Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Neoplasma ; 68(5): 924-937, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33998239

RESUMEN

Homoharringtonine (HHT), was first isolated from the bark of Cephalotaxus harringtonia (Knight ex J. Forbes) K. Koch and Cephalotaxus fortunei Hook trees. The bark extract is used to treat leukemia and in recent years has also been used in traditional Chinese medicine (TCM) to treat solid tumors. However, the inhibitory mechanism of HHT in the progression of hepatocellular carcinoma (HCC) is rarely studied. We aimed to evaluate the antitumor efficacy of HHT on HCC in vitro and in vivo and elucidate the underlying molecular mechanism(s). HCC cell lines, including HCCLM3, HepG2, and Huh7, were used to evaluate the antitumor efficacy of HHT in vitro. Cytotoxicity and proliferative ability were evaluated by MTT and colony formation assays. Cell cycle progression and apoptosis in HHT-treated HCC cells were evaluated by flow cytometry. To determine the migration and invasion abilities of HCC cells, wound-healing and Transwell assays were used. Finally, western blot analysis was used to reveal the proteins involved. We also established a xenograft nude mouse model for in vivo assessments of the preclinical efficacy of HHT, mainly using hematoxylin and eosin staining, immunohistochemistry, ultrasound imaging (USI), and magnetic resonance imaging (MRI). HHT suppressed the proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) of HCC cells, and induced cell cycle arrest at the G2 phase and apoptosis. In the HCC xenograft model, HHT showed an obvious tumor-suppressive effect. Surprisingly, Slug expression was also decreased by HHT via the PI3K/AKT/GSK3ß signaling pathway at least partially suppressed the growth of HCC via the PI3K/AKT/GSK3ß/Slug signaling pathway.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Glucógeno Sintasa Quinasa 3 beta , Homoharringtonina , Neoplasias Hepáticas/tratamiento farmacológico , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Oncol Rep ; 45(1): 139-150, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33416119

RESUMEN

Fangchinoline (FAN), an alkaloid extracted from Stephania tetrandra, has a variety of biological and pharmacological activities, but evidence of its effects on colon adenocarcinoma (COAD) is limited. Therefore, the present study aimed to elucidate the molecular mechanisms by which FAN affects COAD. The cytotoxicity, viability and proliferation of DLD­1 and LoVo cells were assessed in the presence of FAN using MTT and colony formation assays. The effects of FAN on apoptosis and the cell cycle in COAD cells were analysed by flow cytometry, and the migration and invasion of these cells were assessed by wound healing and Transwell experiments. Furthermore, a network pharmacological analysis was conducted to investigate the target of FAN and the results were confirmed by western blotting. In addition, a xenograft model was established in nude mice, and ultrasound imaging was used to assess the preclinical therapeutic effects of FAN in vivo. To the best of our knowledge, the results of this study provided the first evidence that FAN inhibited cellular proliferation, stemness, migration, invasion, angiogenesis and epithelial­mesenchymal transition (EMT), and induced apoptosis and G1­phase cell cycle arrest. Network pharmacological analysis further confirmed that FAN prevented EMT through the epidermal growth factor receptor (EGFR)­phosphoinositide 3­kinase (PI3K)/AKT signalling pathway. Finally, FAN significantly repressed tumour growth and promoted apoptosis in xenografts. Thus, targeting EGFR with FAN may offer a novel therapeutic approach for COAD.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Bencilisoquinolinas/farmacología , Neoplasias del Colon/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Adenocarcinoma/patología , Animales , Bencilisoquinolinas/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/patología , Medicamentos Herbarios Chinos/uso terapéutico , Receptores ErbB/metabolismo , Femenino , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Life Sci ; 258: 118158, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32750435

RESUMEN

AIMS: Glioblastoma multiforme (GBM) is characterized by aggressive infiltration and terrible lethality. The overwhelming majority of chemotherapeutic drugs fail to exhibit the desired treatment effects. Polydatin (PD), which was initially extracted from Polygonum cuspidatum, is distinguished for its outstanding cardioprotective, hepatoprotective, and renal protective effects, as well as significant anticancer activities. However, the anti-GBM effect of PD is unclear. MATERIALS AND METHODS: Cell proliferation and apoptosis after PD intervention were estimated using MTT, colony formation and flow cytometry assays in vitro, while wound-healing and Transwell assays were applied to assess cell migration and invasion. In addition, the anti-GBM effects of PD in vivo were detected in the subcutaneous tumor model of nude mice. Moreover, Western blot, immunofluorescence and immunohistochemical staining assays were employed to elaborate the relevant molecular mechanisms. KEY FINDINGS: The present study demonstrated that PD repressed cell proliferation, migration, invasion and stemness and promoted apoptosis in GBM cells. Moreover, by correlating the molecular characteristics of cancer cells with different sensitivities to PD and employing diverse analytical methods, we ultimately verified that the cytotoxicity of PD was related to EGFR-AKT/ERK1/2/STAT3-SOX2/Snail signaling pathway inhibition, in which multiple components were vital therapeutic targets of GBM. SIGNIFICANCE: This work demonstrated that PD could inhibit proliferation, migration, invasion and stemness and induce apoptosis by restraining multiple components of the EGFR-AKT/ERK1/2/STAT3-SOX2/Snail signaling pathway in GBM cells.


Asunto(s)
Antineoplásicos/uso terapéutico , Glioblastoma/tratamiento farmacológico , Glucósidos/uso terapéutico , Transducción de Señal/efectos de los fármacos , Estilbenos/uso terapéutico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Glucósidos/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT3/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Estilbenos/farmacología
4.
Int J Oncol ; 57(1): 183-196, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32319595

RESUMEN

Cyclovirobuxine D (CVB­D) is an alkaloid, which is mainly derived from Buxus microphylla. It has been reported that CVB­D has positive effects on breast cancer, gastric cancer and other malignant tumors. However, to the best of our knowledge, there are no reports regarding the effects of CVB­D on colorectal cancer (CRC). The purpose of the present study was to determine the anticancer effects of CVB­D and further elucidate its molecular mechanism(s). DLD­1 and LoVo cell lines were selected to evaluate the antitumor effect of CVB­D. Cytotoxicity, viability and proliferation were evaluated by the MTT and colony formation assays. Flow cytometry was used to detect the effects on apoptosis and the cell cycle in CVB­D­treated CRC cells. The migration and invasion abilities of CRC cells were examined by wound healing and Transwell assays. In addition, RNA sequencing, bioinformatics analysis and western blotting were performed to investigate the target of drug action and clarify the molecular mechanisms. A xenograft model was established using nude mice, and ultrasound was employed to assess the preclinical therapeutic effects of CVB­D in vivo. It was identified that CVB­D inhibited the proliferation, migration, stemness, angiogenesis and epithelial­mesenchymal transition of CRC cells, and induced apoptosis and S­phase arrest. In addition, CVB­D significantly inhibited the growth of xenografts. It is notable that CVB­D exerted anticancer effects in CRC cells partly by targeting collagen triple helix repeat containing 1 (CTHRC1), which may be upstream of the AKT and ERK pathways. CVB­D exerted anticancer effects through the CTHRC1­AKT/ERK­Snail signaling pathway. Targeted therapy combining CTHRC1 with CVB­D may offer a promising novel therapeutic approach for CRC treatment.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Colon/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Adenocarcinoma/irrigación sanguínea , Adenocarcinoma/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Neoplasias del Colon/irrigación sanguínea , Neoplasias del Colon/patología , Medicamentos Herbarios Chinos/uso terapéutico , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Interferente Pequeño/metabolismo , RNA-Seq , Factores de Transcripción de la Familia Snail/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Theranostics ; 9(5): 1453-1473, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30867843

RESUMEN

Glioblastoma multiforme (GBM) has been considered the most aggressive glioma type. Temozolomide (TMZ) is the main first-line chemotherapeutic agent for GBM. Decreased mutS homolog 6 (MSH6) expression is clinically recognized as one of the principal reasons for GBM resistance to TMZ. However, the specific functions of MSH6 in GBM, in addition to its role in mismatch repair, remain unknown. Methods: Bioinformatics were employed to analyze MSH6 mRNA and protein levels in GBM clinical samples and to predict the potential cancer-promoting functions and mechanisms of MSH6. MSH6 levels were silenced or overexpressed in GBM cells to assess its functional effects in vitro and in vivo. Western blot, qRT-PCR, and immunofluorescence assays were used to explore the relevant molecular mechanisms. Cu2(OH)PO4@PAA nanoparticles were fabricated through a hydrothermal method. Their MRI and photothermal effects as well as their effect on restraining the MSH6-CXCR4-TGFB1 feedback loop were investigated in vitro and in vivo. Results: We demonstrated that MSH6 is an overexpressed oncogene in human GBM tissues. MSH6, CXCR4 and TGFB1 formed a triangular MSH6-CXCR4-TGFB1 feedback loop that accelerated gliomagenesis, proliferation (G1 phase), migration and invasion (epithelial-to-mesenchymal transition; EMT), stemness, angiogenesis and antiapoptotic effects by regulating the p-STAT3/Slug and p-Smad2/3/ZEB2 signaling pathways in GBM. In addition, the MSH6-CXCR4-TGFB1 feedback loop was a vital marker of GBM, making it a promising therapeutic target. Notably, photothermal therapy (PTT) mediated by Cu2(OH)PO4@PAA + near infrared (NIR) irradiation showed outstanding therapeutic effects, which might be associated with a repressed MSH6-CXCR4-TGFB1 feedback loop and its downstream factors in GBM. Simultaneously, the prominent MR imaging (T1WI) ability of Cu2(OH)PO4@PAA could provide visual guidance for PTT. Conclusions: Our findings indicate that the oncogenic MSH6-CXCR4-TGFB1 feedback loop is a novel therapeutic target for GBM and that PTT is associated with the inhibition of the MSH6-CXCR4-TGFB1 loop.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Glioblastoma/fisiopatología , Glioblastoma/terapia , Hipertermia Inducida/métodos , Fototerapia/métodos , Receptores CXCR4/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Biología Computacional , Modelos Animales de Enfermedad , Retroalimentación Fisiológica , Perfilación de la Expresión Génica , Humanos , Ratones Endogámicos BALB C , Modelos Teóricos , Nanoestructuras/administración & dosificación , Resultado del Tratamiento , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Biomater Sci ; 7(1): 409-418, 2018 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-30488900

RESUMEN

The development of nanoplatforms with integrated therapeutic and imaging functions is necessary for highly efficient cancer therapy. Herein, 3D CuS hollow nanoflowers (HNs) consisting of 2D nanoplates are successfully fabricated through the technique of laser ablation in liquids followed by ion-exchange reactions and applied for the first time as a theranostic nanoagent for magnetic resonance imaging (MRI), photothermal therapy (PTT), and chemotherapy simultaneously. Due to the sufficient and immediate contact between the exposed cupric centers of nanoplates and protons from water molecules, CuS HNs are demonstrated to be capable of being a T1 positive contrast agent for efficient MRI of tumors on the T2-weighted fluid-attenuated inversion recovery imaging (T2-FLAIR) sequence. Besides, the hollow structure makes CuS HNs an efficient nanoplatform for drug loading with a laser-triggered drug release. Moreover, CuS HNs exhibit high photothermal conversion efficiency (30%) and good biocompatibility. The combination of PTT and chemotherapy with CuS HNs provides a significant synergistic therapeutic effect, resulting in a higher tumor inhibition ratio than PTT or chemotherapy alone. This study demonstrates a single-component multifunctional theranostic nanoagent for T2-FLAIR MRI guided thermochemotherapy, which has great potential application in theranostics of cancer.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Cobre/uso terapéutico , Doxorrubicina/uso terapéutico , Nanoestructuras/uso terapéutico , Neoplasias/diagnóstico por imagen , Neoplasias/terapia , Nanomedicina Teranóstica/métodos , Animales , Antibióticos Antineoplásicos/administración & dosificación , Medios de Contraste/química , Medios de Contraste/uso terapéutico , Cobre/química , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/uso terapéutico , Doxorrubicina/administración & dosificación , Liberación de Fármacos , Femenino , Células Hep G2 , Humanos , Hipertermia Inducida/métodos , Rayos Láser , Imagen por Resonancia Magnética/métodos , Ratones Endogámicos BALB C , Ratones Desnudos , Nanoestructuras/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA