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

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Med Sci ; 18(16): 3821-3830, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790058

RESUMEN

Background: Neuronal apoptosis and inflammation in the ventral horn of the spinal cord contribute to denervated muscle atrophy post-burn. Hyperbaric oxygen therapy (HBOT) exerts anti-inflammation and neuroprotection. Furthermore, hypoxia-inducible factor (HIF)-1α has been reported to promote inflammation and apoptosis. We investigated the therapeutic potential of HBOT and the role of HIF-1α post-burn. Methods: Sprague-Dawley rats were divided into three groups: a control group, an untreated burn group receiving burn and sham treatment, and a HBOT group receiving burn injury and HBOT. The burn injury was induced with 75ºC ± 5ºC at the right hindpaw. HBOT (100% oxygen at 2.5 atmosphere, 90 min/day) and sham HBOT (21% oxygen at 1 atmosphere, 90 min/day) was started on day 28 after burn injury and continued for 14 treatments (days 28-41). Incapacitance (hind limb weight bearing) testing was conducted before burn and weekly after burn. At day 42 post-burn, the gastrocnemius muscle and the spinal cord ventral horn were analyzed. Results: HBOT improved burn-induced weight bearing imbalance. At day 42 post-burn, less gastrocnemius muscle atrophy and fibrosis were noted in the HBOT group than in the untreated burn group. In the ventral horn, HBOT attenuated the neuronal apoptosis and glial activation post-burn. The increases in phosphorylated AKT/mTOR post-burn were reduced after HBOT. HBOT also inhibited HIF-1α signaling, as determined by immunofluorescence and western blot. Conclusions: HBOT reduces burn-induced neuronal apoptosis in the ventral horn, possibly through HIF-1α signaling.


Asunto(s)
Quemaduras/terapia , Oxigenoterapia Hiperbárica , Atrofia Muscular/terapia , Animales , Quemaduras/complicaciones , Quemaduras/patología , Modelos Animales de Enfermedad , Masculino , Neuronas Motoras/fisiología , Desnervación Muscular/efectos adversos , Músculo Esquelético/inervación , Músculo Esquelético/patología , Atrofia Muscular/etiología , Atrofia Muscular/patología , Neuroprotección/fisiología , Ratas , Ratas Sprague-Dawley
2.
Support Care Cancer ; 29(11): 6841-6850, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34003380

RESUMEN

BACKGROUND AND OBJECTIVES: Chemotherapy-induced peripheral neuropathy (CIPN) is considered one of the most common sequelae in patients with cancer who experience consistent abnormal sensations or pain symptoms during or after paclitaxel (PAC) chemotherapy. Transient receptor potential vanilloid 1 (TRPV1) and toll-like receptor 4 (TLR4) have been reported to interact in the nervous system in patients with CIPN. The antinociceptive effects of hyperbaric oxygen therapy (HBOT) on CIPN was demonstrated in this study through behavior tests. Using a CIPN rat model, we examined the effects of simultaneous HBOT (SHBOT) administration during chemotherapy and discovered that SHBOT achieved better reversal effects than chemotherapy alone. MATERIALS AND METHODS: Twenty-four rats were randomly allocated to four groups: control, PAC, SHBOT, and HBOT after PAC groups. Behavior tests were performed to evaluate mechanical allodynia and thermal hyperalgesia status. Tissues from the spinal cord and dorsal root ganglions were collected, and TLR4 and TRPV1 expression and microglial activation were investigated through immunofluorescence (IF) staining. RESULTS: The mechanical and thermal behavior tests revealed that HBOT intervention during PAC treatment led to the early alleviation of CIPN symptoms and inhibited CIPN deterioration. IF staining revealed that TLR4, TRPV1, and microglial activation were all upregulated in PAC-injected rats and exhibited early and significant downregulation in SHBOT-treated rats. CONCLUSION: This study is the first to demonstrate that the use of SHBOT during PAC treatment has potential for the early suppression of CIPN initiation and deterioration, indicating that it can alleviate CIPN symptoms and may reverse CIPN in patients undergoing systemic chemotherapy.


Asunto(s)
Antineoplásicos , Oxigenoterapia Hiperbárica , Enfermedades del Sistema Nervioso Periférico , Animales , Antineoplásicos/uso terapéutico , Ganglios Espinales/metabolismo , Humanos , Paclitaxel/efectos adversos , Enfermedades del Sistema Nervioso Periférico/tratamiento farmacológico , Enfermedades del Sistema Nervioso Periférico/terapia , Ratas , Canales Catiónicos TRPV/uso terapéutico , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/uso terapéutico
3.
Medicina (Kaunas) ; 56(6)2020 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-32471203

RESUMEN

Since the outbreak of coronavirus disease 2019 (COVID-19) in Wuhan, China in December 2019 and its subsequent global spread, Taiwan has been combatting this pandemic. COVID-19 is caused by a novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As SARS-CoV-2 can be transmitted through droplets and aerosols, we cannot ignore the risk of transmission during hyperbaric oxygen therapy (HBOT). Our hyperbaric oxygen therapy center prioritizes preventing the spread of COVID-19 and maintaining operation for the patients during the pandemic. The aim of this article is to share the protocol that we have adopted in our hyperbaric oxygen therapy center to help prevent the spread of COVID-19.


Asunto(s)
Infecciones por Coronavirus/prevención & control , Oxigenoterapia Hiperbárica/métodos , Pandemias/prevención & control , Neumonía Viral/prevención & control , COVID-19 , Humanos , Taiwán
4.
Int J Mol Sci ; 20(8)2019 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-31010055

RESUMEN

Hyperbaric oxygen treatment (HBOT) has been used to reduce neuropathic pain. Melatonin and opioid receptors are involved in neuropathic pain, but it is not known if HBOT works through these pathways to achieve its antinociceptive effect. We divided anesthetized rats into two treatment and three sham groups. The two treatment groups received third-degree burns on their right hind paws, one treated in a hyperbaric chamber for a week and the other for two weeks. We evaluated the mechanical paw-withdrawal threshold (MWT) and expression of melatonin receptor 1 (MT1), melatonin receptor 2 (MT2), µ (MOR) and κ (KOR) opioid receptor, brain-derived neurotrophic factor (BDNF), Substance P, and calcitonin gene-related peptide (CGRP) in cuneate nucleus, dorsal horn, and hind paw skin by immunohistochemical, immunofluorescence assays and real-time quantitative polymerase chain reaction (RT-PCR). The group receiving one-week HBOT had increased expressions of MT1, MT2, MOR and KOR and decreased expressions of BDNF, Substance P, and CGRP. Their mechanically measured pain levels returned to normal within a week and lasted three weeks. This anti-allodynia effect lasted twice as long in those treated for two weeks. Our findings suggest that increasing the duration of HBOT can reduce burn-induced mechanical allodynia for an extended period of time in rats. The upregulation of melatonin and opioid receptors observed after one week of HBOT suggests they may be partly involved in attenuation of the mechanical allodynia. Downregulation of BDNF, substance P and CGRP may have also contributed to the overall beneficial effect of HBOT.


Asunto(s)
Quemaduras/complicaciones , Oxigenoterapia Hiperbárica , Neuralgia/etiología , Neuralgia/terapia , Animales , Astrocitos/metabolismo , Astrocitos/patología , Conducta Animal , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Masculino , Bulbo Raquídeo/metabolismo , Nocicepción , Ratas Sprague-Dawley , Receptores de Melatonina/metabolismo , Receptores Opioides/metabolismo , Piel/patología , Asta Dorsal de la Médula Espinal/metabolismo , Sustancia P/metabolismo
5.
Int J Mol Sci ; 19(8)2018 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-30060489

RESUMEN

Hyperbaric oxygen (HBO) treatment has been proven to decrease neuroinflammation in rats. This study aimed to determine the potential mechanism underlying the anti-inflammatory effects of HBO treatment on burn-induced neuroinflammation in rats. Thirty-six adult male Sprague-Dawley (SD) rats were randomly assigned to the following six groups (n = 6 per group): (1) sham burn with sham HBO treatment; (2) sham burn with HBO treatment; (3) burn with one-week sham HBO treatment; (4) burn with two-week sham HBO treatment; (5) burn with one-week HBO treatment; and (6) burn with two-week HBO treatment. SD rats that received third-degree burn injury were used as a full-thickness burn injury model. Subsequently, we analyzed the expression of proteins involved in the galectin-3 (Gal-3)-dependent Toll-like receptor-4 (TLR-4) pathway through enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC) analysis, and Western blotting. A behavior test was also conducted, which revealed that HBO treatment significantly suppressed mechanical hypersensitivity in the burn with HBO treatment group compared to the burn with sham HBO treatment group (p < 0.05). ELISA results showed that tumor necrosis factor α (TNF-α) and interleukin 1 beta (IL-1ß) levels in the dorsal horn of the spinal cord and the skin significantly decreased in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). Western blotting results demonstrated that HBO treatment significantly reduced the expression of Gal-3 and TLR-4 in the dorsal horn of the spinal cord in the burn with HBO treatment group compared with the burn with sham HBO treatment group (p < 0.05). IHC analysis showed that the expression of Gal-3, TLR-4, CD68 and CD45 in the dorsal horn of the spinal cord was significantly lower in the burn with HBO treatment group than in the burn with sham HBO treatment group (p < 0.05), and the expression of CD68 and macrophage migration inhibitory factor (MIF) in the right hind paw skin was significantly lower. The expression of vimentin and fibroblast growth factor in the right hind paw skin was significantly higher after HBO treatment (p < 0.05). This study proved that early HBO treatment relieves neuropathic pain, inhibits the Gal-3-dependent TLR-4 pathway, and suppresses microglia and macrophage activation in a rat model.


Asunto(s)
Quemaduras/terapia , Galectina 3/metabolismo , Oxigenoterapia Hiperbárica , Neuralgia/terapia , Receptor Toll-Like 4/efectos de los fármacos , Animales , Escala de Evaluación de la Conducta , Quemaduras/complicaciones , Quemaduras/metabolismo , Miembro Posterior , Interleucina-1beta/análisis , Masculino , Microglía/metabolismo , Neuralgia/etiología , Ratas , Ratas Sprague-Dawley , Asta Dorsal de la Médula Espinal/metabolismo , Factor de Necrosis Tumoral alfa/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA