Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Healthcare (Basel) ; 10(4)2022 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-35455864

RESUMEN

OBJECTIVES: To evaluate the feasibility of implementation of an extremity exercise program and to examine its preliminary effects in breast cancer survivors suffering from chemotherapy-induced peripheral neuropathy (CIPN). SAMPLE & SETTING: Thirteen breast cancer survivors from one hospital in northern Taiwan. Methods and Variables: A single group with repeated measures, and a quasi-experimental design. The intervention program was a four week, home-based extremity exercise program that was comprised of 10 skilled hand exercises and Buerger-Allen exercises. The Total Neuropathy Scale (clinical version), Functional Assessment of Cancer Therapy/Gynecologic Oncology Group, Neurotoxicity (13-Item Version), Identification Pain Questionnaire, and pain Visual Analogue Scale were used to measure CIPN before exercise (T1), during (T2~T4), and after exercise (T5). Qualitative data were also collected at each time point. Data were analyzed by using descriptive statistics, generalized estimating equations, and directed content analysis. RESULTS: None of the participants reported adverse events during the study period. The extremity exercise program significantly improved patient-reported CIPN after intervention at T4 or T5 but was insignificant on clinician-assessed CIPN. The qualitative data of participant experience indicated that this program is feasible and easy to follow. CONCLUSION: The extremity exercise program is feasible but needs to increase the sample size and prolong the intervention period for confirmation.

2.
Hu Li Za Zhi ; 64(2): 19-27, 2017 Apr.
Artículo en Chino | MEDLINE | ID: mdl-28393335

RESUMEN

Breast cancer is a major disease threating women. From diagnosis and treatment through regular follow-ups, patients with breast cancer may experience physical and mental discomfort and pain caused by disease and treatment-related symptoms. These symptoms may further affect disease progression, emotional state, health function status, quality of life, and survival. Symptom experience is as a dynamic process that expresses a patient's feeling regarding the totality of his or her experience. Symptom experience involves the patient's perception of the frequency, intensity, distress, and meaning that accompany the production and expression of symptoms. Cancer patients often experience multiple symptoms distress. Although these symptoms may occur in isolation, multiple symptoms are typically experienced simultaneously. Therefore, obtaining information from patients about the occurrence and distress of their symptom experience is essential to assess their symptom experiences. The present article presents an overview of symptom experiences, explores breast cancer symptom clusters, and analyzes related factors of influence for patients with breast cancer. The authors hope that the findings will help nurses better understand the symptom experiences of breast cancer patients and further apply this understanding in clinical practice in order to assess symptom experiences correctly and to provide more appropriate interventions to improve quality of care.


Asunto(s)
Neoplasias de la Mama/psicología , Neoplasias de la Mama/enfermería , Femenino , Humanos
3.
Environ Toxicol ; 29(4): 428-39, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22431435

RESUMEN

Prostate cancer has its highest incidence and is becoming a major concern. Many studies have shown that traditional Chinese medicine exhibited antitumor responses. Quercetin, a natural polyphenolic compound, has been shown to induce apoptosis in many human cancer cell lines. Although numerous evidences show multiple possible signaling pathways of quercetin in apoptosis, there is no report to address the role of endoplasmic reticulum (ER) stress in quercetin-induced apoptosis in PC-3 cells. The purpose of this study was to investigate the effects of quercetin on the induction of the apoptotic pathway in human prostate cancer PC-3 cells. Cells were treated with quercetin for 24 and 48 h and at various doses (50-200 µM), and cell morphology and viability decreased significantly in dose-dependent manners. Flow cytometric assay indicated that quercetin at 150 µM caused G0/G1 phase arrest (31.4-49.7%) and sub-G1 phase cells (19.77%) for 36 h treatment and this effect is a time-dependent manner. Western blotting analysis indicated that quercetin induces the G0/G1 phase arrest via decreasing the levels of CDK2, cyclins E, and D proteins. Quercetin also stimulated the protein expression of ATF, GRP78, and GADD153 which is a hall marker of ER stress. Furthermore, PC-3 cells after incubation with quercetin for 48 h showed an apoptotic cell death and DNA damage which are confirmed by DAPI and Comet assays, leading to decrease the antiapoptotic Bcl-2 protein and level of ΔΨm , and increase the proapoptotic Bax protein and the activations of caspase-3, -8, and -9. Moreover, quercetin promoted the trafficking of AIF protein released from mitochondria to nuclei. These data suggest that quercetin may induce apoptosis by direct activation of caspase cascade through mitochondrial pathway and ER stress in PC-3 cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico , Mitocondrias/fisiología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Quercetina/farmacología , Transporte Activo de Núcleo Celular , Factor Inductor de la Apoptosis/metabolismo , Calcio/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Fase G1/efectos de los fármacos , Humanos , Masculino , Especies Reactivas de Oxígeno/metabolismo , Fase de Descanso del Ciclo Celular/efectos de los fármacos , Transducción de Señal
4.
Melanoma Res ; 21(4): 267-73, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21734530

RESUMEN

Melanoma is one of the most common cancers worldwide and its incidence has been increasing over the past few decades. Gallic acid (GA) can inhibit the growth of human cancer cells in vitro and in vivo. However, there is no available information to address the effects of GA on migration and invasion of human skin cancer cells. Matrix metalloproteinases (MMPs), zinc-dependent proteolytic enzymes, play an important role in the invasion, metastasis, and angiogenesis of cancer cells. Therefore, MMPs are one of the targets for agents to suppress and that could inhibit the migration and invasion of cancer cells. GA affected the viable A375.S2 cells by propidium iodide exclusion and flow cytometric analysis. Cell migration and invasion were investigated by Boyden chamber assay and we also determined the levels of protein and mRNA expression cell migration and invasion by gelatin zymography, western blotting, and real-time PCR assays. In this study, we examined the influence of GA on the protein levels and gene expression of MMP-2 and MMP-9 and in-vitro migration and invasiveness of human melanoma cells. GA decreases the MMPs and associated signal pathway protein and MMPs mRNA levels in A375.S2 human melanoma cells. Our findings suggest that GA has antimetastatic potential by decreasing invasiveness of cancer cells. Moreover, this action of GA was involved in the Ras, p-ERK signaling pathways resulting in inhibition of MMP-2 in A375.S2 human melanoma cells. These data, therefore, provide evidence for the role of GA as a potential cancer chemotherapeutic agent, which can markedly inhibit the invasive capacity of melanoma cells.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ácido Gálico/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Melanoma/enzimología , Neoplasias Cutáneas/enzimología , Proteínas ras/antagonistas & inhibidores , Western Blotting , Línea Celular Tumoral , Ensayos de Migración Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Melanoma/genética , Melanoma/patología , Invasividad Neoplásica , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Factores de Tiempo , Proteínas ras/genética , Proteínas ras/metabolismo
5.
Int J Oncol ; 38(6): 1605-13, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21424116

RESUMEN

Diallyl sulfide (DAS), one of the main active constituents of garlic, causes growth inhibition of cancer cells in vitro and promotes immune responses in vivo in experimental settings. However, its effects on the induction of cell cycle and apoptosis in human cervical cancer cells are still unclear. The aims of this study were to explore the anti-cancer effects of DAS in HeLa human cervical cancer cells and to investigate the underlying mechanisms in vitro. Cytotoxicity and apoptosis in HeLa human cervical cancer cells were examined by the morphological changes, viability assay, 4',6-Diamidino-2-phenylindole dihydrochloride (DAPI) staining, comet assay, Western blotting and confocal microscopy examination. The results showed that DAS treatment for 24-72 h resulted in a marked decrease in cell viability time- and dose-dependently. Flow cytometric analysis showed that a 48-h treatment of 75 µM DAS induced G0/G1 cell cycle arrest and sub-G1 phase (apoptosis) in HeLa cells. Typical apoptotic nucleus alterations were observed by fluorescence microscopy in HeLa cells after exposure to DAS using DAPI staining. Cells treated with different concentrations of DAS also showed changes typical of apoptosis such as morphological changes, DNA damage and fragmentation, dysfunction of mitochondria, cytochrome c release and increased expression of pro-caspase-3 and -9. DAS also promoted the release of AIF and Endo G from mitochondria in HeLa cells. In conclusion, DAS induced G0/G1 cell cycle arrest and apoptosis in HeLa cells through caspase- and mitochondria and p53 pathways providing further understanding of the molecular mechanisms of DAS action in cervical cancer. This study, therefore, revealed that DAS significantly inhibits the growth and induces apoptosis of human cervical cancer HeLa cells in vitro.


Asunto(s)
Compuestos Alílicos/farmacología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Mitocondrias/metabolismo , Transducción de Señal/efectos de los fármacos , Sulfuros/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Antioxidantes/farmacología , Apoptosis/genética , Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Modelos Biológicos , Biosíntesis de Proteínas/efectos de los fármacos , Transducción de Señal/genética
6.
Anticancer Res ; 30(10): 4187-92, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21036739

RESUMEN

20-Fluoro-6,7-methylenedioxy-2-phenyl-4-quino-lone (CHM-1) has been reported to induce cell cycle arrest and apoptosis in many types of cancer cells. However, there is no available information to show CHM-1 affecting DNA damage and expression of associated repair genes. Herein, we investigated whether or not CHM-1 induced DNA damage and affected DNA repair gene expression in U-2 OS human osterogenic sarcoma cells. The comet assay showed that incubation of U-2 OS cells with 0, 0.75, 1.5, 3 and 6 µM of CHM-1 led to a longer DNA migration smear (comet tail). DNA gel electrophoresis showed that 3 µM of CHM-1 for 24 and 48 h treatment induced DNA fragmentation in U-2 OS cells. Real-time PCR analysis showed that treatment with 3 µM of CHM-1 for 24 h reduced the mRNA expression levels of ataxia telangiectasia mutated (ATM), ataxia-telangiectasia and Rad3-related (ATR), breast cancer 1, early onset (BRCA1), 14-3-3sigma (14-3-3σ), DNA-dependent serine/threonine protein kinase (DNA-PK) and O(6)-methylguanine-DNA methyltransferase (MGMT) genes in a time-dependent manner. Taken together, the results indicate that CHM-1 caused DNA damage and reduced DNA repair genes in U-2 OS cells, which may be the mechanism for CHM-1-inhibited cell growth and induction of apoptosis.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Daño del ADN , Reparación del ADN/efectos de los fármacos , Dioxoles/farmacología , Osteosarcoma/tratamiento farmacológico , Quinolonas/farmacología , Proteínas 14-3-3/biosíntesis , Proteínas 14-3-3/genética , Proteínas de la Ataxia Telangiectasia Mutada , Proteína BRCA1/biosíntesis , Proteína BRCA1/genética , Biomarcadores de Tumor/biosíntesis , Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Ensayo Cometa , Fragmentación del ADN/efectos de los fármacos , Metilasas de Modificación del ADN/biosíntesis , Metilasas de Modificación del ADN/genética , Reparación del ADN/genética , Enzimas Reparadoras del ADN/biosíntesis , Enzimas Reparadoras del ADN/genética , Proteína Quinasa Activada por ADN/biosíntesis , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Exonucleasas/biosíntesis , Exonucleasas/genética , Exorribonucleasas , Expresión Génica/efectos de los fármacos , Humanos , Osteosarcoma/genética , Proteínas Serina-Treonina Quinasas/biosíntesis , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética
7.
Arch Pharm Res ; 33(8): 1181-91, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20803121

RESUMEN

Dietary polyphenols have been correlated with a reduced risk of developing cancer. Quercetin (a natural polyphenolic compound) induced apoptosis in many human cancer cell lines, including breast cancer MCF-7 cells. However, the involvement of possible signaling pathways and the roles of quercetin in apoptosis are still undefined. The purpose of this study was to investigate the effects of quercetin on the induction of the apoptotic pathway in human breast cancer MCF-7 cells. When MCF-7 cells were treated with quercetin for 24 and 48 h and at various doses (10-175 microM), cell viability decreased significantly in time- and dose-dependent manners. Exposure of MCF-7 cells to 10-175 microM quercetin resulted in an approximate 90.25% decrease in viable cells. To explicate the mechanism underlying the antiproliferative effect of quercetin, cell cycle distribution and apoptosis in MCF-7 cells was investigated after exposure to 150 microM quercetin for 6-48 h. Quercetin caused a remarkable increase in the number of S phase (14.56% to 61.35%) and sub-G1 phase cells (0.1% to 8.32%) in a dose- and time-dependent manner. Quercetin caused S phase arrest by decreasing the protein expression of CDK2, cyclins A and B while increasing the p53 and p57 proteins. Following incubation with quercetin for 48 h, MCF-7 cells showed apoptotic cell death by the decreased levels of Bcl-2 protein and DeltaPsi(m) and increased activations of caspase-6, -8 and -9. Moreover, quercetin increased the AIF protein released from mitochondria to nuclei and the GADD153 protein translocation from endoplasmic reticulum to the nuclei. These data suggested that quercetin may induce apoptosis by direct activation of the caspase cascade through the mitochondrial pathway in MCF-7 cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Quercetina/farmacología , Antineoplásicos Fitogénicos/administración & dosificación , Factor Inductor de la Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis/metabolismo , Neoplasias de la Mama/patología , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Mitocondrias/efectos de los fármacos , Quercetina/administración & dosificación , Factores de Tiempo , Factor de Transcripción CHOP/efectos de los fármacos , Factor de Transcripción CHOP/metabolismo
8.
Int J Oncol ; 37(2): 377-85, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20596665

RESUMEN

The natural antioxidant gallic acid (GA) has demonstrated a significant inhibition of cell proliferation and induction of apoptosis in a series of cancer cell lines. However, there is no available information to show whether GA induces apoptosis in human skin cancer cells. In the present study, we report GA-induced apoptosis in A375.S2 human melanoma cells. GA affected morphological changes, decreased the percentage of viable cells and induced apoptosis in A375.S2 cells in a dose- and time-dependent manner. Observation of the molecular mechanism of apoptosis in A375.S2 cells showed that GA up-regulated the proapoptotic proteins such as Bax, and induced caspase cascade activity, but down-regulated antiapoptotic proteins such as Bcl-2. GA induced reactive oxygen species (ROS) and intracellular Ca2+ productions and decreased the level of mitochondrial membrane potential (DeltaPsim) in A375.S2 cells in a time-dependent manner. GA triggered cytosolic release of apoptotic molecules, cytochrome c, promoted activation of caspase-9 and caspase-3, and ultimately apoptotic cell death. In addition, GA also promoted cytosolic release of apoptosis-inducing factor (AIF) and endonuclease G (Endo G). Therefore, GA may also induce apoptosis through a caspase-independent pathway. Our results suggest that GA might be a potential anticancer compound; however, in depth in vivo studies are needed to elucidate the exact mechanism.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasas/fisiología , Ácido Gálico/farmacología , Melanoma/patología , Antineoplásicos/farmacología , Proteínas Reguladoras de la Apoptosis/metabolismo , Calcio/metabolismo , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocromos c/metabolismo , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Humanos , Melanoma/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Modelos Biológicos , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
9.
J Orthop Res ; 27(12): 1637-44, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19557855

RESUMEN

Novel 2-phenyl-4-quinolone compounds have potent cytotoxic effects on different human cancer cell lines. In this study, we examined anticancer activity and mechanisms of 20-fluoro-6,7-methylenedioxy-2-phenyl-4-quinolone (CHM-1) in human osterogenic sarcoma U-2 OS cells. CHM-1-induced apoptosis was determined by flow cytometric analysis, DAPI staining, Comet assay, and caspase inhibitors. CHM-1-inhibited cell migration and invasion was assessed by a wound healing assay, gelatin zymography, and a Transwell assay. The mechanisms of CHM-1 effects on apoptosis and metastasis signaling pathways were studied using Western blotting and gene expression. CHM-1 induced G2/M arrest and apoptosis at an IC(50) (3 microM) in U-2 OS cells and caspase-3, -8, and -9 were activated. Caspase inhibitors increased cell viability after exposure to CHM-1. CHM-1-induced apoptosis was associated with enhanced ROS generation, DNA damage, decreased DeltaPsi(m) levels, and promotion of mitochondrial cytochrome c release. CHM-1 stimulated mRNA expression of caspase-3, -8, and -9, AIF, and Endo G. In addition, CHM-1 inhibited cell metastasis at a low concentration (<3 microM). CHM-1 inhibited the cell metastasis through the inhibition of MMP-2, -7, and -9. CHM-1 also decreased the levels of MAPK signaling pathways before leading to the inhibition of MMPs. In summary, CHM-1 is a potent inducer of apoptosis, which plays a role in the anticancer activity of CHM-1.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Dioxoles/farmacología , Inhibidores de la Metaloproteinasa de la Matriz , Osteosarcoma/tratamiento farmacológico , Quinolonas/farmacología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Inhibidores de Caspasas , Caspasas/biosíntesis , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ensayo Cometa , Daño del ADN , Ensayos de Selección de Medicamentos Antitumorales , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteosarcoma/genética , Osteosarcoma/patología , Especies Reactivas de Oxígeno/metabolismo
10.
Anticancer Res ; 29(1): 165-74, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19331147

RESUMEN

Capsaicin, a pungent ingredient of red pepper, has been reported to possess antitumor activities. In this study, the effects of capsaicin on human HepG2 cells were investigated. Capsaicin reduced viability by PI incorporation in HepG2 cells in a dose and time dependent manner. Capsaicin promoted intracellular Ca2+ production and reactive oxygen species (ROS). The alpha psi(m) significantly decreased after capsaicin treatment for 24 h. Co-treatment of HepG2 cells with capsaicin and BAPTA (an intracellular Ca2+ chelator) significantly reduced intracellular Ca2+ levels, prevented alpha psi(m) disruption and inhibited apoptosis induction. The protein levels of Bcl-2 decreased and Bax increased in the mitochondrial fraction while the Bax protein decreased, and p53 and cytochrome c protein levels increased in the cytosolic fraction in HepG2 cells after capsaicin treatment for 24 h by Western blot. Immunostaining and confocal microscopic analysis also showed that capsaicin promoted cytoplasmic GADD153 expression and GRP78 nuclear translocation. The caspase-3 activity significantly increased after capsaicin treatment for 24 h. Our results indicated that the capsaicin-induced apoptosis in HepG2 cells may result from the elevation of intracellular Ca2+ production, ROS, disruption of alpha psi(m), regulation of Bcl-2 family protein expression and caspase-3 activity.


Asunto(s)
Apoptosis/efectos de los fármacos , Capsaicina/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Calcio/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Citocromos c/biosíntesis , Chaperón BiP del Retículo Endoplásmico , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/metabolismo , Mitocondrias Hepáticas/patología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Especies Reactivas de Oxígeno/metabolismo
11.
Neurochem Res ; 34(8): 1491-7, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19263217

RESUMEN

Curcumin is reported to be a potent inhibitor of the initiation and promotion of many cancer cells. We investigated to examine whether or not curcumin induce DNA damage in mouse-rat hybrid retina ganglion cell line N18 cells. The Comet assay showed that incubation of N18 cells with 10, 25 and 30 microM of curcumin led to a longer DNA migration smear (Comet tail). The DNA gel electrophoresis showed that 20 microM of curcumin for 24 and 48 h treatment induced DNA damage and fragments in N18 cells. The real time PCR analysis showed that 20 microM of curcumin for 48 h treatment decreased ATM, ATR, BRCA1, 14-3-3sigma, DNA-PK and MGMT mRNA, and ATM and MGMT mRNA expression were inhibited in a time-dependent manner. Our results indicate that curcumin caused DNA damage and inhibited DNA repair genes which may be the factors for curcumin-inhibited cell growth.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Curcumina/farmacología , Reparación del ADN/genética , Células Ganglionares de la Retina/efectos de los fármacos , Animales , Línea Celular , Ensayo Cometa , Daño del ADN , Fibroblastos , Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
12.
Leuk Res ; 33(11): 1505-11, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19250670

RESUMEN

Many evidences have shown that dietary intake of cruciferous vegetables could protect against the risk of various types of malignancies. Benzyl isothiocyanate (BITC), one of the compounds from cruciferous vegetables, had shown induced cell cycle arrest and apoptosis in cancer cells. However, there is no available information to address that BITC affects murine leukemia cells in vitro and in vivo. Here, we investigated in vitro effects of BITC on murine leukemia WEHI-3 cells. BITC decreased the percentage of viable cells via G0/G1 arrest and apoptosis in WEHI-3 cells. BITC induced apoptosis through the dysfunction of mitochondria (decreased the levels of mitochondria membrane potential) and activation of caspase-3. Then we investigated in vivo effects of BITC on murine leukemia WEHI-3 cells and the results indicated that BITC decreased the weights of liver and spleen and it also decreased the percentage of CD11b and Mac-3 markers, indicating that the differentiation of the precursor of macrophage and B cells was inhibited. BITC promoted the activity of macrophage phagocytosis in cells which are isolated from PBMC and peritoneal (i.p.). Taken together, BITC can affect WEHI-3 cells in vitro and in vivo.


Asunto(s)
Isotiocianatos/farmacología , Leucemia Experimental/patología , Fagocitosis/efectos de los fármacos , Animales , Línea Celular Tumoral , Masculino , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA