Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
Más filtros

Medicinas Complementárias
Intervalo de año de publicación
1.
J Photochem Photobiol B ; 239: 112643, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36610350

RESUMEN

Low-level laser therapy, or photobiomodulation, utilizes red or near-infrared light for the treatment of pathological conditions due to the presence of intracellular photoacceptors, such as mitochondrial cytochrome c oxidase, that serve as intermediates for the therapeutic effects. We present an in-detail analysis of the effect of low-intensity LED red light irradiation on the respiratory chain of brain mitochondria. We tested whether low-level laser therapy at 650 nm could alleviate the brain mitochondrial dysfunction in the model of acute hypobaric hypoxia in mice. The irradiation of the mitochondrial fraction of the left cerebral cortex with low-intensity LED red light rescued Complex I-supported respiration during oxidative phosphorylation, normalized the initial polarization of the inner mitochondrial membrane, but has not shown any significant effect on the activity of Complex IV. In comparison, the postponed effect (in 24 h) of the similar transcranial irradiation following hypoxic exposure led to a less pronounced improvement of the mitochondrial functional state, but normalized respiration related to ATP production and membrane polarization. In contrast, the similar irradiation of the mitochondria isolated from control healthy animals exerted an inhibitory effect on CI-supported respiration. The obtained results provide significant insight that can be beneficial for the development of non-invasive phototherapy.


Asunto(s)
Encéfalo , Hipoxia , Terapia por Luz de Baja Intensidad , Mitocondrias , Animales , Ratones , Encéfalo/metabolismo , Encéfalo/efectos de la radiación , Complejo IV de Transporte de Electrones/metabolismo , Hipoxia/complicaciones , Hipoxia/metabolismo , Hipoxia/radioterapia , Rayos Infrarrojos/uso terapéutico , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Presión/efectos adversos , Respiración de la Célula/efectos de la radiación
2.
PLoS One ; 16(12): e0260968, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34860856

RESUMEN

Diabetic retinopathy (DR), the most common complication of diabetes mellitus, is associated with oxidative stress, nuclear factor-κB (NFκB) activation, and excess production of vascular endothelial growth factor (VEGF) and intracellular adhesion molecule-1 (ICAM-1). Muller glial cells, spanning the entirety of the retina, are involved in DR inflammation. Mitigation of DR pathology currently occurs via invasive, frequently ineffective therapies which can cause adverse effects. The application of far-red to near-infrared (NIR) light (630-1000nm) reduces oxidative stress and inflammation in vitro and in vivo. Thus, we hypothesize that 670nm light treatment will diminish oxidative stress preventing downstream inflammatory mechanisms associated with DR initiated by Muller cells. In this study, we used an in vitro model system of rat Müller glial cells grown under normal (5 mM) or high (25 mM) glucose conditions and treated with a 670 nm light emitting diode array (LED) (4.5 J/cm2) or no light (sham) daily. We report that a single 670 nm light treatment diminished reactive oxygen species (ROS) production and preserved mitochondrial integrity in this in vitro model of early DR. Furthermore, treatment for 3 days in culture reduced NFκB activity to levels observed in normal glucose and prevented the subsequent increase in ICAM-1. The ability of 670nm light treatment to prevent early molecular changes in this in vitro high glucose model system suggests light treatment could mitigate early deleterious effects modulating inflammatory signaling and diminishing oxidative stress.


Asunto(s)
Metabolismo Energético , Células Ependimogliales/efectos de la radiación , Glucosa/toxicidad , Rayos Infrarrojos , Mitocondrias/efectos de la radiación , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células Cultivadas , Células Ependimogliales/efectos de los fármacos , Células Ependimogliales/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Ratas , Edulcorantes/toxicidad
3.
Sci Rep ; 11(1): 19114, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34580378

RESUMEN

Bone fracture is a growing public health burden and there is a clinical need for non-invasive therapies to aid in the fracture healing process. Previous studies have demonstrated the utility of electromagnetic (EM) fields in promoting bone repair; however, its underlying mechanism of action is unclear. Interestingly, there is a growing body of literature describing positive effects of an EM field on mitochondria. In our own work, we have previously demonstrated that differentiation of osteoprogenitors into osteoblasts involves activation of mitochondrial oxidative phosphorylation (OxPhos). Therefore, it was reasonable to propose that EM field therapy exerts bone anabolic effects via stimulation of mitochondrial OxPhos. In this study, we show that application of a low intensity constant EM field source on osteogenic cells in vitro resulted in increased mitochondrial membrane potential and respiratory complex I activity and induced osteogenic differentiation. In the presence of mitochondrial inhibitor antimycin A, the osteoinductive effect was reversed, confirming that this effect was mediated via increased OxPhos activity. Using a mouse tibial bone fracture model in vivo, we show that application of a low intensity constant EM field source enhanced fracture repair via improved biomechanical properties and increased callus bone mineralization. Overall, this study provides supporting evidence that EM field therapy promotes bone fracture repair through mitochondrial OxPhos activation.


Asunto(s)
Curación de Fractura/efectos de la radiación , Fracturas Óseas/terapia , Magnetoterapia/métodos , Mitocondrias/efectos de la radiación , Animales , Diferenciación Celular/efectos de la radiación , Línea Celular , Fracturas Óseas/patología , Humanos , Potencial de la Membrana Mitocondrial/efectos de la radiación , Ratones , Mitocondrias/fisiología , Osteoblastos/fisiología , Osteoblastos/efectos de la radiación , Osteogénesis/efectos de la radiación , Fosforilación Oxidativa/efectos de la radiación
4.
Int J Mol Sci ; 22(15)2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34360559

RESUMEN

The tremendous therapeutic potential of photobiomodulation therapy in different branches of medicine has been described in the literature. One of the molecular mechanisms for this treatment implicates the mitochondrial enzyme, cytochrome C oxidase. However, the efficacy and consistency of clinical outcomes with photobiomodulation treatments has been fiercely debated. This work was motivated by this need to improve photobiomodulation devices and delivery approaches. We designed a novel hand-piece with a flat-top beam profile of irradiation. We compared the beam profile versus a standard hand-piece and a fibre probe. We utilized isolated mitochondria and performed treatments at various spots within the beam, namely, the centre, left and right edge. We examined mitochondrial activity by assessing ATP synthesis with the luciferin/luciferase chemiluminescent method as a primary endpoint, while mitochondrial damage was assessed as the secondary endpoint. We observed a uniform distribution of the power density with the flat-top prototype compared to a wide Gaussian beam profile with the standard fibre and standard hand-piece. We noted increased production of ATP in the centre of all three beams with respect to the non-treated controls (p < 0.05). Both the fibre and standard hand-piece demonstrated less increase in ATP synthesis at the edges than the centre (p < 0.05). In contrast, ATP synthesis was increased homogenously in the flat-top handpiece, both in the centre and the edges of the beam. Fibre, standard hand-piece and the flat-top hand-piece prototype have discrete beam distribution characteristics. This significantly affected the mitochondrial activity with respect to their position within the treated areas. Flat-top hand-piece enhances the uniformity of photobiomodulation treatments and can improve the rigour and reproducibility of PBM clinical outcomes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Láseres de Semiconductores/estadística & datos numéricos , Mitocondrias/enzimología , Consumo de Oxígeno , Humanos , Mitocondrias/efectos de la radiación
5.
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
6.
Sci Rep ; 11(1): 13067, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158600

RESUMEN

The plasticity and proliferative capacity of stem cells decrease with aging, compromising their tissue regenerative potential and therapeutic applications. This decline is directly linked to mitochondrial dysfunction. Here, we present an effective strategy to reverse aging of mouse bone marrow mesenchymal stem cells (BM-MSCs) by restoring their mitochondrial functionality using photobiomodulation (PBM) therapy. Following the characterization of young and aged MSCs, our results show that a near-infrared PBM treatment delivering 3 J/cm2 is the most effective modality for improving mitochondrial functionality and aging markers. Furthermore, our results unveil that young and aged MSCs respond differently to the same modality of PBM: whereas the beneficial effect of a single PBM treatment dissipates within 7 h in aged stem cells, it is lasting in young ones. Nevertheless, by applying three consecutive treatments at 24-h intervals, we were able to obtain a lasting rejuvenating effect on aged MSCs. Our findings are of particular significance for improving autologous stem cell transplantation in older individuals who need such therapies most.


Asunto(s)
Senescencia Celular/efectos de la radiación , Terapia por Luz de Baja Intensidad , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de la radiación , Envejecimiento/fisiología , Animales , Biomarcadores/metabolismo , Diferenciación Celular/efectos de la radiación , Linaje de la Célula/efectos de la radiación , Proliferación Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación
7.
Mol Immunol ; 135: 21-27, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33857815

RESUMEN

Ultraviolet A (UVA) irradiation caused skin keratinocytes to accumulate reactive oxygen species (ROS) leading to the skin injury. Thymoquinone (TQ) was identified as the prominent bioactive ingredient in Nigella sativa seeds which was applied in therapying various human diseases. This study aimed to illustrate the role and mechanism of TQ in UVA-induced skin injury. We pre-treated HaCaT cells with TQ and irradiated them by UVA. MTT and Elisa assays were used to evaluate cell viability and apoptosis, as well as cytokine levels. To detect the related parameters of oxidative stress and mitochondrial function, colorimetry, spectrophotometry, bioluminescence, and dual-luciferase reporter methods were used. RT-qPCR and western blotting were performed for expressions of related mRNAs and proteins. TQ significantly improved the UVA-induced cytotoxicity on HaCaT cells. TQ treatment alleviated the oxidative stress and inflammation in UVA-irradiated keratinocytes. Besides, UVA irradiation promoted mitochondrial dysregulation in HaCaT cells leading to cell apoptosis, which could be reversed by TQ treatment. More importantly, NrF2/ARE pathway was activated in TQ-treated cells, while COX-2 was depressed, and inhibiting the pathway or activating COX-2 blocked the therapeutic effect of TQ on UVA-induced skin cell injury. Our study suggested that TQ treatment attenuated the UVA-induced oxidative and inflammatory responses, as well as mitochondrial apoptosis in keratinocytes by COX-2 inhibition via activating NrF2/ARE pathway. This might be a novel sight for preventing the solar radiation damage to the skin.


Asunto(s)
Benzoquinonas/farmacología , Queratinocitos/efectos de la radiación , Mitocondrias/efectos de la radiación , Nigella sativa/metabolismo , Sustancias Protectoras/farmacología , Rayos Ultravioleta/efectos adversos , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Citocinas/metabolismo , Células HaCaT , Humanos , Inflamación/prevención & control , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/efectos de la radiación , Extractos Vegetales/farmacología , Especies Reactivas de Oxígeno/metabolismo , Semillas/metabolismo , Piel/lesiones
8.
Int J Mol Sci ; 22(9)2021 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-33919443

RESUMEN

BACKGROUND: Injury of the trigeminal nerve in oral and maxillofacial surgery can occur. Schwann cell mitochondria are regulators in the development, maintenance and regeneration of peripheral nerve axons. Evidence shows that after the nerve injury, mitochondrial bioenergetic dysfunction occurs and is associated with pain, neuropathy and nerve regeneration deficit. A challenge for research is to individuate new therapies able to normalise mitochondrial and energetic metabolism to aid nerve recovery after damage. Photobiomodulation therapy can be an interesting candidate, because it is a technique involving cell manipulation through the photonic energy of a non-ionising light source (visible and NIR light), which produces a nonthermal therapeutic effect on the stressed tissue. METHODS: The review was based on the following questions: (1) Can photo-biomodulation by red and NIR light affect mitochondrial bioenergetics? (2) Can photobiomodulation support damage to the trigeminal nerve branches? (preclinical and clinical studies), and, if yes, (3) What is the best photobiomodulatory therapy for the recovery of the trigeminal nerve branches? The papers were searched using the PubMed, Scopus and Cochrane databases. This review followed the ARRIVE-2.0, PRISMA and Cochrane RoB-2 guidelines. RESULTS AND CONCLUSIONS: The reliability of photobiomodulatory event strongly bases on biological and physical-chemical evidence. Its principal player is the mitochondrion, whether its cytochromes are directly involved as a photoacceptor or indirectly through a vibrational and energetic variation of bound water: water as the photoacceptor. The 808-nm and 100 J/cm2 (0.07 W; 2.5 W/cm2; pulsed 50 Hz; 27 J per point; 80 s) on rats and 800-nm and 0.2 W/cm2 (0.2 W; 12 J/cm2; 12 J per point; 60 s, CW) on humans resulted as trustworthy therapies, which could be supported by extensive studies.


Asunto(s)
Metabolismo Energético , Terapia por Luz de Baja Intensidad/métodos , Mitocondrias/efectos de la radiación , Regeneración Nerviosa , Recuperación de la Función , Traumatismos del Nervio Trigémino/radioterapia , Animales , Humanos , Traumatismos del Nervio Trigémino/patología
9.
Oxid Med Cell Longev ; 2021: 6626286, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33763170

RESUMEN

Photobiomodulation with 808 nm laser light electively stimulates Complexes III and IV of the mitochondrial respiratory chain, while Complexes I and II are not affected. At the wavelength of 1064 nm, Complexes I, III, and IV are excited, while Complex II and some mitochondrial matrix enzymes seem to be not receptive to photons at that wavelength. Complex IV was also activated by 633 nm. The mechanism of action of wavelengths in the range 900-1000 nm on mitochondria is less understood or not described. Oxidative stress from reactive oxygen species (ROS) generated by mitochondrial activity is an inescapable consequence of aerobic metabolism. The antioxidant enzyme system for ROS scavenging can keep them under control. However, alterations in mitochondrial activity can cause an increment of ROS production. ROS and ATP can play a role in cell death, cell proliferation, and cell cycle arrest. In our work, bovine liver isolated mitochondria were irradiated for 60 sec, in continuous wave mode with 980 nm and powers from 0.1 to 1.4 W (0.1 W increment at every step) to generate energies from 6 to 84 J, fluences from 7.7 to 107.7 J/cm2, power densities from 0.13 to 1.79 W/cm2, and spot size 0.78 cm2. The control was equal to 0 W. The activity of the mitochondria's complexes, Krebs cycle enzymes, ATP production, oxygen consumption, generation of ROS, and oxidative stress were detected. Lower powers (0.1-0.2 W) showed an inhibitory effect; those that were intermediate (0.3-0.7 W) did not display an effect, and the higher powers (0.8-1.1 W) induced an increment of ATP synthesis. Increasing the power (1.2-1.4 W) recovered the ATP production to the control level. The interaction occurred on Complexes III and IV, as well as ATP production and oxygen consumption. Results showed that 0.1 W uncoupled the respiratory chain and induced higher oxidative stress and drastic inhibition of ATP production. Conversely, 0.8 W kept mitochondria coupled and induced an increase of ATP production by increments of Complex III and IV activities. An augmentation of oxidative stress was also observed, probably as a consequence of the increased oxygen consumption and mitochondrial isolation experimental conditions. No effect was observed using 0.5 W, and no effect was observed on the enzymes of the Krebs cycle.


Asunto(s)
Láseres de Semiconductores , Terapia por Luz de Baja Intensidad , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Animales , Bovinos , Respiración de la Célula/efectos de la radiación , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Isocitrato Deshidrogenasa/metabolismo , Peroxidación de Lípido/efectos de la radiación , Malato Deshidrogenasa/metabolismo , Masculino , Fosforilación Oxidativa/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , ATPasas de Translocación de Protón/metabolismo , Superóxidos/metabolismo , Temperatura
10.
J Invest Dermatol ; 141(8): 2056-2066.e10, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33676947

RESUMEN

Near-infrared (NIR) can penetrate the dermis. NIR is able to regulate cutaneous component cells and immune cells and shows significant anti-inflammatory therapeutic effects. However, the mechanisms of these effects are largely unknown. The purpose of this study is to elucidate NIR-induced molecular mechanisms on macrophages because macrophages play initial roles in directing immune responses by their M1 or M2 polarizations. Proteomic analysis revealed that NIR radiation enhanced the expression of mitochondrial respiratory gene citrate synthase. This increased citrate synthase expression was triggered by NIR-induced H3K4 hypermethylation on the citrate synthase gene promoter but not by heat, which led to macrophage M2 polarization and finally resulted in TGFß1 release from CD4+ cells. These cellular effects were validated in human primary macrophages and abdominal NIR-irradiated mouse experiments. In a phorbol 12-myristate 13-acetate‒induced inflammatory model on mouse ear, we confirmed that NIR irradiation induced significant anti-inflammatory effects through decreased M1 counts, reduced TNF-α, and increased CCL22 and/or TGFß1 levels.


Asunto(s)
Dermatitis/terapia , Rayos Infrarrojos/uso terapéutico , Macrófagos/inmunología , Fototerapia/métodos , Animales , Citrato (si)-Sintasa/metabolismo , Dermatitis/inmunología , Dermis/citología , Dermis/inmunología , Dermis/metabolismo , Dermis/efectos de la radiación , Modelos Animales de Enfermedad , Femenino , Humanos , Activación de Macrófagos/efectos de la radiación , Macrófagos/citología , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Ratones , Mitocondrias/enzimología , Mitocondrias/efectos de la radiación , Cultivo Primario de Células , Células THP-1
11.
Int J Med Sci ; 18(1): 109-119, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390779

RESUMEN

Near infrared (NIR) light therapy, or photobiomodulation therapy (PBMT), has gained persistent worldwide attention in recent years as a new novel scientific approach for therapeutic applications in ophthalmology. This ongoing therapeutic adoption of NIR therapy is largely propelled by significant advances in the fields of photobiology and bioenergetics, such as the discovery of photoneuromodulation by cytochrome c oxidase and the elucidation of therapeutic biochemical processes. Upon transcranial delivery, NIR light has been shown to significantly increase cytochrome oxidase and superoxide dismutase activities which suggests its role in inducing metabolic and antioxidant beneficial effects. Furthermore, NIR light may also boost cerebral blood flow and cognitive functions in humans without adverse effects. In this review, we highlight the value of NIR therapy as a novel paradigm for treatment of visual and neurological conditions, and provide scientific evidence to support the use of NIR therapy with emphasis on molecular and cellular mechanisms in eye diseases.


Asunto(s)
Oftalmopatías/terapia , Terapia por Luz de Baja Intensidad/métodos , Apoptosis/efectos de la radiación , Complejo IV de Transporte de Electrones/metabolismo , Oftalmopatías/patología , Humanos , Terapia por Luz de Baja Intensidad/instrumentación , Mitocondrias/metabolismo , Mitocondrias/patología , Mitocondrias/efectos de la radiación , Células Fotorreceptoras de Vertebrados/citología , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/efectos de la radiación , Superóxido Dismutasa/metabolismo , Resultado del Tratamiento
12.
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
13.
Sci Rep ; 10(1): 20382, 2020 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-33230161

RESUMEN

Photobiomodulation (PBM) by far-red (FR) to near-infrared (NIR) light has been demonstrated to restore the function of damaged mitochondria, increase the production of cytoprotective factors and prevent cell death. Our laboratory has shown that FR PBM improves functional and structural outcomes in animal models of retinal injury and retinal degenerative disease. The current study tested the hypothesis that a brief course of NIR (830 nm) PBM would preserve mitochondrial metabolic state and attenuate photoreceptor loss in a model of retinitis pigmentosa, the P23H transgenic rat. P23H rat pups were treated with 830 nm light (180 s; 25 mW/cm2; 4.5 J/cm2) using a light-emitting diode array (Quantum Devices, Barneveld, WI) from postnatal day (p) 10 to p25. Sham-treated rats were restrained, but not treated with 830 nm light. Retinal metabolic state, function and morphology were assessed at p30 by measurement of mitochondrial redox (NADH/FAD) state by 3D optical cryo-imaging, electroretinography (ERG), spectral-domain optical coherence tomography (SD-OCT), and histomorphometry. PBM preserved retinal metabolic state, retinal function, and retinal morphology in PBM-treated animals compared to the sham-treated group. PBM protected against the disruption of the oxidation state of the mitochondrial respiratory chain observed in sham-treated animals. Scotopic ERG responses over a range of flash intensities were significantly greater in PBM-treated rats compared to sham controls. SD-OCT studies and histological assessment showed that PBM preserved the structural integrity of the retina. These findings demonstrate for the first time a direct effect of NIR PBM on retinal mitochondrial redox status in a well-established model of retinal disease. They show that chronic proteotoxic stress disrupts retinal bioenergetics resulting in mitochondrial dysfunction, and retinal degeneration and that therapies normalizing mitochondrial metabolism have considerable potential for the treatment of retinal degenerative disease.


Asunto(s)
Metabolismo Energético/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Mitocondrias/efectos de la radiación , Degeneración Retiniana/radioterapia , Retinitis Pigmentosa/radioterapia , Animales , Modelos Animales de Enfermedad , Electrorretinografía , Flavina-Adenina Dinucleótido/metabolismo , Rayos Infrarrojos , Mitocondrias/metabolismo , NAD/metabolismo , Oxidación-Reducción , Ratas , Ratas Transgénicas , Degeneración Retiniana/diagnóstico por imagen , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Fotorreceptoras Retinianas Bastones/patología , Células Fotorreceptoras Retinianas Bastones/efectos de la radiación , Retinitis Pigmentosa/diagnóstico por imagen , Retinitis Pigmentosa/metabolismo , Retinitis Pigmentosa/patología , Tomografía de Coherencia Óptica , Resultado del Tratamiento
14.
Radiat Res ; 194(3): 236-245, 2020 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-32942301

RESUMEN

Autophagy has been reported to play a radioresistance role in high-dose-rate irradiation. However, its mechanisms and roles in continuous low-dose-rate (CLDR) irradiation have not been clearly understood. Iodine-125 (I-125) seed brachytherapy is a modality of CLDR irradiation and has been used in the treatment of various cancers. In this study, we investigated the mechanisms and roles of autophagy induced by I-125 seed radiation in human esophageal squamous cell carcinoma (ESCC) cell lines (Eca-109 and EC-109) and a xenograft mouse model. The results of this work showed that I-125 seed radiation induced a dose-dependent increase in autophagy in both cell lines. In Eca-109 cells, I-125 seed radiation-induced endoplasmic reticulum (ER) stress, manifesting as the increased levels of intracellular Ca2+ and Grp78/BiP, and activated PERK-eIF2α, IRE1, and ATF6 pathways of the unfolded protein response. Knockdown of PERK led to the decreased expression of autophagy marker, LC3B-II. Inhibition of autophagy by chloroquine or knockdown of ATG5 enhanced I-125 seed radiation-induced cell proliferation inhibition and apoptosis. Interestingly, chloroquine did not aggravate ER stress but promoted apoptosis via the mitochondrial pathway. The animal experiment showed that inhibition of autophagy by chloroquine improved the efficacy of I-125 seed radiation. In summary, our data demonstrate that I-125 seed CLDR radiation induces ER stress-mediated autophagy in ESCC. Autophagy plays a pro-survival role in I-125 seed CLDR irradiation, and chloroquine is a potential candidate for use in combination therapy with I-125 seed radiation treatment to improve efficacy against ESCC.


Asunto(s)
Autofagia/efectos de la radiación , Estrés del Retículo Endoplásmico/efectos de la radiación , Carcinoma de Células Escamosas de Esófago/patología , Carcinoma de Células Escamosas de Esófago/radioterapia , Radioisótopos de Yodo/uso terapéutico , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Chaperón BiP del Retículo Endoplásmico , Humanos , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación
15.
Diabetologia ; 63(9): 1900-1915, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32661752

RESUMEN

AIMS/HYPOTHESIS: Diabetic macular oedema (DME) is the leading cause of visual impairment in people with diabetes. Intravitreal injections of vascular endothelial growth factor inhibitors or corticosteroids prevent loss of vision by reducing DME, but the injections must be given frequently and usually for years. Here we report laboratory and clinical studies on the safety and efficacy of 670 nm photobiomodulation (PBM) for treatment of centre-involving DME. METHODS: The therapeutic effect of PBM delivered via a light-emitting diode (LED) device was tested in transgenic mice in which induced Müller cell disruption led to photoreceptor degeneration and retinal vascular leakage. We also developed a purpose-built 670 nm retinal laser for PBM to treat DME in humans. The effect of laser-delivered PBM on improving mitochondrial function and protecting against oxidative stress was studied in cultured rat Müller cells and its safety was studied in pigmented and non-pigmented rat eyes. We then used the retinal laser to perform PBM in an open-label, dose-escalation Phase IIa clinical trial involving 21 patients with centre-involving DME. Patients received 12 sessions of PBM over 5 weeks for 90 s per treatment at a setting of 25, 100 or 200 mW/cm2 for the three sequential cohorts of 6-8 patients each. Patients were recruited from the Sydney Eye Hospital, over the age of 18 and had centre-involving DME with central macular thickness (CMT) of >300 µm with visual acuity of 75-35 Log minimum angle of resolution (logMAR) letters (Snellen visual acuity equivalent of 20/30-20/200). The objective of this trial was to assess the safety and efficacy of laser-delivered PBM at 2 and 6 months. The primary efficacy outcome was change in CMT at 2 and 6 months. RESULTS: LED-delivered PBM enhanced photoreceptor mitochondrial membrane potential, protected Müller cells and photoreceptors from damage and reduced retinal vascular leakage resulting from induced Müller cell disruption in transgenic mice. PBM delivered via the retinal laser enhanced mitochondrial function and protected against oxidative stress in cultured Müller cells. Laser-delivered PBM did not damage the retina in pigmented rat eyes at 100 mW/cm2. The completed clinical trial found a significant reduction in CMT at 2 months by 59 ± 46 µm (p = 0.03 at 200 mW/cm2) and significant reduction at all three settings at 6 months (25 mW/cm2: 53 ± 24 µm, p = 0.04; 100 mW/cm2: 129 ± 51 µm, p < 0.01; 200 mW/cm2: 114 ± 60 µm, p < 0.01). Laser-delivered PBM was well tolerated in humans at settings up to 200 mW/cm2 with no significant side effects. CONCLUSIONS/INTERPRETATION: PBM results in anatomical improvement of DME over 6 months and may represent a safe and non-invasive treatment. Further testing is warranted in randomised clinical trials. TRIAL REGISTRATION: ClinicalTrials.gov NCT02181400 Graphical abstract.


Asunto(s)
Retinopatía Diabética/radioterapia , Células Ependimogliales/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Edema Macular/radioterapia , Anciano , Animales , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Mitocondrias/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Ratas , Tomografía de Coherencia Óptica
16.
J Nanobiotechnology ; 18(1): 99, 2020 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-32690085

RESUMEN

BACKGROUND: CuS-modified hollow mesoporous organosilica nanoparticles (HMON@CuS) have been preferred as non-invasive treatment for cancer, as near infrared (NIR)-induced photo-thermal effect (PTT) and/or photo-dynamic effect (PDT) could increase cancer cells' apoptosis. However, the certain role of HMON@CuS-produced-PTT&PDT inducing gastric cancer (GC) cells' mitochondrial damage, remained unclear. Moreover, theranostic efficiency of HMON@CuS might be well improved by applying multi-modal imaging, which could offer an optimal therapeutic region and time window. Herein, new nanotheranostics agents were reported by Gd doped HMON decorated by CuS nanocrystals (called HMON@CuS/Gd). RESULTS: HMON@CuS/Gd exhibited appropriate size distribution, good biocompatibility, L-Glutathione (GSH) responsive degradable properties, high photo-thermal conversion efficiency (82.4%) and a simultaneous reactive oxygen species (ROS) generation effect. Meanwhile, HMON@CuS/Gd could efficiently enter GC cells, induce combined mild PTT (43-45 °C) and PDT under mild NIR power density (0.8 W/cm2). Surprisingly, it was found that PTT might not be the only factor of cell apoptosis, as ROS induced by PDT also seemed playing an essential role. The NIR-induced ROS could attack mitochondrial transmembrane potentials (MTPs), then promote mitochondrial reactive oxygen species (mitoROS) production. Meanwhile, mitochondrial damage dramatically changed the expression of anti-apoptotic protein (Bcl-2) and pro-apoptotic protein (Bax). Since that, mitochondrial permeability transition pore (mPTP) was opened, followed by inducing more cytochrome c (Cyto C) releasing from mitochondria into cytosol, and finally activated caspase-9/caspase-3-depended cell apoptosis pathway. Our in vivo data also showed that HMON@CuS/Gd exhibited good fluorescence (FL) imaging (wrapping fluorescent agent), enhanced T1 imaging under magnetic resonance imaging (MRI) and infrared thermal (IRT) imaging capacities. Guided by FL/MRI/IRT trimodal imaging, HMON@CuS/Gd could selectively cause mild photo-therapy at cancer region, efficiently inhibit the growth of GC cells without evident systemic toxicity in vivo. CONCLUSION: HMON@CuS/Gd could serve as a promising multifunctional nanotheranostic platform and as a cancer photo-therapy agent through inducing mitochondrial dysfunction on GC.


Asunto(s)
Mitocondrias , Imagen Multimodal/métodos , Compuestos de Organosilicio , Fototerapia/métodos , Neoplasias Gástricas , Antineoplásicos/química , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sulfato de Cobre , Humanos , Imagen por Resonancia Magnética , Mitocondrias/patología , Mitocondrias/efectos de la radiación , Compuestos de Organosilicio/química , Compuestos de Organosilicio/farmacología , Neoplasias Gástricas/diagnóstico por imagen , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Nanomedicina Teranóstica
17.
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
18.
Cell Mol Life Sci ; 77(14): 2815-2838, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31583425

RESUMEN

Biological effects of high fluence low-power (HFLP) lasers have been reported for some time, yet the molecular mechanisms procuring cellular responses remain obscure. A better understanding of the effects of HFLP lasers on living cells will be instrumental for the development of new experimental and therapeutic strategies. Therefore, we investigated sub-cellular mechanisms involved in the laser interaction with human hepatic cell lines. We show that mitochondria serve as sub-cellular "sensor" and "effector" of laser light non-specific interactions with cells. We demonstrated that despite blue and red laser irradiation results in similar apoptotic death, cellular signaling and kinetic of biochemical responses are distinct. Based on our data, we concluded that blue laser irradiation inhibited cytochrome c oxidase activity in electron transport chain of mitochondria. Contrary, red laser triggered cytochrome c oxidase excessive activation. Moreover, we showed that Bcl-2 protein inhibited laser-induced toxicity by stabilizing mitochondria membrane potential. Thus, cells that either overexpress or have elevated levels of Bcl-2 are protected from laser-induced cytotoxicity. Our findings reveal the mechanism how HFLP laser irradiation interfere with cell homeostasis and underscore that such laser irradiation permits remote control of mitochondrial function in the absence of chemical or biological agents.


Asunto(s)
Complejo IV de Transporte de Electrones/genética , Transporte de Electrón/efectos de la radiación , Terapia por Luz de Baja Intensidad , Fototerapia , Apoptosis/efectos de la radiación , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Transporte de Electrón/genética , Regulación de la Expresión Génica/efectos de la radiación , Células Hep G2 , Humanos , Potencial de la Membrana Mitocondrial/genética , Potencial de la Membrana Mitocondrial/efectos de la radiación , Mitocondrias/genética , Mitocondrias/efectos de la radiación , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/efectos de la radiación , Oxidación-Reducción/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo
19.
J Food Biochem ; 43(2): e12708, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-31353662

RESUMEN

This study investigated the effect of a hawthorn polyphenol extract (HPE) on ultraviolet B (UVB)-induced damage in HaCaT cells and mice. High-performance liquid chromatography/electrospray ionization tandem mass spectrometry was used to analyze the phenolic composition of HPE. The protective effects of HPE and its main components were compared in HaCaT cells. An enzyme-linked immunosorbent assay was used to detect DNA damage (8-hydroxydeoxyguanosine levels). Flow cytometry and western blotting were used to measure the extent of apoptosis and the levels of apoptosis-related proteins, respectively. Treatment with HPE or its polyphenol components inhibited the UVB-induced damage by removing an excess of reactive oxygen species (ROS), reducing DNA damage and p53 activation, regulating the protein expression of B-cell lymphoma 2 family members toward antiapoptotic ratios, and reducing caspase activation. Similar effects were observed in a UVB-irradiated mouse skin, as detected using terminal deoxynucleotidyl transferase dUTP nick-end labeling, immunohistochemistry, and western blotting assays. These results suggest that HPE can be used as a natural dietary supplement for the prevention and treatment of UVB radiation-induced skin damage. PRACTICAL APPLICATIONS: Hawthorn (Crataegus pinnatifida) shows antioxidant, anti-inflammatory, and lipid-lowering effects. As natural, healthy, and effective additives, HPEs have been widely used in food and health products. The results of this study reveal the molecular mechanisms underlying HPE effects, showing that HPE reverses the effects of UVB irradiation via removal of an excess of ROS and reduction of DNA damage and p53 expression in vitro and in vivo. Consequently, HPE upregulates the expression of antiapoptotic BCL-2 and downregulates that of proapoptotic BAX, thereby reducing the activation of caspase-3/9 and inhibiting apoptosis. These findings suggest that HPE can be used as the base ingredient for antiphotoaging food products. This study provides both theoretical and experimental background for hawthorn deep processing and utilization.


Asunto(s)
Crataegus/química , Mitocondrias/metabolismo , Extractos Vegetales/administración & dosificación , Polifenoles/administración & dosificación , Envejecimiento de la Piel/efectos de la radiación , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Femenino , Frutas/química , Humanos , Ratones , Ratones Endogámicos BALB C , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/efectos de la radiación , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Envejecimiento de la Piel/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Rayos Ultravioleta/efectos adversos
20.
Med Eng Phys ; 71: 108-113, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31303375

RESUMEN

The purpose of this study was to investigate the effect of different doses of photobiomodulation (PBM) on mitochondrial respiratory complexes and oxidative cellular energy metabolic enzymes in the mitochondria of brain, muscle, and C6 glioma cells after different time intervals. C6 cells were irradiated with an AlGaInP laser at 10, 30, and 60 J/cm2 for 20, 60, and 120 s, respectively. After irradiation, the cells were maintained in serum-free Dulbecco's Modified Eagle's medium for 24 h, and biochemical measurements were made subsequently. Mitochondrial suspensions from adult rat skeletal muscles/brains were irradiated with an AlGaInP laser at the abovementioned doses. In one group, the reaction was stopped 5 min after irradiation and in the other 60 min after irradiation. Both the C6 cells that received the doses of 10 and 30 J/cm² showed increased complex I activity; the cells that were irradiated at 30 J/cm2 showed increased hexokinase activity. Five minutes after the introduction of PBM of the muscle mitochondria (at 30 and 60 J/cm2), the activity of complex I increased, while the activity of complex IV increased only at 60 J/cm2. One hour after the laser session, complex II activity increased in the cells treated with 10 and 60 J/cm²; however, complex IV activity showed an increase in all PBM groups. In brain mitochondria, 5 min after irradiation only the activity of complex IV increased in all PBM groups. One hour after the laser session, complex II activity increased at 60 J/cm2, and complex IV activity increased for all PBM groups when compared to controls. PBM could increase the activity of respiratory chain complexes in an apparently dose- and time-dependent manner.


Asunto(s)
Astrocitoma/patología , Encéfalo/citología , Terapia por Luz de Baja Intensidad , Mitocondrias/efectos de la radiación , Músculos/citología , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Transporte de Electrón/efectos de la radiación , Humanos , Mitocondrias/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA