Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Food Funct ; 15(3): 1294-1309, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38197246

RESUMEN

Hypoglycemia can potentially cause severe damage to the central nervous system. The ketogenic diet (KD), characterized by high-fat and extremely low-carbohydrate content, can modulate homeostasis and nutrient metabolism, thereby influencing body health. However, the effects and underlying mechanisms of KD on hypoglycemia-induced brain injury have not been thoroughly investigated. We aimed to explore the modulating effects of KD on cognitive functions and elucidate the underlying mechanisms. In this study, one-month-old mice were fed with KD for 2 weeks, and the changes in the gut microbiota were detected using the 16S rRNA gene amplicon sequencing method. The hypoglycemic model of mice was established using insulin, and the potential protective effect of KD on hypoglycemia-induced brain injury in mice was evaluated through immunofluorescence staining, western blotting, transmission electron microscopy, and Golgi staining. Our results showed that the intestinal flora of Dorea increased and Rikenella decreased in KD-fed mice. KD can not only alleviate anxiety-like behavior induced by hypoglycemia, but also increase the proportion of mushroom dendritic spines in the hippocampus by modulating changes in the gut microbiota. KD regulated synaptic plasticity by increasing the levels of SPN, PSD95, and SYP, which relieve cognitive impairment caused by hypoglycemia. Moreover, KD can promote the proliferation and survival of adult neural stem cells in the hippocampus, while reducing apoptosis by suppressing the activation of the IRE1-XBP1 and ATF6 endoplasmic reticulum stress pathways in mice with hypoglycemia. This study provides new evidence for demonstrating that KD may alleviate cognitive dysfunctions caused by hypoglycemia by modulating the gut microbiota.


Asunto(s)
Lesiones Encefálicas , Disfunción Cognitiva , Dieta Cetogénica , Hipoglucemia , Ratones , Animales , Dieta Cetogénica/métodos , ARN Ribosómico 16S , Estrés del Retículo Endoplásmico , Dieta Alta en Grasa
2.
Metabolism ; 152: 155769, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38158076

RESUMEN

BACKGROUND: Lipophagy is a vital biological process that maintains the balance of intracellular lipid metabolism in nonalcoholic fatty liver disease (NAFLD). However, the precise regulatory mechanism of RNF186 in hepatic lipophagy is still unclear. This study investigates the roles and mechanisms of RNF186 in the regulation of lipophagy during the development of NAFLD. METHODS: In this study, we employed RNF186 knockout mice as well as human liver cells and mouse primary hepatocytes (MPHs) to investigate the role and mechanisms of RNF186 in lipophagy during the progression of NAFLD. Additionally, liver specimens from individuals with NAFLD were examined to assess the expression of RNF186 and its associated factors. RESULTS: Here, we provide evidence that depletion of RNF186 enhances lipophagy in hepatocytes of a NAFLD model. Mechanistically, RNF186 acts as an E3 ubiquitin ligase that targets cytoplasmic HMGB1 for lysine 48 (K48)- and K63-linked ubiquitination, leading to its subsequent proteasomal degradation. Importantly, the translocation of HMGB1 from the nucleus to the cytoplasm is responsible for inducing lipophagy in NAFLD samples. Knockdown of HMGB1 significantly reduces the activation of lipophagy and mediates the decrease in lipid accumulation caused by RNF186 depletion in hepatocytes. Furthermore, we find that maintaining the nuclear HMGB1 level and inhibiting its nuclear-cytoplasmic shuttling are critical for the proper function of RNF186 in NAFLD. Additionally, the expression of RNF186 and HMGB1 in human NAFLD samples, along with factors related to lipophagy, suggest that RNF186 may play a similar role in the pathogenesis of human fatty liver. CONCLUSION: RNF186 deficiency accelerates hepatic lipophagy in NAFLD through the inhibition of ubiquitination and degradation of cytoplasmic HMGB1. Consequently, targeting the RNF186-HMGB1 axis may offer a promising strategy for the prevention and treatment of NAFLD.


Asunto(s)
Proteína HMGB1 , Enfermedad del Hígado Graso no Alcohólico , Animales , Humanos , Ratones , Autofagia/genética , Citoplasma/metabolismo , Hepatocitos/metabolismo , Proteína HMGB1/genética , Proteína HMGB1/metabolismo , Metabolismo de los Lípidos/genética , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
3.
Biochem Biophys Rep ; 36: 101583, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38053620

RESUMEN

Angiogenesis is critical for wound healing and tissue repair. Umbilical cord mesenchymal stem cells (UCMSCs)-conditioned medium has certain actions to promote angiogenesis, and is expected for wound healing and tissue repair. However, recent studies showed that the pro-angiogenic efficacy of unprocessed MSCs-conditioned medium is low, and insufficient for tissue repair. Autophagy is a process for protein recycling and a contributor for cell exocrine, which may enhance pro-angiogenic efficacy of the conditioned medium by stimulating cytokine release from UCMSCs. Therefore, in this study we attempted to obtain enhanced autophagy in UCMSCs using different concentrations of rapamycin and compared pro-angiogenic functions of the conditioned media. The in vitro data showed that although 100 nM-10 µM rapamycin all could induce autophagy in UCMSCs, 100 nM was the best dose to optimize the angiogenic effect of the conditioned medium. The in vivo data also showed that pro-angiogenic effect of the optimized conditioned medium was more obvious than that of the control conditioned medium (0 nM group) in the injected matrigel plaques. Further, the expressions of VEGF, FGF-2, MMP-9, PDGF-α and PDGF-ß were markedly increased in UCMSCs treated with 100 nM rapamycin. In conclusion, appropriately enhancing autophagy of UCMSC can improve pro-angiogenic efficacy of the conditioned medium, which may optimize therapeutic applications of UCMSCs-conditioned medium in wound healing and tissue repair.

4.
Int Immunopharmacol ; 124(Pt B): 111025, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37827056

RESUMEN

Oxaliplatin is a chemotherapy drug currently utilized in the treatment of advanced cancer patients. However, its tolerability poses a limitation to its clinical application. Studies have demonstrated that the presence of tumor-associated macrophages is positively correlated with poor prognosis in various solid tumors, including hepatocellular carcinoma (HCC), and is a significant factor contributing to oxaliplatin resistance. Therefore, targeting tumor-associated macrophages may be an effective strategy to improve the efficacy of oxaliplatin in the treatment of HCC patients. CD24 is a novel target for tumor therapy that can interact with the inhibitory receptor Siglec-10 on tumor-associated macrophages, transmitting immune inhibitory signals and inhibiting macrophage phagocytosis function. In this study, we utilized RNAi technology to inhibit the expression of CD24 in tumor cells and combined it with oxaliplatin, resulting in reduced tumor invasion, migration, and proliferation, as well as increased cell apoptosis. Furthermore, immunofluorescence and flow cytometry results indicated that both the single treatment group and combination treatment group enhanced the infiltration of immune cells. This study presents a novel approach to identifying combination therapy and targets for the clinical treatment of HCC with oxaliplatin.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Oxaliplatino/farmacología , Oxaliplatino/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , ARN Interferente Pequeño/genética , Proteínas Portadoras , Línea Celular Tumoral , Antígeno CD24/genética , Antígeno CD24/metabolismo
5.
Explor Target Antitumor Ther ; 2(3): 266-291, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-36046433

RESUMEN

Metastasis is still the primary cause of cancer-related mortality. However, the underlying mechanisms of cancer metastasis are not yet fully understood. Currently, the epithelial-mesenchymal transition, metabolic remodeling, cancer cell intercommunication and the tumor microenvironment including diverse stromal cells, are reported to affect the metastatic process of cancer cells. Calcium ions (Ca2+) are ubiquitous second messengers that manipulate cancer metastasis by affecting signaling pathways. Diverse transporter/pump/channel-mediated Ca2+ currents form Ca2+ oscillations that can be decoded by Ca2+-binding proteins, which are promising prognostic biomarkers and therapeutic targets of cancer metastasis. This paper presents a review of the advances in research on the mechanisms underlying cancer metastasis and the roles of Ca2+-related signals in these events.

6.
J Cardiovasc Pharmacol Ther ; 26(1): 3-11, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32851865

RESUMEN

Endothelial-to-mesenchymal transition (EndMT) is a complex biological process by which endothelial cells lose their endothelial cell characteristics and acquire mesenchymal cell properties under certain physiological or pathological conditions. Recently, it has been found that EndMT plays an important role in the occurrence and development of fibrotic cardiovascular diseases. In this review, we first summarize the main induction pathways involved in EndMT process. In addition, we discuss the role of EndMT in fibrotic cardiovascular diseases and its potential implication in new therapeutic interventions.


Asunto(s)
Células Endoteliales/patología , Transición Epitelial-Mesenquimal , Cardiopatías/patología , Miocardio/patología , Animales , Células Endoteliales/metabolismo , Fibrosis , Cardiopatías/metabolismo , Humanos , Miocardio/metabolismo , Transducción de Señal
7.
Front Aging Neurosci ; 12: 512097, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33328952

RESUMEN

Background: There is a significant gender difference in the incidence and symptoms of Alzheimer's disease (AD), but its mechanisms are not completely understood. Recent studies showed that NLRP1 inflammasome was overexpressed in females under some pathological conditions such as nodular melanoma. Whether NLRP1 signals have a gender difference in AD has not been elucidated. This study was designed to investigate gender difference on the expressions of NLRP1 signals including NLRP1, Capase-1 and IL-1ß in the brains of APP/PS1+/- mice. Methods: Female and male APP/PS1+/- mice (30-weeks-old) were used in this study. Amyloid-ß (Aß) plaques were stained with Congo red dye and cell apoptosis was detected by TUNEL staining. Expressions of NLRP1, Capase-1 and IL-1ß were measured by immunofluorescent staining and Western blotting assay. Results: The numbers of Aß plaques in cortex and hippocampus and neuronal apoptosis in cortex were 4 and 2-folds in females than males, respectively (P < 0.001). The average size of Aß plaques in both cortex (females: 3527.11 ± 539.88 µm2 vs. males: 1920.44 ± 638.49 µm2) and hippocampus (females: 1931 ± 308.61 µm2 vs. males: 1038.55 ± 220.40 µm2) were also larger in females than males (P < 0.01). More interestingly, expressions of NLRP1, Caspase-1, and IL-1ß were markedly increased in the cortex of females as compared with males. Conclusions: These findings show that NLRP1 signals are higher in brains of female APP/PS1+/- mice than males, which may be related to the gender differences of AD.

8.
Mol Cancer ; 19(1): 58, 2020 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-32171304

RESUMEN

Circular RNAs (circRNAs), one type of non-coding RNA, were initially misinterpreted as nonfunctional products of pre-mRNA mis-splicing. Currently, circRNAs have been proven to manipulate the functions of diverse molecules, including non-coding RNAs, mRNAs, DNAs and proteins, to regulate cell activities in physiology and pathology. Accumulating evidence indicates that circRNAs play critical roles in tumor genesis, development, and sensitivity to radiation and chemotherapy. Radiotherapy and chemotherapy are two primary types of intervention for most cancers, but their therapeutic efficacies are usually retarded by intrinsic and acquired resistance. Thus, it is urgent to develop new strategies to improve therapeutic responses. To achieve this, clarification of the underlying mechanisms affecting therapeutic responses in cancer is needed. This review summarizes recent progress and mechanisms of circRNAs in cancer resistance to radiation and chemotherapy, and it discusses the limitations of available knowledge and potential future directions.


Asunto(s)
Biomarcadores de Tumor/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/patología , ARN Circular/genética , Tolerancia a Radiación/genética , Animales , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/radioterapia
9.
Biochem Pharmacol ; 175: 113863, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32081791

RESUMEN

NLRP3 inflammasome is an intracellular protein complex that initiates cellular injury via assembly of NLRP3, ASC and caspase-1 in response to microbial infection and sterile stressors. The importance of NLRP3 inflammasome in immunity and human diseases has been well documented. Up to now, targeted inhibition of the assembly of NLRP3 inflammasome complex and of its activation was thought to be therapeutic strategy for associated diseases. Recent studies show that a host of molecules such as NIMA-related kinase 7 (Nek7) and DEAD-box helicase 3 X-linked (DDX3X) and a large number of biological mediators including cytokines, microRNAs, nitric oxide, carbon monoxide, nuclear factor erythroid-2 related factor 2 (Nrf2) and cellular autophagy participate in the activation and inactivation of NLRP3 inflammasome. This review summarizes current understanding of the molecular basis of NLRP3 inflammasome activation and inactivation. This knowledge may lead to development of new therapies directed at NLRP3 inflammasome related diseases.


Asunto(s)
Inflamasomas/metabolismo , Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Procesamiento Proteico-Postraduccional , Autofagia/inmunología , Citocinas/metabolismo , ARN Helicasas DEAD-box/metabolismo , Humanos , Inflamación/patología , Factor 2 Relacionado con NF-E2/metabolismo , Quinasas Relacionadas con NIMA/metabolismo , Óxido Nítrico/metabolismo
10.
Br J Pharmacol ; 176(9): 1190-1205, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30801705

RESUMEN

Due to its Ca2+ buffering capacity, the mitochondrion is one of the most important intracellular organelles in regulating Ca2+ dynamic oscillation. Mitochondrial calcium uniporter (MCU) is the primary mediator of Ca2+ influx into mitochondria, manipulating cell energy metabolism, ROS production, and programmed cell death, all of which are critical for carcinogenesis. The understanding of the uniporter complex was significantly boosted by recent groundbreaking discoveries that identified the uniporter pore-forming subunit MCU and its regulatory molecules, including MCU-dominant negative ß subunit (MCUb), essential MCU regulator (EMRE), MCU regulator 1 (MCUR1), mitochondrial calcium uptake (MICU) 1, MICU2, and MICU3. These provide the means and molecular platform to investigate MCU complex (uniplex)-mediated impaired Ca2+ signalling in physiology and pathology. This review aims to summarize the progress of the understanding regulatory mechanisms of uniplex, roles of uniplex-mediated Ca2+ signalling in cancer, and potential pharmacological inhibitors of MCU.


Asunto(s)
Canales de Calcio/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Antineoplásicos/farmacología , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Humanos , Terapia Molecular Dirigida
11.
Cell Physiol Biochem ; 48(6): 2302-2317, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30114704

RESUMEN

BACKGROUND/AIMS: Multidrug resistance (MDR) induced by the ABC transporter subfamily B member 1 (ABCB1) and subfamilyG member 2 (ABCG2) limits successful cancer chemotherapy and no commercially available MDR modulator is used in the clinic. In the current study, we aimed to investigate the effects of PCI29732 on the enhancement of chemotherapeutic agents. METHODS: Cell cytotoxicity and reversal effect were measured with MTT assay. Additionally, flow cytometry was employed to detect the accumulation and efflux of the drugs. We investigated the interaction of PCI29732 and the substrate binding sites of ABCG2 was investigated via the photo-labeling of ABCG2 with the [125I] iodoarylazidoprazosin. The vanadate-sensitive ATPase activity of ABCG2 was measured to identify whether the drug affected the ATPase activity. RT-PCR and Western blot were utilized to analyze mRNA and protein expression respectively. RESULTS: Here, we found that PCI29732 significantly enhanced the efficacy of substrate chemotherapeutic agents in ABCG2-overexpressing cells and also in xenografts harboring the H460/MX20 cell that overexpress ABCG2, but not in their parental sensitive cells and ABCB1-overexpressing cells. Mechanistically, the intracellular accumulations of doxorubicin and Rhodamine 123 were increased in ABCG2-overexpressing S1-MI-80 cells with the presence of PCI29732. PCI29732 stimulated the ATPase activity of ABCG2 at low concentrations. However at the high concentrations, PCI29732 inhibited the ATPase activity, and competed with [125I]-iodoarylazidoprazosin for photo-affinity labeling of ABCG2. PCI29732 did neither alter the mRNA or protein expression levels of ABCG2 nor the phosphorylation levels of AKT and ERK1/2. CONCLUSION: Our study demonstrates that PCI29732 inhibits the function of ABCG2 by competitively binding to the ATP-binding site of ABCG2 and enhances the anti-tumor efficacy of substrate chemotherapeutic agents, This findings encourages the development of combinational chemotherapy for the treatment of ABCG2- overexpressing cancer patients.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Ciclopentanos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Agammaglobulinemia Tirosina Quinasa , Animales , Antineoplásicos/metabolismo , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Doxorrubicina/metabolismo , Doxorrubicina/farmacología , Humanos , Ratones , Ratones Desnudos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Rodamina 123/metabolismo
12.
Nat Commun ; 9(1): 2605, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973582

RESUMEN

Various types of nanoparticles have been proposed for targeted drug delivering, imaging, and tracking of therapeutic agents. However, highly biocompatible nanoparticles with structure-induced fluorescence and capability to conjugate with biomarkers and drugs remain lacking. This research proposes and synthesizes fluorescent nanoparticles (f-PNPs) assembled by cyclic peptides to combine imaging and drug delivering for esophageal cancer (EC). To achieve tumor targeting, f-PNPs are first conjugated with RGD moieties to selectively target EC cells via αvß3 integrin; the nanoparticles are then embedded with epirubicin (EPI). Cell viability assays and analysis of tissue histology reveal that EPI-loaded RGD-f-PNPs (RGD-f-PNPs/EPI) led to significantly reduced cardiotoxicity and improved anti-tumor activity compared to EPI alone. Moreover, the drug delivery to tumor sites and therapeutic responses could be monitored with near-infrared fluorescence using RGD-f-PNPs/EPI. This unique nanoparticle system may lead to potential approaches for bioorganic fluorescence-based delivering, imaging, and drug release tracking.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Epirrubicina/farmacología , Neoplasias Esofágicas/diagnóstico por imagen , Neoplasias Esofágicas/tratamiento farmacológico , Imagen Molecular/métodos , Animales , Antibióticos Antineoplásicos/farmacocinética , Línea Celular Tumoral , Composición de Medicamentos , Monitoreo de Drogas/métodos , Epirrubicina/farmacocinética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Fluorescencia , Humanos , Inyecciones Intraperitoneales , Integrina alfaVbeta3/metabolismo , Masculino , Ratones , Ratones Desnudos , Nanopartículas/administración & dosificación , Nanopartículas/química , Oligopéptidos/química , Oligopéptidos/metabolismo , Péptidos Cíclicos/química , Péptidos Cíclicos/metabolismo , Espectroscopía Infrarroja Corta , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Lett ; 432: 169-179, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-29908962

RESUMEN

Esophageal cancer (EC) is the 6th leading cause of cancer mortality worldwide with poor prognosis, hence more effective chemotherapeutic drugs for this deadly disease are urgently needed. We previously reported that high expression of Orai1, a store-operated Ca2+entry (SOCE) channel, was associated with poor survival rate in EC patients and Orai1-mediated intracellular Ca2+ oscillations regulated cancer cell proliferation. Previous studies suggested that Orai1-mediated SOCE is a promising target for EC chemotherapy. Here, we evaluated the anti-cancer effect of a novel SOCE inhibitor, RP4010, in cultured EC cells and xenograft models. Compared to other previously reported SOCE channel inhibitors, RP4010 is more potent in blocking SOCE and inhibiting cell proliferation in EC and other cancer cells. Treatment with RP4010 resulted in reduction of intracellular Ca2+ oscillations, caused cell cycle arrest at G0/G1 phase in vitro, decreased nuclear translocation of nuclear factor kappa B (NF-κB) in vivo and in vitro, and inhibited tumor growth in vivo. Taken together, data demonstrated the therapeutic potential of RP4010 in EC patients via inhibition of SOCE-mediated intracellular Ca2+ signaling.


Asunto(s)
Antineoplásicos/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio/química , Calcio/metabolismo , Carcinoma de Células Escamosas/patología , Neoplasias Esofágicas/patología , Proteína ORAI1/metabolismo , Compuestos Orgánicos/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis , Señalización del Calcio/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Movimiento Celular , Proliferación Celular , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína ORAI1/genética , Compuestos Orgánicos/uso terapéutico , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
FASEB J ; 32(1): 404-416, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28928244

RESUMEN

Zinc, an essential micronutrient, has a cancer preventive role. Zinc deficiency has been shown to contribute to the progression of esophageal cancer. Orai1, a store-operated Ca2+ entry (SOCE) channel, was previously reported to be highly expressed in tumor tissues removed from patients with esophageal squamous cell carcinoma (ESCC) with poor prognosis, and elevation of its expression contributes to both hyperactive intracellular Ca2+ oscillations and fast cell proliferation in human ESCC cells. However, the molecular basis of cancer preventive functions of zinc and its association with Orai1-mediated cell proliferation remains unknown. The present study shows that zinc supplementation significantly inhibits proliferation of ESCC cell lines and that the effect of zinc is reversible with N,N,N',N'-tetrakis (2-pyridylmethyl) ethylenediamine, a specific Zn2+ chelator, whereas nontumorigenic esophageal epithelial cells are significantly less sensitive to zinc treatment. Fluorescence live cell imaging revealed that extracellular Zn2+ exerted rapid inhibitory effects on Orai1-mediated SOCE and on intracellular Ca2+ oscillations in the ESCC cells. Knockdown of Orai1 or expression of Orai1 mutants with compromised zinc binding significantly diminished sensitivity of the cancer cells to zinc treatment in both SOCE and cell proliferation analyses. These data suggest that zinc may inhibit cell proliferation of esophageal cancer cells through Orai1-mediated intracellular Ca2+ oscillations and reveal a possible molecular basis for zinc-induced cancer prevention and Orai1-SOCE signaling pathway in cancer cells.-Choi, S., Cui, C., Luo, Y., Kim, S.-H., Ko, J.-K., Huo, X., Ma, J., Fu, L.-W., Souza, R. F., Korichneva, I., Pan, Z. Selective inhibitory effects of zinc on cell proliferation in esophageal squamous cell carcinoma through Orai1.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Proteína ORAI1/metabolismo , Zinc/farmacología , Sustitución de Aminoácidos , Señalización del Calcio/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quelantes/farmacología , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago , Etilenodiaminas/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteína ORAI1/antagonistas & inhibidores , Proteína ORAI1/genética
15.
Acta Pharm Sin B ; 7(1): 3-17, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28119804

RESUMEN

The intracellular calcium ions (Ca2+) act as second messenger to regulate gene transcription, cell proliferation, migration and death. Accumulating evidences have demonstrated that intracellular Ca2+ homeostasis is altered in cancer cells and the alteration is involved in tumor initiation, angiogenesis, progression and metastasis. Targeting derailed Ca2+ signaling for cancer therapy has become an emerging research area. This review summarizes some important Ca2+ channels, transporters and Ca2+-ATPases, which have been reported to be altered in human cancer patients. It discusses the current research effort toward evaluation of the blockers, inhibitors or regulators for Ca2+ channels/transporters or Ca2+-ATPase pumps as anti-cancer drugs. This review is also aimed to stimulate interest in, and support for research into the understanding of cellular mechanisms underlying the regulation of Ca2+ signaling in different cancer cells, and to search for novel therapies to cure these malignancies by targeting Ca2+ channels or transporters.

16.
Zhong Yao Cai ; 35(6): 863-6, 2012 Jun.
Artículo en Chino | MEDLINE | ID: mdl-23236815

RESUMEN

OBJECTIVE: To investigate the effect of Gecko crude peptides (GCPS) on human liver carcinoma HepG2 cells and its mechanism. METHODS: MTT assay was used to analyze the effect of the GCPS on the proliferation of HepG2 Cell; Nucleus change of HepG2 treated with GCP was observed by Hoechst33258 fluorescence staining, and BAX and BCL-2 were detected with western-blot assay. RESULTS: GCPS could inhibit the proliferation of HepG2 Cell in a time and dosage dependent way, and its half-maximal inhibitory concentration (IC50) was 1.2 mg/mL; HepG2 pretreated with GCPS showed apoptotic morphological changes. GCPS (1.6 mg/mL, 0.8 mg/mL) could decrease the expression of BCL-2 protein, and increase the expression of BAX protein. CONCLUSION: GCPS can inhibit the proliferation of HepG2 cell. The mechanism may be related to the induction apoptosis of HepG2.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Lagartos , Materia Medica/farmacología , Péptidos/farmacología , Animales , Western Blotting , Relación Dosis-Respuesta a Droga , Células Hep G2 , Humanos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Coloración y Etiquetado/métodos , Factores de Tiempo , Proteína X Asociada a bcl-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...