Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.044
Filtrar
1.
Oncol Rep ; 52(4)2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39092576

RESUMEN

Senescent cells are known to secrete proteins, including inflammatory cytokines and damage­associated molecular patterns. This phenomenon is known as the senescence­associated secretory phenotype (SASP). SASP in cancer stromal fibroblasts is involved in cancer growth and progression. Conversely, metformin, an antidiabetic drug, has been reported to inhibit SASP induction by inhibiting the activation of NF­κB, a regulator of SASP. To date, at least to the best of our knowledge, there have been no reports regarding cellular senescence in fibroblasts and tumor progression via the SASP­mediated paracrine pathway. The present study thus aimed to elucidate the induction mechanisms of SASP in radiation­induced fibroblasts and to determine its effects on cancer progression via the paracrine pathway. Furthermore, the present study aimed to determine whether controlling SASP using metformin suppresses cancer progression. A well­differentiated esophageal cancer cell line established by the authors' department and fibroblasts isolated and cultured from the non­cancerous esophageal mucosa of resected esophageal cancer cases were used for the experiments. Fibroblasts were irradiated with 8 Gy radiation, and the changes in the expression of the senescence markers, SA­ß­gal, p21, p16 and NF­κB were evaluated using immunofluorescent staining and western blot analysis in the presence or absence of metformin treatment. The culture supernatants of irradiated fibroblasts treated with metformin and those treated without metformin were collected and added to the cancer cells to evaluate their proliferative, invasive and migratory abilities. Vimentin and E­cadherin expression levels were also evaluated using immunofluorescent staining and western blot analysis. The expression levels of p16, p21 and NF­κB in irradiated fibroblasts were attenuated by treatment with metformin. Supernatants collected from irradiated fibroblasts exhibited the proliferative activity of esophageal cancer cells, and the promotion of migratory and invasion abilities, which may be due to epithelial­mesenchymal transition and changes in cell morphology. These reactions were confirmed to be suppressed by the addition of the supernatant of cultured fibroblasts pre­treated with metformin. On the whole, the present study demonstrates that fibroblasts in the cancer stroma may be involved in tumor progression through cellular senescence.


Asunto(s)
Fibroblastos Asociados al Cáncer , Proliferación Celular , Senescencia Celular , Neoplasias Esofágicas , Metformina , Metformina/farmacología , Humanos , Senescencia Celular/efectos de los fármacos , Senescencia Celular/efectos de la radiación , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/efectos de la radiación , Fibroblastos Asociados al Cáncer/patología , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , FN-kappa B/metabolismo , Línea Celular Tumoral , Fenotipo Secretor Asociado a la Senescencia , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de la radiación , Hipoglucemiantes/farmacología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Fibroblastos/efectos de los fármacos
2.
Zhongguo Zhong Yao Za Zhi ; 49(11): 2906-2919, 2024 Jun.
Artículo en Chino | MEDLINE | ID: mdl-39041150

RESUMEN

Rheumatoid arthritis(RA) is a condition in which the joints are in a weakly acidic environment. In RA, RA fibroblastlike synoviocytes( RAFLS) in the joints become abnormally activated and secrete a large amount of matrix metalloproteinases(MMPs), and the receptor protein CD44 on the cell membrane is specifically upregulated. Xuetongsu(XTS), an active ingredient in the Tujia ethnomedicine Xuetong, is known to inhibit the proliferation of RAFLS. However, its development and utilization have been limited due to poor targeting ability. A biomimetic XTS-Prussian blue nanoparticles(PB NPs) drug delivery system called THMPX NPs which can target CD44 was constructed in this study. The surface of THMPX NPs was modified with hyaluronic acid(HA) and a long chain of triglycerol monostearate(TGMS) and 3-aminobenzeneboronic acid(PBA)(PBA-TGMS). The overexpressed MMPs and H+ in inflammatory RAFLS can synergistically cleave the PBA-TGMS on the surface of the nanoparticles, exposing HA to interact with CD44. This allows THMPX NPs to accumulate highly in RAFLS, and upon near-infrared light irradiation, generate heat and release XTS, thereby inhibiting the proliferation and migration of RAFLS. Characterization revealed that THMPX NPs were uniform cubes with a diameter of(190. 3±4. 7) nm and an average potential of(-15. 3± 2. 3) m V. Upon near-infrared light irradiation for 5 min, the temperature of THMPX NPs reached 41. 5 ℃, indicating MMPs and H+-triggered drug release. Safety assessments showed that THMPX NPs had a hemolysis rate of less than 4% and exhibited no cytotoxicity against normal RAW264. 7 and human fibroblast-like synoviocytes(HFLS). In vitro uptake experiments demonstrated the significant targeting ability of THMPX NPs to RAFLS. Free radical scavenging experiments revealed excellent free radical clearance capacity of THMPX NPs, capable of removing reactive oxygen species in RAFLS. Cell counting kit-8 and scratch assays demonstrated that THMPX NPs significantly suppressed the viability and migratory ability of RAFLS. This study provides insights into the development of innovative nanoscale targeted drugs from traditional ethnic medicines for RA treatment.


Asunto(s)
Movimiento Celular , Proliferación Celular , Metaloproteinasas de la Matriz , Nanopartículas , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Nanopartículas/química , Humanos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Metaloproteinasas de la Matriz/metabolismo , Metaloproteinasas de la Matriz/genética , Ferrocianuros/química , Concentración de Iones de Hidrógeno , Sinoviocitos/efectos de los fármacos , Sinoviocitos/efectos de la radiación , Sinoviocitos/metabolismo , Rayos Láser , Receptores de Hialuranos/metabolismo , Receptores de Hialuranos/genética , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/farmacología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/metabolismo
3.
Lasers Med Sci ; 39(1): 180, 2024 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-39001934

RESUMEN

This study investigates the impact of Photobiomodulation (PBM) at different wavelengths on non-superficial cancer cells. Utilizing three laser protocols (650 nm, 810 nm, and 915 nm), the research explores cytotoxic effects, ROS generation, and cell migration. Results reveal varied responses across cell lines, with 810 nm PBM inducing significant ROS levels and inhibiting PAN-1 cell migration. The study suggests potential therapeutic applications for PBM in non-superficial cancers, emphasizing the need for further exploration in clinical settings.


Asunto(s)
Movimiento Celular , Terapia por Luz de Baja Intensidad , Especies Reactivas de Oxígeno , Humanos , Terapia por Luz de Baja Intensidad/métodos , Movimiento Celular/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Neoplasias/radioterapia
4.
Nat Commun ; 15(1): 5512, 2024 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-38951525

RESUMEN

Microglia are important players in surveillance and repair of the brain. Implanting an electrode into the cortex activates microglia, produces an inflammatory cascade, triggers the foreign body response, and opens the blood-brain barrier. These changes can impede intracortical brain-computer interfaces performance. Using two-photon imaging of implanted microelectrodes, we test the hypothesis that low-intensity pulsed ultrasound stimulation can reduce microglia-mediated neuroinflammation following the implantation of microelectrodes. In the first week of treatment, we found that low-intensity pulsed ultrasound stimulation increased microglia migration speed by 128%, enhanced microglia expansion area by 109%, and a reduction in microglial activation by 17%, indicating improved tissue healing and surveillance. Microglial coverage of the microelectrode was reduced by 50% and astrocytic scarring by 36% resulting in an increase in recording performance at chronic time. The data indicate that low-intensity pulsed ultrasound stimulation helps reduce the foreign body response around chronic intracortical microelectrodes.


Asunto(s)
Electrodos Implantados , Microelectrodos , Microglía , Ondas Ultrasónicas , Microglía/efectos de la radiación , Microglía/metabolismo , Animales , Masculino , Reacción a Cuerpo Extraño/prevención & control , Reacción a Cuerpo Extraño/etiología , Ratones , Corteza Cerebral/efectos de la radiación , Corteza Cerebral/citología , Interfaces Cerebro-Computador , Movimiento Celular/efectos de la radiación , Ratas
5.
J Photochem Photobiol B ; 257: 112966, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38970968

RESUMEN

BACKGROUND/AIM: Although photobiomodulation therapy (PBMt) is available to alleviate post-operative side effects of malignant diseases, its application is still controversial due to some potential of cancer recurrence and occurrence of a secondary malignancy. We investigated effect of PBMt on mitochondrial function in HT29 colon cancer cells. METHODS: HT29 cell proliferation was determined with MTT assay after PBMt. Immunofluorescent staining was performed to determine mitochondrial biogenesis and reactive oxygen species (ROS). Mitochondrial membrane potential was measured with Mitotracker. Western blotting was executed to determine expression of fission, fusion, UCP2, and cyclin B1 and D1 proteins. In vivo study was performed by subcutaneously inoculating cancer cells into nude mice and immunohistochemistry was done to determine expression of FIS1, MFN2, UCP2, and p-AKT. RESULTS: The proliferation and migration of HT29 cells reached maximum with PBMt (670 nm, light emitting diode, LED) at 2.0 J/cm2 compared to control (P < 0.05) with more expression of cyclin B1 and cyclin D1 (P < 0.05). Immunofluorescent staining showed that ROS and mitochondrial membrane potential were enhanced after PBMt compared to control. ATP synthesis of mitochondria was also higher in the PBMt group than in the control (P < 0.05). Expression levels of fission and fusion proteins were significantly increased in the PBMt group than in the control (P < 0.05). Electron microscopy revealed that the percentage of mitochondria showing fission was not significantly different between the two groups. Oncometabolites including D-2-hydoxyglutamate in the supernatant of cell culture were higher in the PBMt group than in the control with increased UCP2 expression (P < 0.05). Both tumor size and weight of xenograft in nude mice model were bigger and heavier in the PBMt group than in the control (P < 0.05). Immunohistologically, mitochondrial biogenesis proteins UCP2 and p-AKT in xenograft of nude mice were expressed more in the PBMt group than in the control (P < 0.05). CONCLUSIONS: Treatment with PBM using red light LED may induce proliferation and progression of HT29 cancer cells by increasing mitochondrial activity and fission.


Asunto(s)
Proliferación Celular , Neoplasias del Colon , Potencial de la Membrana Mitocondrial , Ratones Desnudos , Mitocondrias , Especies Reactivas de Oxígeno , Humanos , Células HT29 , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Animales , Proliferación Celular/efectos de la radiación , Ratones , Especies Reactivas de Oxígeno/metabolismo , Neoplasias del Colon/patología , Neoplasias del Colon/radioterapia , Neoplasias del Colon/metabolismo , Potencial de la Membrana Mitocondrial/efectos de la radiación , Terapia por Luz de Baja Intensidad , Movimiento Celular/efectos de la radiación , Ciclina B1/metabolismo , Dinámicas Mitocondriales/efectos de la radiación , Ciclina D1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
6.
Molecules ; 29(14)2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39064938

RESUMEN

Doxorubicin (DOX) has been an effective antitumor agent for human liver cancer cells; however, an overdose might lead to major side effects appearing in clinical applications. In this work, we present a strategy of combining DOX and blue light (BL) irradiation for the antitumor treatment of HepG2 cells (one typical human liver cancer cell line). It is demonstrated that synergetic DOX and BL can significantly reduce cell proliferation and increase the apoptotic rate of HepG2 cells in comparison to individual DOX treatment. The additional BL irradiation is further helpful for enhancing the inhibition of cell migration and invasion. Analyses of reactive oxygen species (ROS) level and Western blotting reveal that the strategy results in more ROS accumulation, mitochondrial damage, and the upregulation of proapoptotic protein (Bcl-2) and downregulation of antiapoptotic protein (Bax). In addition to the improved therapeutic effect, the non-contact BL irradiation is greatly helpful for reducing the dosage of DOX, and subsequently reduces the side effects caused by the DOX drug. These findings offer a novel perspective for the therapeutic approach toward liver cancer with high efficiency and reduced side effects.


Asunto(s)
Apoptosis , Movimiento Celular , Proliferación Celular , Doxorrubicina , Luz , Neoplasias Hepáticas , Especies Reactivas de Oxígeno , Doxorrubicina/farmacología , Humanos , Células Hep G2 , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/radioterapia , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Luz Azul
7.
Int J Mol Sci ; 25(12)2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38928220

RESUMEN

We hypothesize that the injection of JP4-039, a mitochondria-targeted nitroxide, prior to irradiation of the mouse retina may decrease apoptosis and reduce neutrophil and macrophage migration into the retina. In our study, we aimed to examine the effects of JP4-039 in the mouse retina using fluorescent microscopy, a terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, and flow cytometry. Forty-five mice and one eye per mouse were used. In Group 1, fluorescent microscopy was used to determine retinal uptake of 10 µL (0.004 mg/µL) of intravitreally injected BODIPY-labeled JP4-039 at 0, 15, and 60 min after injection. In Group 2, the TUNEL assay was performed to investigate the rate of apoptosis after irradiation in addition to JP4-039 injection, compared to controls. In Group 3, flow cytometry was used to determine the extent of inflammatory cell migration into the retina after irradiation in addition to JP4-039 injection, compared to controls. Maximal retinal uptake of JP4-039 was 15 min after intravitreal injection (p < 0.0001). JP4-039-treated eyes had lower levels of retinal apoptosis (35.8 ± 2.5%) than irradiated controls (49.0 ± 2.7%; p = 0.0066) and demonstrated reduced migration of N1 cells (30.7 ± 11.7% vs. 77.7 ± 5.3% controls; p = 0.004) and M1 cells (76.6 ± 4.2 vs. 88.1 ± 3.7% controls, p = 0.04). Pretreatment with intravitreally injected JP4-039 reduced apoptosis and inflammatory cell migration in the irradiated mouse retina, marking the first confirmed effect of this molecule in retinal tissue. Further studies may allow for safety profiling and potential use for patients with radiation retinopathy.


Asunto(s)
Apoptosis , Movimiento Celular , Mitocondrias , Retina , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Ratones , Retina/efectos de los fármacos , Retina/metabolismo , Retina/efectos de la radiación , Retina/patología , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Ratones Endogámicos C57BL , Masculino , Óxidos de Nitrógeno/farmacología , Inflamación/patología
8.
J Photochem Photobiol B ; 257: 112963, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38908147

RESUMEN

The therapeutic potential of blue light photobiomodulation in cancer treatment, particularly in inhibiting cell proliferation and promoting cell death, has attracted significant interest. Oral squamous cell carcinoma (OSCC) is a prevalent form of oral cancer, necessitating innovative treatment approaches to improve patient outcomes. In this study, we investigated the effects of 420 nm blue LED light on OSCC and explored the underlying mechanisms. Our results demonstrated that 420 nm blue light effectively reduced OSCC cell viability and migration, and induced G2/M arrest. Moreover, we observed that 420 nm blue light triggered endoplasmic reticulum (ER) stress and mitochondrial dysfunction in OSCC cells, leading to activation of the CHOP signal pathway and alterations in the levels of Bcl-2 and Bax proteins, ultimately promoting cell apoptosis. Additionally, blue light suppressed mitochondrial gene expression, likely due to its damage to mitochondrial DNA. This study highlights the distinct impact of 420 nm blue light on OSCC cells, providing valuable insights into its potential application as a clinical treatment for oral cancer.


Asunto(s)
Apoptosis , Carcinoma de Células Escamosas , Supervivencia Celular , Estrés del Retículo Endoplásmico , Luz , Mitocondrias , Neoplasias de la Boca , Humanos , Estrés del Retículo Endoplásmico/efectos de la radiación , Mitocondrias/efectos de la radiación , Mitocondrias/metabolismo , Neoplasias de la Boca/radioterapia , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Línea Celular Tumoral , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Movimiento Celular/efectos de la radiación , Transducción de Señal/efectos de la radiación , Proteína X Asociada a bcl-2/metabolismo , Proteína X Asociada a bcl-2/genética , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Luz Azul
9.
Biochim Biophys Acta Gen Subj ; 1868(9): 130650, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38830560

RESUMEN

Glioblastoma (GBM), a highly aggressive malignant tumor of the central nervous system, is mainly treated with radiotherapy. However, since irradiation may lead to the acquisition of migration ability by cancer cells, thereby promoting tumor metastasis and invasion, it is important to understand the mechanism of cell migration enhancement in order to prevent recurrence of GBM. The receptor for advanced glycation end products (RAGE) is a pattern recognition receptor activated by high mobility group box 1 (HMGB1). In this study, we found that RAGE plays a role in the enhancement of cell migration by γ-irradiation in human GBM A172 cells. γ-Irradiation induced actin remodeling, a marker of motility acquisition, and enhancement of cell migration in A172 cells. Both phenotypes were suppressed by specific inhibitors of RAGE (FPS-ZM1 and TTP488) or by knockdown of RAGE. The HMGB1 inhibitor ethyl pyruvate similarly suppressed γ-irradiation-induced enhancement of cell migration. In addition, γ-irradiation-induced phosphorylation of STAT3 was suppressed by RAGE inhibitors, and a STAT3 inhibitor suppressed γ-irradiation-induced enhancement of cell migration, indicating that STAT3 is involved in the migration enhancement downstream of RAGE. Our results suggest that HMGB1-RAGE-STAT3 signaling is involved in radiation-induced enhancement of GBM cell migration, and may contribute to GBM recurrence by promoting metastasis and invasion.


Asunto(s)
Movimiento Celular , Rayos gamma , Glioblastoma , Proteína HMGB1 , Fenotipo , Receptor para Productos Finales de Glicación Avanzada , Factor de Transcripción STAT3 , Humanos , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/radioterapia , Movimiento Celular/efectos de la radiación , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteína HMGB1/metabolismo , Línea Celular Tumoral , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Fosforilación/efectos de la radiación , Piruvatos/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/radioterapia , Benzamidas
10.
J Photochem Photobiol B ; 257: 112957, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38941921

RESUMEN

Phototherapy has been extensively used to prevent and treat signs of aging and stimulate wound healing, and phototherapy through light-emitting diodes (LEDs). In contrast to LED, organic LED (OLED) devices are composed of organic semiconductors that possess novel characteristics. We investigated the regenerative potential of OLED for restoring cellular potential from senescence and thus delaying animal aging. Bone marrow-derived stem cells (BMSCs) and adipose-derived stem cells (ADSCs) were isolated from the control and OLED- treated groups to evaluate their proliferation, migration, and differentiation potentials. Cellular senescence was evaluated using a senescence-associated ß-galactosidase (SA-ß-gal) activity assay and gene expression biomarker assessment. OLED treatment significantly increased the cell proliferation, colony formation, and migration abilities of stem cells. SA-ß-gal activity was significantly decreased in both ADSCs and BMSCs in the OLED-treated group. Gene expression biomarkers from treated mice indicated a significant upregulation of IGF-1 (insulin growthfactor-1). The upregulation of the SIRT1 gene inhibited the p16 and p19 genes then to downregulate the p53 expressions for regeneration of stem cells in the OLED-treated group. Our findings indicated that the survival rates of 10-month aging senescence-accelerated mouse prone 8 mice were prolonged and that their gross appearance improved markedly after OLED treatment. Histological analysis of skin and brain tissue also indicated significantly greater collagen fibers density, which prevents ocular abnormalities and ß-amyloid accumulation. Lordokyphosis and bone characteristics were observed to resemble those of younger mice after OLED treatment. In conclusion, OLED therapy reduced the signs of aging and enhanced stem-cell senescence recovery and then could be used for tissue regeneration.


Asunto(s)
Senescencia Celular , Sirtuina 1 , Regulación hacia Arriba , Animales , Sirtuina 1/metabolismo , Sirtuina 1/genética , Ratones , Regulación hacia Arriba/efectos de la radiación , Senescencia Celular/efectos de la radiación , Longevidad/efectos de la radiación , Proliferación Celular/efectos de la radiación , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/genética , Diferenciación Celular/efectos de la radiación , Movimiento Celular/efectos de la radiación , Envejecimiento , Células Madre/citología , Células Madre/metabolismo , Células Madre/efectos de la radiación , beta-Galactosidasa/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Tejido Adiposo/citología
11.
PLoS Negl Trop Dis ; 18(5): e0012227, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38814992

RESUMEN

BACKGROUND: Photobiomodulation has exhibited promise in mitigating the local effects induced by Bothrops snakebite envenoming; however, the mechanisms underlying this protection are not yet fully understood. Herein, the effectiveness of photobiomodulation effects on regenerative response of C2C12 myoblast cells following exposure to Bothrops jararacussu venom (BjsuV), as well as the mechanisms involved was investigated. METHODOLOGY/PRINCIPAL FINDINGS: C2C12 myoblast cells were exposed to BjsuV (12.5 µg/mL) and irradiated once for 10 seconds with laser light of 660 nm (14.08 mW; 0.04 cm2; 352 mW/cm2) or 780 nm (17.6 mW; 0.04 cm2; 440 mW/ cm2) to provide energy densities of 3.52 and 4.4 J/cm2, and total energies of 0.1408 and 0.176 J, respectively. Cell migration was assessed through a wound-healing assay. The expression of MAPK p38-α, NF-Кß, Myf5, Pax-7, MyoD, and myogenin proteins were assessed by western blotting analysis. In addition, interleukin IL1-ß, IL-6, TNF-alfa and IL-10 levels were measured in the supernatant by ELISA. The PBM applied to C2C12 cells exposed to BjsuV promoted cell migration, increase the expression of myogenic factors (Pax7, MyF5, MyoD and myogenin), reduced the levels of proinflammatory cytokines, IL1-ß, IL-6, TNF-alfa, and increased the levels of anti-inflammatory cytokine IL-10. In addition, PBM downregulates the expression of NF-kB, and had no effect on p38 MAKP. CONCLUSION/SIGNIFICANCE: These data demonstrated that protection of the muscle cell by PBM seems to be related to the increase of myogenic factors as well as the modulation of inflammatory mediators. PBM therapy may offer a new therapeutic strategy to address the local effects of snakebite envenoming by promoting muscle regeneration and reducing the inflammatory process.


Asunto(s)
Bothrops , Venenos de Crotálidos , Citocinas , Terapia por Luz de Baja Intensidad , Mioblastos , Miogenina , Animales , Mioblastos/efectos de los fármacos , Mioblastos/efectos de la radiación , Mioblastos/metabolismo , Ratones , Terapia por Luz de Baja Intensidad/métodos , Citocinas/metabolismo , Línea Celular , Venenos de Crotálidos/toxicidad , Miogenina/metabolismo , Miogenina/genética , Factor de Transcripción PAX7/metabolismo , Factor de Transcripción PAX7/genética , FN-kappa B/metabolismo , Proteína MioD/metabolismo , Proteína MioD/genética , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Factor 5 Regulador Miogénico/metabolismo , Factor 5 Regulador Miogénico/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Mordeduras de Serpientes/radioterapia , Serpientes Venenosas
12.
J Photochem Photobiol B ; 256: 112940, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38776590

RESUMEN

Photobiomodulation (PBM) is a well-established medical technology that employs diverse light sources like lasers or light-emitting diodes to generate diverse photochemical and photophysical reactions in cells, thereby producing beneficial clinical outcomes. In this study, we introduced an 830 nm near-infrared (NIR) laser irradiation system combined with a microscope objective to precisely and controllably investigate the impact of PBM on the migration and viability of human adipose mesenchymal stem cells (hADSCs). We observed a biphasic dose-response in hADSCs' viability and migration after PBM exposure (0-10 J/cm2), with the 5 J/cm2 group showing significantly higher cell viability and migration ability than other groups. Additionally, at the optimal dose of 5 J/cm2, we used nanoparticle tracking analysis (NTA) and found a 6.25-fold increase in the concentration of extracellular vesicles (EVs) derived from hADSCs (PBM/ADSC-EVs) compared to untreated cells (ADSC-EVs). Both PBM/ADSC-EVs and ADSC-EVs remained the same size, with an average diameter of 56 nm measured by the ExoView R200 system, which falls within the typical size range for exosomes. These findings demonstrate that PBM not only improves the viability and migration of hADSCs but also significantly increases the EV yield.


Asunto(s)
Movimiento Celular , Supervivencia Celular , Vesículas Extracelulares , Células Madre Mesenquimatosas , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Movimiento Celular/efectos de la radiación , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/efectos de la radiación , Tejido Adiposo/citología , Tejido Adiposo/efectos de la radiación , Terapia por Luz de Baja Intensidad , Relación Dosis-Respuesta en la Radiación , Células Cultivadas , Rayos Infrarrojos
13.
BMC Cancer ; 24(1): 664, 2024 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-38822331

RESUMEN

Recent studies have shown that blue light-emitting diode (LED) light has anti-tumor effects, suggesting the possibility of using visible light in cancer therapy. However, the effects of blue light irradiation on cells in the tumor microenvironment, including tumor-associated macrophages (TAMs), are unknown. Here, THP-1 cells were cultured in the conditioned medium (CM) of HCT-116 cells to prepare TAMs. TAMs were divided into LED-irradiated and control groups. Then, the effects of blue LED irradiation on TAM activation were examined. Expression levels of M2 macrophage markers CD163 and CD206 expression were significantly decreased in LED-irradiated TAMs compared with the control group. While control TAM-CM could induce HCT-116 cell migration, these effects were not observed in cells cultured in TAM-CM with LED irradiation. Vascular endothelial growth factor (VEGF) secretion was significantly suppressed in LED-exposed TAMs. PD-L1 expression was upregulated in HCT-116 cells cultured with TAM-CM but attenuated in cells cultured with LED-irradiated TAM-CM. In an in vivo model, protein expression levels of F4/80 and CD163, which are TAM markers, were reduced in the LED-exposed group. These results indicate that blue LED light may have an inhibitory effect on TAMs, as well as anti-tumor effects on colon cancer cells.


Asunto(s)
Luz Azul , Neoplasias del Colon , Macrófagos Asociados a Tumores , Animales , Humanos , Ratones , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Movimiento Celular/efectos de la radiación , Neoplasias del Colon/radioterapia , Medios de Cultivo Condicionados/farmacología , Células HCT116 , Activación de Macrófagos/efectos de la radiación , Macrófagos/metabolismo , Macrófagos/efectos de la radiación , Fototerapia/métodos , Receptores de Superficie Celular/metabolismo , Microambiente Tumoral/efectos de la radiación , Macrófagos Asociados a Tumores/metabolismo , Macrófagos Asociados a Tumores/efectos de la radiación , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Radiat Res ; 202(1): 87-95, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38720240

RESUMEN

Radiotherapy with cell cycle-specific anticancer agents has become an important option in the control of both primary tumors and metastases. Here, we used image analysis algorithms that enable quick segmentation and tracking to describe a radiobiological approach for the optimized selection of cell cycle-targeting anticancer drugs for radiotherapy. We confirmed cell cycle-synchronization using human cervical cancer HeLa cells expressing a fluorescent ubiquitination-based cell cycle indicator (FUCCI) as a cell cycle-monitoring probe. Cells synchronized in the G1 and G2 phases were irradiated with X rays at 0.5-2 Gy. Each cell was identified using Cellpose, a deep learning-based algorithm for cellular segmentation, and the velocity and direction of migration were analyzed using the TrackMate plugin in Fiji ImageJ. G1 phase synchronized cells showed a dose-dependent decrease in velocity after irradiation, while G2 cells tended to increase their velocity. The migration pattern of all cells appeared to be a random walk model, regardless of the exposure dose. In addition, we used cisplatin to arrest the cell cycle. HeLa-FUCCI cells arrested at the G2 phase via cisplatin treatment showed enhanced cell migration after X-ray exposure. These results indicated that anticancer agents that arrest the cell cycle of cancer cells in a specific phase may enhance cell migration after radiotherapy. Our approach, using cellular segmentation and tracking algorithms, could enhance the radiobiological assessment of cell cycle-specific migration after irradiation to aid in optimizing radiotherapy using cell cycle-targeting agents.


Asunto(s)
Ciclo Celular , Movimiento Celular , Humanos , Movimiento Celular/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Células HeLa , Rayos X , Ciclo Celular/efectos de la radiación , Ciclo Celular/efectos de los fármacos , Antineoplásicos/farmacología , Radiobiología/métodos , Cisplatino/farmacología
15.
Br J Cancer ; 131(1): 37-48, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38740970

RESUMEN

BACKGROUND: Cancer cells in severely hypoxic regions have been reported to invade towards tumour blood vessels after surviving radiotherapy in a postirradiation reoxygenation- and hypoxia-inducible factor (HIF)-dependent manner and cause recurrence. However, how HIF induces invasiveness of irradiated and reoxygenated cancer cells remains unclear. METHODS: Here, we identified human minor histocompatibility antigen 1 (HMHA1), which has been suggested to function in cytoskeleton dynamics and cellular motility, as a responsible factor and elucidated its mechanism of action using molecular and cellular biology techniques. RESULTS: HMHA1 expression was found to be induced at the transcription initiation level in a HIF-dependent manner under hypoxia. Boyden chamber invasion assay revealed that the induction of HMHA1 expression is required for the increase in invasion of hypoxic cancer cells. Reoxygenation treatment after ionising radiation in vitro that mimics dynamic changes of a microenvironment in hypoxic regions of tumour tissues after radiation therapy further enhanced HMHA1 expression and invasive potential of HMHA1 wildtype cancer cells in ROS- and HIF-dependent manners, but not of HMHA1 knockout cells. CONCLUSION: These results together provide insights into a potential molecular mechanism of the acquisition of invasiveness by hypoxic cancer cells after radiotherapy via the activation of the ROS/HIF/HMHA1 axis.


Asunto(s)
Invasividad Neoplásica , Humanos , Hipoxia de la Célula , Línea Celular Tumoral , Movimiento Celular/efectos de la radiación , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo
16.
J Photochem Photobiol B ; 255: 112907, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677259

RESUMEN

OBJECTIVE: The objective of this study is to investigate the variances in transcriptome gene expression of normal oral mucosa-derived mesenchymal stem cell (OM-MSC), oral leukoplakia-derived MSC (OLK-MSC) and oral squamous cell carcinoma-derived MSC(OSCC-MSC). as Additionally, the study aims to compare the in vitro proliferation, migration, invasion ability, and response to photodynamic therapy (PDT) of these three MSC, HOK, DOK, leuk1, and Cal27 cell lines. METHODS: HOK, DOK, leuk1, Cal27 cells were cultured in vitro. 3 MSC cells were obtained from OM, OLK, OSCC tissue (n = 3) and identified through flow cytometry. They were also cultured in vitro for osteogenic and lipogenic-induced differentiation. Based on the Illumina HiSeq high-throughput sequencing platform, OM-MSC, OLK-MSC, OSCC-MSC (n = 3) were subjected to transcriptome sequencing, functional annotation, and enrichment analysis of differentially expressed genes and related genes. CCK8 assay, wound healing assay, and transwell assay were performed to compare the proliferation, migration, and invasion of the seven types of cells. The 7 cells were incubated with 0, 0.125 mM, 0.25 mM, 0.5 mM, 1 mM, and 2 mM of the photosensitizer (5-aminolevulinic acid, 5-ALA) in vitro. Subsequently, they were irradiated with a 150 mM, 635 nm laser for 1 min, and the cell activity was detected using the CCK8 assay after 24 h. The mitochondrial changes in the 7 cells before and after the treatment of PDT were detected using the JC-10 probe, and the changes in ATP content were measured before and after the PDT treatment. RESULTS: OM-MSC, OLK-MSC, and OSCC-MSC expressed positive MSC surface markers. After osteogenic and lipogenic-induced differentiation culture, stained calcium nodules and lipid droplets were visible, meeting the identification criteria of MSC. Pathway enrichment analysis revealed that the differentially expressed genes (DEGs) of OSCC-MSC compared to OLK-MSC were primarily associated with the PI3K-Akt signaling pathway and tumor-related pathways. OSCC-MSC exhibited stronger migratory and invasive abilities compared to Cal27. The IC50 values required for OM, OLK, and OSCC-derived MSC were lower than those required for epithelial cells treated with PDT, which were 1.396 mM, 0.9063 mM, and 2.924 mM, respectively. Cell membrane and mitochondrial disruption were observed in seven types of cells after 24 h of PDT treatment. However, HOK, DOK, leuk1, and Cal27 cells had an ATP content increased. CONCLUSIONS: OLK, OSCC epithelial cells require higher concentrations of 5-ALA for PDT treatment than MSC of the same tissue origin. The concentration of 5-ALA required increases with increasing cell malignancy. Differences in the response of epithelial cells and MSC to PDT treatment may have varying impacts on OLK recurrence and malignancy.


Asunto(s)
Carcinoma de Células Escamosas , Movimiento Celular , Proliferación Celular , Células Epiteliales , Leucoplasia Bucal , Células Madre Mesenquimatosas , Mucosa Bucal , Neoplasias de la Boca , Fotoquimioterapia , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Mucosa Bucal/patología , Mucosa Bucal/citología , Leucoplasia Bucal/patología , Leucoplasia Bucal/terapia , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/metabolismo , Neoplasias de la Boca/patología , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/terapia , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Fármacos Fotosensibilizantes/farmacología , Línea Celular Tumoral , Ácido Aminolevulínico/farmacología , Diferenciación Celular/efectos de los fármacos , Transcriptoma/efectos de los fármacos
17.
J Radiat Res ; 65(3): 303-314, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38637316

RESUMEN

Angiosarcoma is a rare refractory soft-tissue tumor with a poor prognosis and is treated by radiotherapy. The fibroblast growth factor 1 (FGF1) mutant, with enhanced thermostability due to several substituted amino acids, inhibits angiosarcoma cell metastasis, yet the mechanism of action is unclear. This study aims to clarify the FGF1 mutant mechanism of action using ISOS-1 mouse angiosarcoma cells. The wild-type FGF1 or FGF1 mutant was added to ISOS-1 cells and cultured, evaluating cell numbers over time. The invasive and migratory capacity of ISOS-1 cells was assessed by transwell analysis. ISOS-1 cell radiosensitivity was assessed by colony formation assay after X-ray irradiation. To examine whether mitogen-activated protein kinase (MEK) inhibitor counteracts the FGF1 mutant effects, a combination of MEK inhibitor and FGF1 mutant was added to ISOS-1 cells and cultured. The FGF1 mutant was observed to inhibit ISOS-1 cell proliferation, invasion and migration by sustained FGF1 signaling activation. A MEK inhibitor suppressed the FGF1 mutant-induced inhibition of proliferation, invasion and migration of ISOS-1 cells. Furthermore, the FGF1 mutant enhanced radiosensitivity of ISOS-1 cells, but MEK inhibition suppressed the increased radiosensitivity. In addition, we found that the FGF1 mutant strongly inhibits actin polymerization, suggesting that actin cytoskeletal dynamics are closely related to ISOS-1 cell radiosensitivity. Overall, this study demonstrated that in ISOS-1 cells, the FGF1 mutant inhibits proliferation, invasion and migration while enhancing radiosensitivity through sustained activation of the MEK-mediated signaling pathway.


Asunto(s)
Movimiento Celular , Proliferación Celular , Factor 1 de Crecimiento de Fibroblastos , Hemangiosarcoma , Sistema de Señalización de MAP Quinasas , Invasividad Neoplásica , Tolerancia a Radiación , Animales , Ratones , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Tolerancia a Radiación/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Línea Celular Tumoral , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Hemangiosarcoma/patología , Hemangiosarcoma/metabolismo , Hemangiosarcoma/radioterapia
18.
Pathol Res Pract ; 257: 155293, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38615508

RESUMEN

Lysophosphatidic acid (LPA) binds to its specific G protein-coupled LPA receptors (LPA1 to LPA6), resulting in the activation of various cellular functions. LPA receptor-mediated signaling facilitates tumor progression in human malignancies. In the present study, we investigated whether LPA receptor-mediated signaling contributes to cellular responses to X-ray irradiation in osteosarcoma MG-63 cells. After X-ray irradiation (2, 4 and 8 Gy), LPAR2 and LPAR3 expression levels in MG-63 cells were significantly elevated in a dose-dependent manner, but no change of LPAR1 expression level was observed. The cell growth activities of MG-63 cells irradiated with X-rays (2, 4 and 8 Gy) were reduced by LPA. Conversely, LPA3 agonist (2 S)-OMPT enhanced the cell growth activities of X-ray irradiated MG-63 cells. The cell movement of MG-63 cells exposed to X-ray irradiation (8 Gy) was inhibited by (2 S)OMPT. In cell survival assay, (2 S)-OMPT suppressed the cell survival to cisplatin (CDDP) of MG-63 cells irradiated with X-rays (8 Gy). The cell survival to CDDP of X-ray irradiated cells was elevated by LPA3 knockdown. Moreover, we evaluated the effects of LPA2 on the cell survival to CDDP of MG-63 cells exposed to X-ray irradiation (8 Gy). The cell survival to CDDP of X-ray irradiated cells was increased by LPA2 agonist GRI-977143 and reduced by LPA2 knockdown. These results suggest that LPA receptor-signaling participates in the modulation of cellular functions induced by X-ray irradiation in osteosarcoma cells.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Receptores del Ácido Lisofosfatídico , Humanos , Neoplasias Óseas/patología , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Lisofosfolípidos/farmacología , Lisofosfolípidos/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/radioterapia , Receptores del Ácido Lisofosfatídico/metabolismo , Receptores del Ácido Lisofosfatídico/efectos de la radiación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Rayos X
20.
Oncol Rep ; 47(5)2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35417035

RESUMEN

Irradiation with a specific wavelength of light using light­emitting diodes (LEDs) has various effects on cells and organisms. Recently, the antitumor effects of visible blue light on tumor cells were reported; however, the mechanism and effects on the tumor microenvironment remain unclear. Human colon cancer cells (HCT­116) were injected into the rectal wall of nude mice. Tumors were irradiated with a 465­nm LED light at 30 mW/cm2 for 30 min. Tumor volumes and the expression levels of opsin 3 (Opn3), autophagy­related factors, cancer­associated fibroblast (CAF) markers, and programmed cell death 1­ligand (PD­L1) were measured. Additionally, human intestinal fibroblasts were cultured in HCT116­conditioned medium (CM) to prepare CAFs. CAFs were divided into an LED group and a control group, and the effect of the LED light on CAF activation in colon cancer cells was examined. Irradiation with blue LED light suppressed tumor growth; Opn3 expression was localized to the cell membrane in the LED group. Irradiated tumors exhibited increased autophagy­related gene expression. Furthermore, in the LED group, TGF­ß and α­SMA expression levels in the fibroblasts were decreased. Regarding CAFs, α­SMA and IL­6 expression levels were decreased in the LED group. HCT­116 cells cultured in CAF­CM with LED irradiation showed no enhanced migration or invasion. In the HCT­116 cells cultured in CM of CAFs irradiated with LED, the relative increase in PD­L1 expression was lower than that noted in the CAF­CM without LED irradiation. Blue LED light may have a direct antitumor effect on colon cancer and also an inhibitory effect on CAFs.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias del Colon , Luz , Animales , Antígeno B7-H1/metabolismo , Fibroblastos Asociados al Cáncer/efectos de la radiación , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de la radiación , Neoplasias del Colon/genética , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Opsinas de Bastones/metabolismo , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...