Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 710: 149874, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38581950

RESUMEN

Synaptic plasticity is crucial as it dynamically molds the strength and connectivity of neural circuits, influencing learning, memory, and the development of neurological disorders. Metformin, a widely prescribed anti-diabetic medication, has been shown to readily cross the blood-brain barrier (BBB) and the placenta. However, its prolonged impact on neuronal morphology and functions remains underexplored. In this study, we investigated the influence of metformin on dendrite development and synaptic plasticity in embryonic brains and primary rat cortical neurons. Our findings reveal a negative modulation of dendrite development by metformin, as evidenced by altered dendritic arborization, impaired dendritic spine morphology and disruptions in synaptic plasticity, suggesting a potential link between metformin exposure and aberrations in neuronal connectivity. In addition, we extend our insights to the impact of maternal metformin exposure on embryonic brains, revealing a significant inhibition of dendrite development in E18.5 rat brains. In conclusion, this study adds to the expanding knowledge base on the non-metabolic effects of metformin, emphasizing the significance of assessing its potential influence on both neuronal structure and function. There is an urgent need for further investigations into the enduring impact of prolonged metformin administration on the structural and functional aspects of neurons.


Asunto(s)
Plasticidad Neuronal , Neuronas , Embarazo , Femenino , Ratas , Animales , Plasticidad Neuronal/fisiología , Aprendizaje , Barrera Hematoencefálica , Dendritas
2.
Artículo en Inglés | MEDLINE | ID: mdl-38682237

RESUMEN

The authors' previous research has shown the pivotal roles of Cyclin-dependent kinase 5 (CDK5) and its regulatory protein p35 in nerve growth factor (NGF)-induced differentiation of sympathetic neurons in PC12 cells. During the process of differentiation, neurons are susceptible to environmental influences, including the effects of drugs. Metformin is commonly used in the treatment of diabetes and its associated symptoms, particularly in diabetic neuropathy, which is characterized by dysregulation of the sympathetic neurons. However, the impacts of metformin on sympathetic neuronal differentiation remain unknown. In this study, we investigated the impact of metformin on NGF-induced sympathetic neuronal differentiation using rat pheochromocytoma PC12 cells as a model. We examined the regulation of TrkA-p35/CDK5 signaling in NGF-induced PC12 differentiation. Our results demonstrate that metformin reduces NGF-induced PC12 differentiation by inactivating the TrkA receptor, subsequently inhibiting ERK and EGR1. Inhibition of this cascade ultimately leads to the down-regulation of p35/CDK5 in PC12 cells. Furthermore, metformin inhibits the activation of the pre-synaptic protein Synapsin-I, a substrate of CDK5, in PC12 differentiation. Additionally, metformin alters axonal and synaptic bouton formation by inhibiting p35 at both the axons and axon terminals in fully differentiated PC12 cells. In summary, our study elucidates that metformin inhibits sympathetic neuronal differentiation in PC12 cells by disrupting TrkA/ERK/EGR1 and p35/CDK5 signaling. This research contributes to uncovering a novel signaling mechanism in drug response during sympathetic neuronal differentiation, enhancing our understanding of the intricate molecular processes governing this critical aspect of neurodevelopment.

3.
Chemosphere ; 358: 142124, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38677614

RESUMEN

Metformin, the most commonly prescribed drug for the treatment of diabetes, is increasingly used during pregnancy to address various disorders such as diabetes, obesity, preeclampsia, and metabolic diseases. However, its impact on neocortex development remains unclear. Here, we investigated the direct effects of metformin on neocortex development, focusing on ERK and p35/CDK5 regulation. Using a pregnant rat model, we found that metformin treatment during pregnancy induces small for gestational age (SGA) and reduces relative cortical thickness in embryos and neonates. Additionally, we discovered that metformin inhibits neural progenitor cell proliferation in the sub-ventricular zone (SVZ)/ventricular zone (VZ) of the developing neocortex, a process possibly mediated by ERK inactivation. Furthermore, metformin induces neuronal apoptosis in the SVZ/VZ area of the developing neocortex. Moreover, metformin retards neuronal migration, cortical lamination, and differentiation, potentially through p35/CDK5 inhibition in the developing neocortex. Remarkably, compensating for p35 through in utero electroporation partially rescues metformin-impaired neuronal migration and development. In summary, our study reveals that metformin disrupts neocortex development by inhibiting neuronal progenitor proliferation, neuronal migration, cortical layering, and cortical neuron maturation, likely via ERK and p35/CDK5 inhibition. Consequently, our findings advocate for caution in metformin usage during pregnancy, given its potential adverse effects on fetal brain development.

4.
Anticancer Res ; 44(2): 543-553, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38307563

RESUMEN

BACKGROUND/AIM: Bladder cancer remains a significant global health concern, necessitating a deeper understanding of the molecular mechanisms underlying its progression. Cyclin-Dependent Kinase 5 (CDK5) has recently emerged as a potential player in bladder cancer pathogenesis. This study investigated the involvement of CDK5 in bladder cancer, emphasizing its potential as a therapeutic target. MATERIALS AND METHODS: The expression levels of CDK5 and p35 (CDK5 regulatory protein) and their roles in the tumor grade and malignancy of patient samples were evaluated using western blot analysis and immunohistochemistry. In addition, tumor cancer genome atlas (TCGA) was utilized to evaluate survival rate in patients with bladder cancer. We further confirmed the role of CDK5 with in vitro experiments using western blot analysis, immunocytochemistry, cell culture-based proliferation and migration assays. RESULTS: Higher CDK5 and p35 were associated with a higher tumor grade and poor survival rate in patients with bladder cancer. To confirm the role of CDK5 in vitro, we over-expressed CDK5 in bladder cancer cells. The results showed that the over-expression of CDK5 enhanced bladder cancer cell proliferation and migration. In addition, CDK5 inhibition by a pan-CDK inhibitor, Roscovitine (RV), significantly reduced proliferation of bladder cancer cells. Indeed, the migration and adhesion of bladder cancer cells were inhibited by RV treatment. CONCLUSION: CDK5 might play important roles in bladder cancer progression and be a potential diagnostic and therapeutic target in the near future.


Asunto(s)
Neoplasias de la Vejiga Urinaria , Humanos , Proliferación Celular , Quinasa 5 Dependiente de la Ciclina/genética , Quinasa 5 Dependiente de la Ciclina/metabolismo , Roscovitina , Tasa de Supervivencia , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
5.
Artículo en Inglés | MEDLINE | ID: mdl-36482936

RESUMEN

Antrodia salmonea (AS) is a genus of Antrodia, an epiphyte of Cunninghamia konishii in Taiwan. AS has been reported to have potential therapeutic effects on different diseases, including diarrhea, abdominal pain, and hypertension. AS has been reported to have anticancer effects on numerous cancer types, such as ovarian carcinoma and triple-negative breast cancer. Our previous studies demonstrated that antrocins and triterpenoids are possibly bioactive compositions. However, the effects of AS on prostate cancer remain unknown. Therefore, we investigated the role of AS in prostate cancer growth, apoptosis, and cell cycle regulation. The results showed that AS extracts significantly inhibited the proliferation of prostate cancer LNCaP cells in a dose-dependent manner and increased the levels of apoptotic markers (cleaved PARP and cleaved caspase 3/8/9). In addition, the cell cycle-related proteins CDK1, CDK2, CDK4, and their respective specific regulators Cyclin B1, Cyclin A, and Cyclin D were also affected. Besides, AS treatment increased p53 protein levels and slowed its degradation in LNCaP cells. Interestingly, we found that AS treatment reduced both total protein and Ser-81 phosphorylation levels of the androgen receptor (AR). Notably, the increase of nuclear p53 was accompanied by the down-regulation of AR, suggesting a reverse regulation between p53 and AR in LNCaP cells was triggered by AS treatment. These findings suggest that AS extracts trigger the apoptosis of prostate cancer cells through the reverse regulation of p53 and AR and elucidate that AS extracts might be a potential treatment for androgen-dependent prostate cancer in the near future.

6.
Chin J Physiol ; 65(4): 209-214, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36073569

RESUMEN

Antrodia salmonea (AS) is a fungus, which belongs to a fungal family of Taiwanofungus salmoneus with the features of anti-oxidant, anti-inflammatory, and anticancer. Recent studies have shown that AS has anti-cancer functions in ovarian and breast cancer. However, the effects of AS on prostate cancer (PCa) proliferation remain unknown. Therefore, we investigated the role of AS in PCa proliferation through apoptosis, and cell cycle regulation in PCa cell lines. Our results showed that Antrodia salmonea extract (ASE) inhibited PCa cells growth with a dose-dependent manner. In addition, ASE decreased the anchorage-independent growth formation ability in PC3 cells. Moreover, ASE-induced cell growth inhibition in PCa cells (DU145, PC3) was correlated to decreased cell cycle-related proteins such as cyclin A/B and cyclin-dependent kinase CDK1/2/4, and increased cell cycle inhibitor proteins p21. Besides, ASE decreased the total protein level of epidermal growth factor receptor and its downstream signaling pathways Akt and Erk in both PCa cells. We found that apoptotic markers such as cleaved-PARP protein levels increased significantly in DU145 cells indicating ASE might induce apoptosis. In conclusion, our results suggest that ASE may have the ability to induce PCa cell death through regulating cell cycle arrest and apoptosis pathways.


Asunto(s)
Apoptosis , Neoplasias de la Próstata , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Humanos , Masculino , Extractos Vegetales/farmacología , Polyporales , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo
7.
Molecules ; 27(14)2022 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-35889228

RESUMEN

Plant-parasitic nematodes (PPNs) constitute the most damaging group of plant pathogens. Plant infections by root-knot nematodes (RKNs) alone could cause approximately 5% of global crop loss. Conventionally, chemical-based methods are used to control PPNs at the expense of the environment and human health. Accordingly, the development of eco-friendly and safer methods has been urged to supplement or replace chemical-based methods for the control of RKNs. Using microorganisms or their metabolites as biological control agents (BCAs) is a promising approach to controlling RKNs. Among the metabolites, volatile organic compounds (VOCs) have gained increasing attention because of their potential in the control of not only RKNs but also other plant pathogens, such as insects, fungi, and bacteria. This review discusses the biology of RKNs as well as the status of various control strategies. The discovery of VOCs emitted by bacteria from various environmental sources and their application potential as BCAs in controlling RKNs are specifically addressed.


Asunto(s)
Tylenchoidea , Compuestos Orgánicos Volátiles , Animales , Bacterias , Humanos , Enfermedades de las Plantas/parasitología , Enfermedades de las Plantas/prevención & control , Plantas/parasitología , Compuestos Orgánicos Volátiles/farmacología
8.
Biomolecules ; 11(6)2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34207842

RESUMEN

Medullary thyroid cancer (MTC) is a neuroendocrine tumor that arises from the parafollicular C-cells, which produces the hormone calcitonin. RET is a transmembrane receptor protein-tyrosine kinase, which is highly expressed in MTC. Our previous studies reported that cyclin-dependent kinase 5 (CDK5) plays a crucial role in cancer progression, including MTC. However, the role of CDK5 in GDNF-induced RET signaling in medullary thyroid cancer proliferation remains unknown. Here, we investigated RET activation and its biochemically interaction with CDK5 in GDNF-induced medullary thyroid cancer proliferation. Our results demonstrated that GDNF stimulated RET phosphorylation and thus subsequently resulted in CDK5 activation by its phosphorylation. Activated CDK5 further caused STAT3 activation by its specific phosphorylation at Ser727. Moreover, we also found that GDNF treatment enhanced ERK1/2 and EGR1 activity, which is involved in p35 activation. Interestingly, we identified for the first time that CDK5 physically interacted with RET protein in MTC. Overall, our results provide a new mechanism for medullary thyroid cancer cell proliferation, suggesting that targeting CDK5 may be a promising therapeutic candidate for human medullary thyroid cancer in the near future.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Proteínas Proto-Oncogénicas c-ret/metabolismo , Factor de Transcripción STAT3/metabolismo , Carcinoma Neuroendocrino/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Quinasa 5 Dependiente de la Ciclina/genética , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Fosforilación , Proteínas Proto-Oncogénicas c-ret/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Factor de Transcripción STAT3/genética , Transducción de Señal , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología
9.
Int J Mol Sci ; 20(16)2019 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-31395805

RESUMEN

Cyclin-dependent kinase 5 (CDK5) is a unique member of the cyclin-dependent kinase family. CDK5 is activated by binding with its regulatory proteins, mainly p35, and its activation is essential in the development of the central nervous system (CNS) and neurodegeneration. Recently, it has been reported that CDK5 plays important roles in regulating various biological and pathological processes, including cancer progression. Concerning prostate cancer, the androgen receptor (AR) is majorly involved in tumorigenesis, while CDK5 can phosphorylate AR and promotes the proliferation of prostate cancer cells. Clinical evidence has also shown that the level of CDK5 is associated with the progression of prostate cancer. Interestingly, inhibition of CDK5 prevents prostate cancer cell growth, while drug-triggered CDK5 hyperactivation leads to apoptosis. The blocking of CDK5 activity by its small interfering RNAs (siRNA) or Roscovitine, a pan-CDK inhibitor, reduces the cellular AR protein level and triggers the death of prostate cancer cells. Thus, CDK5 plays a crucial role in the growth of prostate cancer cells, and AR regulation is one of the important pathways. In this review paper, we summarize the significant studies on CDK5-mediated regulation of prostate cancer cells. We propose that the CDK5-p35 complex might be an outstanding candidate as a diagnostic marker and potential target for prostate cancer treatment in the near future.


Asunto(s)
Quinasa 5 Dependiente de la Ciclina/metabolismo , Neoplasias de la Próstata/patología , Andrógenos/análisis , Andrógenos/metabolismo , Animales , Apoptosis , Carcinogénesis/metabolismo , Carcinogénesis/patología , Quinasa 5 Dependiente de la Ciclina/análisis , Humanos , Masculino , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/terapia , Receptores Androgénicos/análisis , Receptores Androgénicos/metabolismo , Factor de Transcripción STAT3/análisis , Factor de Transcripción STAT3/metabolismo
10.
Toxins (Basel) ; 11(4)2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30925742

RESUMEN

Arecoline is the primary alkaloid in betel nuts, which are known as a risk factor for oral submucosal fibrosis and oral cancer. Lung cancer is a severe type of carcinoma with high cell motility that is difficult to treat. However, the detailed mechanisms of the correlation between Arecoline and lung cancer are not fully understood. Here, we investigated the effect of Arecoline on migration in lung cancer cell lines and its potential mechanism through the muscarinic acetylcholine receptor 3 (mAChR3)-triggered EGFR/Src/FAK pathway. Our results indicate that different concentrations of Arecoline treatment (10 µM, 20 µM, and 40 µM) significantly increased the cell migration ability in A549 and CL1-0 cells and promoted the formation of the filamentous actin (F-actin) cytoskeleton, which is a crucial element for cell migration. However, migration of H460, CL1-5, and H520 cell lines, which have a higher migration ability, was not affected by Arecoline treatment. The EGFR/c-Src/Fak pathway, which is responsible for cell migration, was activated by Arecoline treatment, and a decreased expression level of E-cadherin, which is an epithelial marker, was observed in Arecoline-treated cell lines. Blockade of the EGFR/c-Src/Fak pathway with the inhibitors of EGFR (Gefitinib) or c-Src (Dasatinib) significantly prevented Arecoline-promoted migration in A549 cells. Gefitinib or Dasatinib treatment significantly disrupted the Arecoline-induced localization of phospho-Y576-Fak during focal adhesion in A549 cells. Interestingly, Arecoline-promoted migration in A549 cells was blocked by a specific mAChR3 inhibitor (4-DAMP) or a neutralizing antibody of matrix metalloproteinase (MMP7 or Matrilysin). Taken together, our findings suggest that mAChR3 might play an essential role in Arecoline-promoted EGFR/c-Src/Fak activation and migration in an A549 lung cancer cell line.


Asunto(s)
Arecolina/farmacología , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias Pulmonares/metabolismo , Receptor Muscarínico M3/metabolismo , Familia-src Quinasas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Humanos , Antagonistas Muscarínicos/farmacología , Piperidinas/farmacología , Receptor Muscarínico M3/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos
11.
Oral Oncol ; 67: 124-130, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28351566

RESUMEN

Being one of the most aggressive cancers of oral cavity, tongue squamous cell carcinoma (TSCC) constitutes 41% of all oral carcinomas. Despite considerable improvements in multimodal diagnosis and treatment techniques, TSCC still remains to be one of the most lethal cancer types in the head and neck region. MicroRNAs are endogenously synthesized, small, non-coding RNAs, which are responsible for post-transcriptional regulation of mRNA expression. They are involved in regulation of almost all biological processes through their spatial and temporal expression. Their deregulation participates in pathogenesis of various diseases, including human TSCC, where they can act as potent oncogenes or tumor suppressors. Extensive microRNA profiling in TSCC samples and further in vitro and in vivo functional characterization of differentially expressed microRNAs revealed their contribution to the underlying molecular mechanisms of TSCC initiation, development, progression, metastasis, chemo-radioresistance, and recurrence. They are suggested as diagnostic and prognostic biomarkers for TSCC due to their differential expression in tumor tissues and their stability in body fluids like plasma, oral cytology, and saliva. MicroRNAs are, therefore, considered amongst the most promising candidates for development of novel therapeutic approaches against TSCC. In this review, we summarized important findings including our own works on microRNAs as implicated in TSCC and the new insights into the roles of microRNAs in squamous cell carcinoma of tongue.


Asunto(s)
Carcinoma de Células Escamosas/patología , MicroARNs/genética , Neoplasias de la Lengua/patología , Carcinoma de Células Escamosas/tratamiento farmacológico , Humanos , Neoplasias de la Lengua/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...