseases and the pathological improvements following MB and PBM treatment.


Asunto(s)
Encefalopatías/tratamiento farmacológico , Encefalopatías/radioterapia , Terapia por Luz de Baja Intensidad/métodos , Azul de Metileno/administración & dosificación , Mitocondrias/efectos de los fármacos , Neuroprotección/fisiología , Animales , Encefalopatías/diagnóstico , Humanos , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Neuroprotección/efectos de los fármacos
6.
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
7.
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
8.
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
9.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA