Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Neuroinflammation ; 18(1): 256, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34740378

RESUMEN

BACKGROUND: Neurotoxic microglia and astrocytes begin to activate and participate in pathological processes after spinal cord injury (SCI), subsequently causing severe secondary damage and affecting tissue repair. We have previously reported that photobiomodulation (PBM) can promote functional recovery by reducing neuroinflammation after SCI, but little is known about the underlying mechanism. Therefore, we aimed to investigate whether PBM ameliorates neuroinflammation by modulating the activation of microglia and astrocytes after SCI. METHODS: Male Sprague-Dawley rats were randomly divided into three groups: a sham control group, an SCI + vehicle group and an SCI + PBM group. PBM was performed for two consecutive weeks after clip-compression SCI models were established. The activation of neurotoxic microglia and astrocytes, the level of tissue apoptosis, the number of motor neurons and the recovery of motor function were evaluated at different days post-injury (1, 3, 7, 14, and 28 days post-injury, dpi). Lipocalin 2 (Lcn2) and Janus kinase-2 (JAK2)-signal transducer and activator of transcription-3 (STAT3) signaling were regarded as potential targets by which PBM affected neurotoxic microglia and astrocytes. In in vitro experiments, primary microglia and astrocytes were irradiated with PBM and cotreated with cucurbitacin I (a JAK2-STAT3 pathway inhibitor), an adenovirus (shRNA-Lcn2) and recombinant Lcn2 protein. RESULTS: PBM promoted the recovery of motor function, inhibited the activation of neurotoxic microglia and astrocytes, alleviated neuroinflammation and tissue apoptosis, and increased the number of neurons retained after SCI. The upregulation of Lcn2 and the activation of the JAK2-STAT3 pathway after SCI were suppressed by PBM. In vitro experiments also showed that Lcn2 and JAK2-STAT3 were mutually promoted and that PBM interfered with this interaction, inhibiting the activation of microglia and astrocytes. CONCLUSION: Lcn2/JAK2-STAT3 crosstalk is involved in the activation of neurotoxic microglia and astrocytes after SCI, and this process can be suppressed by PBM.


Asunto(s)
Astrocitos/efectos de la radiación , Terapia por Luz de Baja Intensidad , Microglía/efectos de la radiación , Recuperación de la Función/efectos de la radiación , Traumatismos de la Médula Espinal/patología , Animales , Astrocitos/metabolismo , Janus Quinasa 2/metabolismo , Janus Quinasa 2/efectos de la radiación , Lipocalina 2/metabolismo , Lipocalina 2/efectos de la radiación , Masculino , Microglía/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Enfermedades Neuroinflamatorias/patología , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/efectos de la radiación , Transducción de Señal/efectos de la radiación , Traumatismos de la Médula Espinal/metabolismo , Regulación hacia Arriba
2.
PLoS One ; 16(10): e0257896, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34610026

RESUMEN

INTRODUCTION: Peripheral artery disease (PAD) is a highly morbid condition in which impaired blood flow to the limbs leads to pain and tissue loss. Previously we identified 670 nm electromagnetic energy (R/NIR) to increase nitric oxide levels in cells and tissue. NO elicits relaxation of smooth muscle (SMC) by stimulating potassium efflux and membrane hyperpolarization. The actions of energy on ion channel activity have yet to be explored. Here we hypothesized R/NIR stimulates vasodilation through activation of potassium channels in SMC. METHODS: Femoral arteries or facial arteries from C57Bl/6 and Slo1-/- mice were isolated, pressurized to 60 mmHg, pre-constricted with U46619, and irradiated twice with energy R/NIR (10 mW/cm2 for 5 min) with a 10 min dark period between irradiations. Single-channel K+ currents were recorded at room temperature from cell-attached and excised inside-out membrane patches of freshly isolated mouse femoral arterial muscle cells using the patch-clamp technique. RESULTS: R/NIR stimulated vasodilation requires functional activation of the large conductance potassium channels. There is a voltage dependent outward current in SMC with light stimulation, which is due to increases in the open state probability of channel opening. R/NIR modulation of channel opening is eliminated pharmacologically (paxilline) and genetically (BKca α subunit knockout). There is no direct action of light to modulate channel activity as excised patches did not increase the open state probability of channel opening. CONCLUSION: R/NIR vasodilation requires indirect activation of the BKca channel.


Asunto(s)
Radiación Electromagnética , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efectos de la radiación , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/efectos de la radiación , Transducción de Señal/efectos de la radiación , Vasodilatación/efectos de la radiación , Animales , Estimulación Eléctrica/métodos , Terapia por Estimulación Eléctrica/métodos , Arteria Femoral/metabolismo , Técnicas de Inactivación de Genes , Subunidades alfa de los Canales de Potasio de Gran Conductancia Activados por Calcio/genética , Potenciales de la Membrana/efectos de la radiación , Ratones , Ratones Noqueados , Óxido Nítrico/metabolismo , Técnicas de Placa-Clamp , Enfermedad Arterial Periférica/metabolismo , Enfermedad Arterial Periférica/terapia
3.
Cell Death Dis ; 12(11): 1029, 2021 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-34716300

RESUMEN

Breast cancer is a major threat to women's health and estrogen receptor-positive (ER+) breast cancer exhibits the highest incidence among these cancers. As the primary estrogen, estradiol strongly promotes cellular proliferation and radiotherapy, as a standard treatment, exerts an excellent therapeutic effect on ER+ breast cancer. Therefore, we herein wished to explore the mechanism(s) underlying the inhibitory effects of radiation on the proliferation of ER+ breast cancer cells. We used the ER+ breast cancer cell lines MCF7 and T47D, and their complementary tamoxifen-resistant cell lines in our study. The aforementioned cells were irradiated at different doses of X-rays with or without exogenous estradiol. CCK8 and clone-formation assays were used to detect cellular proliferation, enzyme-linked immunosorbent assay (ELISA) to determine estradiol secretion, western immunoblotting analysis and quantitative real-time PCR to evaluate the expression of proteins, and immunofluorescence to track endoplasmic reticulum stress-related processes. Finally, BALB/C tumor-bearing nude mice were irradiated with X-rays to explore the protein expression in tumors using immunohistochemistry. We found that ionizing radiation significantly reduced the phosphorylation of estrogen receptors and the secretion of estradiol by ER+ breast cancer cells. CYP19A (aromatase) is an enzyme located in the endoplasmic reticulum, which plays a critical role in estradiol synthesis (aromatization), and we further demonstrated that ionizing radiation could induce endoplasmic reticulum stress with or without exogenous estradiol supplementation, and that it downregulated the expression of CYP19A through ER-phagy. In addition, ionizing radiation also promoted lysosomal degradation of CYP19A, reduced estradiol synthesis, and inhibited the proliferation of tamoxifen-resistant ER+ breast cancer cells. We concluded that ionizing radiation downregulated the expression of CYP19A and reduced estradiol synthesis by inducing endoplasmic reticulum stress in ER+ breast cancer cells, thereby ultimately inhibiting cellular proliferation.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/radioterapia , Proliferación Celular/efectos de la radiación , Regulación hacia Abajo/efectos de la radiación , Estrés del Retículo Endoplásmico/efectos de la radiación , Estradiol/biosíntesis , Radiación Ionizante , Receptores de Estrógenos/metabolismo , Transducción de Señal/efectos de la radiación , Animales , Aromatasa/metabolismo , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/efectos de la radiación , Estradiol/farmacología , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosforilación/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Tamoxifeno/farmacología , Resultado del Tratamiento , Carga Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Int J Mol Sci ; 22(18)2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34576119

RESUMEN

Although apoptosis of keratinocytes has been relatively well studied, there is a lack of information comparing potentially proapoptotic treatments for healthy and diseased skin cells. Psoriasis is a chronic autoimmune-mediated skin disease manifested by patches of hyperproliferative keratinocytes that do not undergo apoptosis. UVB phototherapy is commonly used to treat psoriasis, although this has undesirable side effects, and is often combined with anti-inflammatory compounds. The aim of this study was to analyze if cannabidiol (CBD), a phytocannabinoid that has anti-inflammatory and antioxidant properties, may modify the proapoptotic effects of UVB irradiation in vitro by influencing apoptotic signaling pathways in donor psoriatic and healthy human keratinocytes obtained from the skin of five volunteers in each group. While CBD alone did not have any major effects on keratinocytes, the UVB treatment activated the extrinsic apoptotic pathway, with enhanced caspase 8 expression in both healthy and psoriatic keratinocytes. However, endoplasmic reticulum (ER) stress, characterized by increased expression of caspase 2, was observed in psoriatic cells after UVB irradiation. Furthermore, decreased p-AKT expression combined with increased 15-d-PGJ2 level and p-p38 expression was observed in psoriatic keratinocytes, which may promote both apoptosis and necrosis. Application of CBD partially attenuated these effects of UVB irradiation both in healthy and psoriatic keratinocytes, reducing the levels of 15-d-PGJ2, p-p38 and caspase 8 while increasing Bcl2 expression. However, CBD increased p-AKT only in UVB-treated healthy cells. Therefore, the reduction of apoptotic signaling pathways by CBD, observed mainly in healthy keratinocytes, suggests the need for further research into the possible beneficial effects of CBD.


Asunto(s)
Apoptosis/efectos de los fármacos , Cannabidiol/farmacología , Queratinocitos/citología , Queratinocitos/efectos de la radiación , Psoriasis/patología , Rayos Ultravioleta , Biomarcadores/metabolismo , Línea Celular , Dinoprostona/farmacología , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Queratinocitos/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Prostaglandina D2/análogos & derivados , Prostaglandina D2/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
5.
Int J Mol Sci ; 22(14)2021 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-34299021

RESUMEN

In this article, we provide an extensive review of the recent literature of the signaling pathways modulated by Pulsed Electromagnetic Fields (PEMFs) and PEMFs clinical application. A review of the literature was performed on two medical electronic databases (PubMed and Embase) from 3 to 5 March 2021. Three authors performed the evaluation of the studies and the data extraction. All studies for this review were selected following these inclusion criteria: studies written in English, studies available in full text and studies published in peer-reviewed journal. Molecular biology, identifying cell membrane receptors and pathways involved in bone healing, and studying PEMFs target of action are giving a solid basis for clinical applications of PEMFs. However, further biology studies and clinical trials with clear and standardized parameters (intensity, frequency, dose, duration, type of coil) are required to clarify the precise dose-response relationship and to understand the real applications in clinical practice of PEMFs.


Asunto(s)
Fracturas Óseas/radioterapia , Magnetoterapia/métodos , Osteogénesis/efectos de la radiación , Transducción de Señal/efectos de la radiación , Células Madre/efectos de la radiación , Bases de Datos Factuales , Campos Electromagnéticos , Humanos , Osteogénesis/genética , Transducción de Señal/genética , Células Madre/metabolismo
6.
Sci Rep ; 11(1): 13371, 2021 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-34183697

RESUMEN

The severity of tissue injury in burn wounds from associated inflammatory and immune sequelae presents a significant clinical management challenge. Among various biophysical wound management approaches, low dose biophotonics treatments, termed Photobiomodulation (PBM) therapy, has gained recent attention. One of the PBM molecular mechanisms of PBM treatments involves photoactivation of latent TGF-ß1 that is capable of promoting tissue healing and regeneration. This work examined the efficacy of PBM treatments in a full-thickness burn wound healing in C57BL/6 mice. We first optimized the PBM protocol by monitoring tissue surface temperature and histology. We noted this dynamic irradiance surface temperature-monitored PBM protocol improved burn wound healing in mice with elevated TGF-ß signaling (phospho-Smad2) and reduced inflammation-associated gene expression. Next, we investigated the roles of individual cell types involved in burn wound healing following PBM treatments and noted discrete effects on epithelieum, fibroblasts, and macrophage functions. These responses appear to be mediated via both TGF-ß dependent and independent signaling pathways. Finally, to investigate specific contributions of TGF-ß1 signaling in these PBM-burn wound healing, we utilized a chimeric TGF-ß1/ß3 knock-in (TGF-ß1Lß3/Lß3) mice. PBM treatments failed to activate the chimeric TGF-ß1Lß3/Lß3 complex and failed to improve burn wound healing in these mice. These results suggest activation of endogenous latent TGF-ß1 following PBM treatments plays a key role in burn wound healing. These mechanistic insights can improve the safety and efficacy of clinical translation of PBM treatments for tissue healing and regeneration.


Asunto(s)
Quemaduras/metabolismo , Quemaduras/radioterapia , Factor de Crecimiento Transformador beta1/metabolismo , Cicatrización de Heridas/efectos de la radiación , Animales , Línea Celular , Inflamación/metabolismo , Inflamación/radioterapia , Terapia por Luz de Baja Intensidad , Masculino , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Transducción de Señal/efectos de la radiación
7.
Anticancer Res ; 41(3): 1407-1420, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33788732

RESUMEN

BACKGROUND/AIM: Recurrence and metastasis of cancer caused by cancer stem cells (CSCs) is a challenge to overcome. Low level laser therapy is a new treatment strategy to suppress their invasiveness. We have assessed the inhibitory effects of 470 nm blue LED on the invasiveness of them to determine the molecular mechanisms of anti-invasiveness. MATERIALS AND METHODS: The effects of blue LEDs on their viability, proliferation and invasion were analyzed using MTT and transwell methods. In addition, the anti-invasiveness effect of blue LED on them was evaluated by zymography, semi-quantitative RT-PCR and western blot analysis. RESULTS: Irradiation with blue LED at 3 J/cm2 resulted in inhibition of their viability, proliferation and invasiveness. Their matrix metalloproteinase 2 (MMP-2) and MMP-9 activities were reduced by blue LED irradiation. Semi-quantitative RT-PCR also showed similar results. In addition, western blotting analyses showed that cyclooxygenase-2 (COX-2) and prostaglandin E2 (PGE2) synthesis were significantly inhibited by LED irradiation in CD133+ colorectal CSCs. CONCLUSION: Down-regulation of the COX-2/PGE2 signaling pathway by blue LED irradiation led to reduce expression of MMP-2 and MMP-9, inhibiting the invasiveness of CD133+ colorectal CSC.


Asunto(s)
Antígeno AC133/metabolismo , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Láseres de Semiconductores , Células Madre Neoplásicas/efectos de la radiación , Transducción de Señal/efectos de la radiación , Antígeno AC133/genética , Proliferación Celular/genética , Proliferación Celular/efectos de la radiación , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Ciclooxigenasa 2/genética , Regulación hacia Abajo/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Invasividad Neoplásica , Células Madre Neoplásicas/metabolismo , Células Tumorales Cultivadas
8.
J Immunol Res ; 2021: 6664453, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33628851

RESUMEN

BACKGROUND: The immune mechanisms underlying low-intensity ultrasound- (LIUS-) mediated suppression of inflammation and tumorigenesis remain poorly determined. METHODS: We used microarray datasets from the NCBI GEO DataSet repository and conducted comprehensive data-mining analyses, where we examined the gene expression of 1376 innate immune regulators (innatome genes (IGs) in cells treated with LIUS. RESULTS: We made the following findings: (1) LIUS upregulates proinflammatory IGs and downregulates metastasis genes in cancer cells, and LIUS upregulates adaptive immunity pathways but inhibits danger-sensing and inflammation pathways and promote tolerogenic differentiation in bone marrow (BM) cells. (2) LIUS upregulates IGs encoded for proteins localized in the cytoplasm, extracellular space, and others, but downregulates IG proteins localized in nuclear and plasma membranes, and LIUS downregulates phosphatases. (3) LIUS-modulated IGs act partially via several important pathways of reactive oxygen species (ROS), reverse signaling of immune checkpoint receptors B7-H4 and BTNL2, inflammatory cytokines, and static or oscillatory shear stress and heat generation, among which ROS is a dominant mechanism. (4) LIUS upregulates trained immunity enzymes in lymphoma cells and downregulates trained immunity enzymes and presumably establishes trained tolerance in BM cells. (5) LIUS modulates chromatin long-range interactions to differentially regulate IGs expression in cancer cells and noncancer cells. CONCLUSIONS: Our analysis suggests novel molecular mechanisms that are utilized by LIUS to induce tumor suppression and inflammation inhibition. Our findings may lead to development of new treatment protocols for cancers and chronic inflammation.


Asunto(s)
Citocinas/metabolismo , Proteínas de Punto de Control Inmunitario/metabolismo , Neoplasias/etiología , Neoplasias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Escape del Tumor/inmunología , Ondas Ultrasónicas , Inmunidad Adaptativa , Células Cultivadas , Perfilación de la Expresión Génica , Humanos , Hipertermia Inducida/métodos , Proteínas de Punto de Control Inmunitario/genética , Inmunidad Innata , Inmunomodulación/efectos de la radiación , Modelos Biológicos , Neoplasias/patología , Neoplasias/terapia , Transducción de Señal/efectos de la radiación
9.
Aging (Albany NY) ; 13(8): 11010-11025, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33535179

RESUMEN

Ultra-violet (UV) radiation (UVR) causes significant oxidative injury to retinal pigment epithelium (RPE) cells. Obacunone is a highly oxygenated triterpenoid limonoid compound with various pharmacological properties. Its potential effect in RPE cells has not been studied thus far. Here in ARPE-19 cells and primary murine RPE cells, obacunone potently inhibited UVR-induced reactive oxygen species accumulation, mitochondrial depolarization, lipid peroxidation and single strand DNA accumulation. UVR-induced RPE cell death and apoptosis were largely alleviated by obacunone. Obacunone activated Nrf2 signaling cascade in RPE cells, causing Keap1-Nrf2 disassociation, Nrf2 protein stabilization and nuclear translocation. It promoted transcription and expression of antioxidant responsive element-dependent genes. Nrf2 silencing or CRISPR/Cas9-induced Nrf2 knockout almost reversed obacunone-induced RPE cytoprotection against UVR. Forced activation of Nrf2 cascade, by Keap1 knockout, similarly protected RPE cells from UVR. Importantly, obacunone failed to offer further RPE cytoprotection against UVR in Keap1-knockout cells. In vivo, intravitreal injection of obacunone largely inhibited light-induced retinal damage. Collectively, obacunone protects RPE cells from UVR-induced oxidative injury through activation of Nrf2 signaling cascade.


Asunto(s)
Benzoxepinas/farmacología , Limoninas/farmacología , Degeneración Macular/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Epitelio Pigmentado de la Retina/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Benzoxepinas/uso terapéutico , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , ADN de Cadena Simple/efectos de los fármacos , ADN de Cadena Simple/efectos de la radiación , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Inactivación de Genes , Humanos , Inyecciones Intravítreas , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Limoninas/uso terapéutico , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/efectos de la radiación , Degeneración Macular/etiología , Degeneración Macular/patología , Ratones , Membranas Mitocondriales/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo/genética , Estrés Oxidativo/efectos de la radiación , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Epitelio Pigmentado de la Retina/citología , Epitelio Pigmentado de la Retina/patología , Epitelio Pigmentado de la Retina/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación
10.
Int J Mol Sci ; 22(3)2021 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-33540902

RESUMEN

Human epidermal keratinocytes are constantly exposed to UV radiation. As a result, there is a significant need for safe and effective compounds to protect skin cells against this environmental damage. This study aimed to analyze the effect of phytocannabinoid-cannabinoid (CBD)-on the proteome of UVA/B irradiated keratinocytes. The keratinocytes were cultured in a three-dimensional (3D) system, designed to mimic epidermal conditions closely. The obtained results indicate that CBD protected against the harmful effects of UVA/B radiation. CBD decreased the expression of proinflammatory proteins, including TNFα/NFκB and IκBKB complex and decreased the expression of proteins involved in de novo protein biosynthesis, which are increased in UVA/B-irradiated cells. Additionally, CBD enhanced the UV-induced expression of 20S proteasome subunits. CBD also protected protein structures from 4-hydroxynonenal (HNE)-binding induced by UV radiation, which primarily affects antioxidant enzymes. CBD-through its antioxidant/anti-inflammatory activity and regulation of protein biosynthesis and degradation-protects skin cells against UVA/B-induced changes. In the future, its long-term use in epidermal cells should be investigated.


Asunto(s)
Cannabidiol/farmacología , Queratinocitos/efectos de los fármacos , Proteoma/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Rayos Ultravioleta , Aldehídos/farmacología , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Cannabidiol/química , Técnicas de Cultivo de Célula , Células Cultivadas , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Quinasa I-kappa B/metabolismo , Queratinocitos/metabolismo , Queratinocitos/efectos de la radiación , Estructura Molecular , Complejos Multiproteicos/metabolismo , FN-kappa B/metabolismo , Análisis de Componente Principal , Proteoma/efectos de la radiación , Transducción de Señal/efectos de la radiación , Factor de Necrosis Tumoral alfa/metabolismo
11.
Lasers Med Sci ; 36(6): 1169-1175, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32996021

RESUMEN

Although it was demonstrated that curcumin-mediated antimicrobial photodynamic therapy (aPDT) is effective for reducing the viability of microbial cells and the vitality of oral biofilms, the cytotoxicity of this therapeutic approach for host cells has not been yet elucidated. Hence, the aim of this study was to evaluate the cytotoxicity and apoptotic effects of curcumin-mediated aPDT on mouse fibroblasts. Cells were treated with 0.6 or 6 µmol.L-1 curcumin combined with 0.075 or 7.5 J.cm-2 LED at 455 nm. Cytotoxicity was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and crystal violet (CV) assays, while quantitative reverse transcriptase-PCR (qRT-PCR) was used to assess the expression of Bax, Bad, Bcl-2, VDAC-1, cytochrome C, and Fas-L genes for apoptosis. The differences between groups were detected by Kruskal-Wallis and post hoc Dunn's tests for MTT and CV assays and by ANOVA and post hoc Tukey test for qRT-PCR (P < 0.05). The effect of 0.6 µmol.L-1 curcumin plus 0.075 J.cm-2 LED (minimum parameter) did not differ statistically from control group; however, the combination of 0.6 µmol.L-1 curcumin plus 7.5 J.cm-2 LED reduced viable cells in 34%, while the combinations of 6 µmol.L-1 curcumin plus 0.075 and 7.5 J.cm-2 LED reduced viable cells in 47% and 99%, respectively. aPDT increased significantly the relative expression of Bax/Bcl-2, cytochrome C, VDAC-1, and Fas-L genes, without influence on the ratio Bad/Bcl-2. Therefore, curcumin-mediated aPDT activated Bcl-2 apoptosis signaling pathways in mouse fibroblasts regarding present conditions, reducing the viability of cells with the increase of curcumin concentrations and light energies.


Asunto(s)
Antiinfecciosos/farmacología , Apoptosis/efectos de los fármacos , Curcumina/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fotoquimioterapia , Transducción de Señal/efectos de los fármacos , Animales , Apoptosis/efectos de la radiación , Fibroblastos/efectos de la radiación , Ratones , Transducción de Señal/efectos de la radiación
12.
J Ethnopharmacol ; 269: 113694, 2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33321189

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ultra Violet (UV) radiation is the major reason for reactive oxygen species (ROS) forming, skin cell damage, melanin production, and could horribly cause skin cancer. Saussureae Involucratae Herba (SIH) is the aerial part of Saussurea involucrata Matsum. & Koidz. This Material Medica is popular with both in Uyghur and Chinese medicines filed. SIH is one of the famous species of the Asteraceae family and which prescribed for skin protection from UV-induced damage according to China Pharmacopeia (2020). However, the detailed working mechanism involved is still not elucidated. AIM OF THE STUDY: We would like to probe the potential transduction pathway of SIH against UV-induced skin cell damages in cultured B16F10 cells. METHODS: Western blot, luciferase assay, laser confocal, RT-PCR and flow cytometer were employed here to verify the protective pharmaceutical value of SIH in cultured B16F10 cells after UV pre-treatment. RESULTS: Our result revealed that SIH attenuates ROS formation after UV-induced damage in B16F10 cells in a dose-dependent manner. Moreover, the transcriptional and translational anti-oxidative encoding genes were up-regulated under the presence of SIH. Further studies showed that SIH activated transcriptional activity of anti-oxidant response element (ARE). Moreover, we found that SIH dramatically stimulates PI3K/Akt phosphorylation in cultured B16F10 cells, this result was further verified by its specific inhibitors, LY294002 and Tocris. CONCLUSION: Our findings concluded that SIH protect melanoma cells from UV damages via activating PI3K/Akt signaling and which could provide scientific evidence for anti-UV pharmaceutical values of this herbal extract.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Extractos Vegetales/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Saussurea , Transducción de Señal/efectos de los fármacos , Rayos Ultravioleta/efectos adversos , Animales , Melanoma Experimental , Ratones , Fosfatidilinositol 3-Quinasas/efectos de la radiación , Extractos Vegetales/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Especies Reactivas de Oxígeno/efectos de la radiación , Transducción de Señal/fisiología , Transducción de Señal/efectos de la radiación
13.
Phytomedicine ; 81: 153424, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33278782

RESUMEN

BACKGROUD: Exposure to high-dose radiation, such as after a nuclear accident or radiotherapy, elicits severe intestinal damage and is associated with a high mortality rate. In treating patients exhibiting radiation-induced intestinal dysfunction, countermeasures to radiation are required. In principle, the cellular event underlying radiation-induced gastrointestinal syndrome is intestinal stem cell (ISC) apoptosis in the crypts. High-dose irradiation induces the loss of ISCs and impairs intestinal barrier function, including epithelial regeneration and integrity. Notch signaling plays a critical role in the maintenance of the intestinal epithelium and regulates ISC self-renewal. Ghrelin, a hormone produced mainly by enteroendocrine cells in the gastrointestinal tract, has diverse physiological and biological functions. PURPOSE: We investigate whether ghrelin mitigates radiation-induced enteropathy, focusing on its role in maintaining epithelial function. METHODS: To investigate the effect of ghrelin in radiation-induced epithelial damage, we analyzed proliferation and Notch signaling in human intestinal epithelial cell. And we performed histological analysis, inflammatory response, barrier functional assays, and expression of notch related gene and epithelial stem cell using a mouse model of radiation-induced enteritis. RESULTS: In this study, we found that ghrelin treatment accelerated the reversal of radiation-induced epithelial damage including barrier dysfunction and defective self-renewing property of ISCs by activating Notch signaling. Exogenous injection of ghrelin also attenuated the severity of radiation-induced intestinal injury in a mouse model. CONCLUSION: These data suggest that ghrelin may be used as a potential therapeutic agent for radiation-induced enteropathy.


Asunto(s)
Ghrelina/farmacología , Enfermedades Intestinales/tratamiento farmacológico , Mucosa Intestinal/citología , Receptores Notch/metabolismo , Células Madre/efectos de la radiación , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Humanos , Enfermedades Intestinales/etiología , Enfermedades Intestinales/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de la radiación , Masculino , Ratones Endogámicos C57BL , Traumatismos por Radiación , Protectores contra Radiación/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Células Madre/efectos de los fármacos , Células Madre/patología , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/efectos de la radiación
14.
Aging (Albany NY) ; 13(2): 2575-2592, 2020 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-33316778

RESUMEN

Radiation therapy is widely used to treat a variety of malignant tumors, including non-small-cell lung cancer (NSCLC). However, ionizing radiation (IR) paradoxically promotes radioresistance, metastasis and recurrence by inducing epithelial-mesenchymal transition (EMT) and cancer stem cells (CSCs). Here, we developed two NSCLC radioresistant (RR) cell lines (A549-RR and H1299-RR) and characterized their motility, cell cycle distribution, DNA damage, and CSC production using migration/invasion assays, flow cytometry, comet assays, and sphere formation, respectively. We also evaluated their tumorigenicity in vivo using a mouse xenograft model. We found that invasion and spheroid formation by A549-RR and H1299-RR cells were increased as compared to their parental cells. Furthermore, as compared to radiation alone, the combination of ß-elemene administration with radiation increased the radiosensitivity of A549 cells and reduced expression of EMT/CSC markers while inhibiting the Prx-1/NF-kB /iNOS signaling pathway. Our findings suggest that NSCLC radioresistance is associated with EMT, enhanced CSC phenotypes, and activation of the Prx-1/NF-kB/iNOS signaling pathway. They also suggest that combining ß-elemene with radiation may be an effective means of overcoming radioresistance in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/terapia , Neoplasias Pulmonares/terapia , Tolerancia a Radiación/efectos de los fármacos , Fármacos Sensibilizantes a Radiaciones/farmacología , Sesquiterpenos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de la radiación , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación
15.
PLoS One ; 15(8): e0236727, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32750068

RESUMEN

Low-power laser irradiation (LPLI) is clinically used to modulate inflammation, proliferation and apoptosis. However, its molecular mechanisms are still not fully understood. This study aimed to describe the effects of LPLI upon inflammatory, apoptotic and proliferation markers in submandibular salivary glands (SMGs) in an experimental model of chronic disorder, 24h after one time irradiation. Diabetes was induced in rats by the injection of streptozotocin. After 29 days, these animals were treated with LPLI in the SMG area, and euthanized 24h after this irradiation. Treatment with LPLI significantly decreased diabetes-induced high mobility group box 1 (HMGB1) and tumor necrosis factor alpha (TNF-α) expression, while enhancing the activation of the transcriptional factor cAMP response element binding (CREB) protein. LPLI also reduced the expression of bax, a mitochondrial apoptotic marker, favoring the cell survival. These findings suggest that LPLI can hamper the state of chronic inflammation and favor homeostasis in diabetic rats SMGs.


Asunto(s)
Diabetes Mellitus Experimental/radioterapia , Terapia por Luz de Baja Intensidad , Transducción de Señal/efectos de la radiación , Glándula Submandibular/efectos de la radiación , Animales , Apoptosis , AMP Cíclico/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Femenino , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Fosforilación , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/metabolismo
16.
Aging (Albany NY) ; 12(13): 13187-13205, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32620714

RESUMEN

Cerebral ischemia/reperfusion (CIR) injury occurs when blood flow is restored in the brain, causing secondary damage to the ischemic tissues. Previous studies have shown that electroacupuncture (EA) treatment contributes to brain protection against CIR injury through modulating autophagy. Studies indicated that SIRT1-FOXO1 plays a crucial role in regulating autophagy. Here we investigated the mechanisms underlying the neuroprotective effect of EA and its role in modulating autophagy via the SIRT1-FOXO1 signaling pathway in rats with CIR injury. EA pretreatment at "Baihui", "Quchi" and "Zusanli" acupoints (2/15Hz, 1mA, 30 min/day) was performed for 5 days before the rats were subjected to middle cerebral artery occlusion, and the results indicated that EA pretreatment substantially reduced the Longa score and infarct volume, increased the dendritic spine density and lessened autophagosomes in the peri-ischemic cortex of rats. Additionally, EA pretreatment also reduced the ratio of LC3-II/LC3-I, the levels of Ac-FOXO1 and Atg7, and the interaction of Ac-FOXO1 and Atg7, but increased the levels of p62, SIRT1, and FOXO1. The above effects were abrogated by the SIRT1 inhibitor EX527. Thus, we presume that EA pretreatment elicits a neuroprotective effect against CIR injury, potentially by suppressing autophagy via activating the SIRT1-FOXO1 signaling pathway.


Asunto(s)
Autofagia/efectos de la radiación , Isquemia Encefálica/metabolismo , Electroacupuntura , Proteínas del Tejido Nervioso/metabolismo , Sirtuina 1/metabolismo , Animales , Autofagosomas/metabolismo , Masculino , Neuroprotección/efectos de la radiación , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de la radiación
17.
J Dermatol Sci ; 98(1): 41-49, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32376153

RESUMEN

BACKGROUND: Endoplasmic reticulum (ER) calcium depletion-induced ER stress is a crucial signal for keratinocyte differentiation and barrier homeostasis, but its effects on the epidermal tight junction (TJ) have not been characterized. Ultraviolet B (UVB) causes ER calcium release in keratinocytes and disrupts epidermal TJ, however, the involvement of ER stress in the UVB-induced TJ alterations remains unknown. OBJECTIVES: To investigate the effect of ER stress by pharmacological ER calcium depletion or UVB on the TJ integrity in normal human epidermal keratinocytes (NHEK). METHODS: NHEK were exposed to ER calcium pump inhibitor thapsigargin (Tg) or UVB. ER stress markers and TJ molecules expression, TJ and F-actin structures, and TJ barrier function were analyzed. RESULTS: Tg or UVB exposure dose-dependently triggered unfolded protein response (UPR) in NHEK. Low dose Tg induced the IRE1α-XBP1 pathway and strengthened TJ barrier. Contrary, high dose Tg activated PERK phosphorylation and disrupted TJ by F-actin disorganization. UVB disrupted TJ and F-actin structures dose dependently. IRE1α RNase inhibition induced or exacerbated TJ and F-actin disruption in the presence of low dose Tg or UVB. High dose Tg increased RhoA activity. 4-PBA or Rho kinase (ROCK) inhibitor partially prevented the disruption of TJ and F-actin following high dose Tg or UVB. CONCLUSIONS: ER stress has bimodal effects on the epidermal TJ depending on its intensity. The IRE1α pathway is critical for the maintenance of TJ integrity during mild ER stress. Severe ER stress-induced UPR or ROCK signalling mediates the disruption of TJ through cytoskeletal disorganization during severe ER stress.


Asunto(s)
Calcio/metabolismo , Estrés del Retículo Endoplásmico/efectos de la radiación , Queratinocitos/patología , Uniones Estrechas/patología , Rayos Ultravioleta/efectos adversos , Amidas/farmacología , Línea Celular , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/patología , Retículo Endoplásmico/efectos de la radiación , Estrés del Retículo Endoplásmico/efectos de los fármacos , Endorribonucleasas/metabolismo , Humanos , Queratinocitos/citología , Queratinocitos/efectos de la radiación , Fenilbutiratos/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Piridinas/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/efectos de la radiación , Respuesta de Proteína Desplegada/efectos de los fármacos , Respuesta de Proteína Desplegada/efectos de la radiación , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
18.
Lasers Med Sci ; 35(9): 1927-1936, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32162133

RESUMEN

Phototherapy has been used to treat postoperative pain and inflammatory response in rheumatoid arthritis. Confidence in this approach, however, is impaired by lack of understanding of the light-triggered cellular and molecular mechanisms. The purpose of this study was to characterize the response of human synoviocyte MH7A cells to visible LED red light in an attempt to elucidate the associated action mechanism. Human synoviocyte MH7A cells were treated with 630-nm LED light after stimulation of tumor necrosis factor-α (TNF-α). The effects of light radiation on cell proliferation and migration were detected by MTT assay and scratch test. The expressions of inflammatory cytokines were measured using RT-qPCR. This was followed by detection of the levels of extracellular proteins IL-6 and IL-8 after differential radiation. Furthermore, the expression levels and activation of proteins on PI3K/AKT/mTOR signaling pathway were examined with Western blot. In terms of the proliferation and migration, repeated radiation with LED red light (630 nm, 26 and 39 J/cm2) exerted an inhibitory effect on synoviocyte MH7A cells. Expression of inflammatory factors (IL-6, IL-1ß, IL-8, and MMP-3) was reduced; meanwhile, the expression of anti-inflammatory factor IL-10 was promoted. At the protein level, treatment with 39 J/cm2 of LED red light could decrease the level of extracellular protein (IL-6 and IL-8) and affect the expression and phosphorylation of proteins on TRPV4/PI3K/AKT/mTOR signaling pathway induced by TNF-α. These results demonstrated that LED red light (630 nm) inhibits proliferation and migration of MH7A cells. The growth-inhibiting effects of LED red light on human synoviocyte MH7A cells appear to be associated with regulation of the TRPV4/PI3K/AKT/mTOR signaling pathway.


Asunto(s)
Terapia por Luz de Baja Intensidad , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Sinoviocitos/efectos de la radiación , Serina-Treonina Quinasas TOR/metabolismo , Canales Catiónicos TRPV/metabolismo , Línea Celular , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Citocinas/metabolismo , Relación Dosis-Respuesta en la Radiación , Regulación de la Expresión Génica/efectos de la radiación , Humanos , Mediadores de Inflamación/metabolismo , Modelos Biológicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal/efectos de la radiación , Sinoviocitos/efectos de los fármacos , Sinoviocitos/metabolismo , Sinoviocitos/patología
19.
J Immunotoxicol ; 17(1): 31-42, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32013650

RESUMEN

In the study here, the potential applicability of KMRC011 - an agonist of toll-like receptor-5 - as a countermeasure for radiation toxicities was evaluated. Following a single 5.5 Gy total body irradiation (TBI, surface absorbed dose = 7 Gy) of Co60 γ-rays, mortality rates and degrees of pathological lesions that developed over 80 days were compared in monkeys that received TBI only and a group that was injected once with KMRC011 (10 µg/kg) after TBI. Compared to the TBI-only hosts (80%), the death rate was significantly improved by the use of KMRC011 (40%), all deaths in both groups occurred in the period from Days 19-24 post-TBI. Further analysis of monkeys that survived until the end of the experiment showed that AST and ALT levels were elevated only in the TBI group, and that radiation-induced tissue damage was alleviated by the KMRC011 injection. Additionally, expression of cell death-related proteins was lower in tissues from the KMRC011-treated hosts than in those in the TBI-only group. Other measured parameters, including body weight, food uptake, and hematological values did not significantly differ between the two groups over the entire period. The results of this study, thus demonstrate that KMRC011 could potentially be used as a medical countermeasure for the treatment of acute radiation exposure.


Asunto(s)
Fragmentos de Péptidos/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Receptor Toll-Like 5/agonistas , Animales , Evaluación Preclínica de Medicamentos , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/efectos de la radiación , Inyecciones Intramusculares , Macaca fascicularis , Masculino , Fragmentos de Péptidos/uso terapéutico , Traumatismos Experimentales por Radiación/inmunología , Protectores contra Radiación/uso terapéutico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Transducción de Señal/efectos de la radiación , Receptor Toll-Like 5/metabolismo , Irradiación Corporal Total
20.
Lasers Surg Med ; 52(4): 358-372, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31321797

RESUMEN

BACKGROUND AND OBJECTIVES: This study was aimed at determining the effects of blue light photobiomodulation on primary adult mouse dermal fibroblasts (AMDFs) and the associated signaling pathways. STUDY DESIGN/MATERIALS AND METHODS: Cultured AMDFs from adult C57BL/6 mice were irradiated by blue light from a light-emitting diode (wavelength = 463 ± 50 nm; irradiance = 5 mW/cm2 ; energy density = 4-8 J/cm2 ). The cells were analyzed using mass spectrometry for proteomics/phosphoproteomics, AlamarBlue assay for mitochondrial activity, time-lapse video for cell migration, quantitative polymerase chain reaction for gene expression, and immunofluorescence for protein expression. RESULTS: Proteomic/phosphoproteomic analysis showed inhibition of extracellular signal-regulated kinases/mammalian target of rapamycin and casein kinase 2 pathways, cell motility-related networks, and multiple metabolic processes, including carbon metabolism, biosynthesis of amino acid, glycolysis/gluconeogenesis, and the pentose phosphate pathway. Functional analysis demonstrated inhibition of mitochondrial activities, cell migration, and mitosis. Expression of growth promoting insulin-like growth factor 1 and fibrosis-related genes, including transforming growth factor ß1 (TGFß1) and collagen type 1 ɑ2 chain diminished. Protein expression of α-smooth muscle actin, an important regulator of myofibroblast functions, was also suppressed. CONCLUSIONS: Low-level blue light exerted suppressive effects on AMDFs, including suppression of mitochondrial activity, metabolism, cell motility, proliferation, TGFß1 levels, and collagen I production. Low-level blue light can be a potential treatment for the prevention and reduction of tissue fibrosis, such as hypertrophic scar and keloids. Lasers Surg. Med. © 2019 Wiley Periodicals, Inc.


Asunto(s)
Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Terapia por Luz de Baja Intensidad , Proteómica , Animales , Adhesión Celular/efectos de la radiación , Técnicas de Cultivo de Célula , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Citocinas/metabolismo , Fibroblastos/patología , Fibrosis/prevención & control , Láseres de Semiconductores , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/efectos de la radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA