Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 96
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell Tissue Res ; 395(1): 53-62, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37985496

RESUMEN

Glomerular epithelial protein-1 (Glepp1), a R3 subtype family of receptor-type protein tyrosine phosphatases, plays important role in the activation of Src family kinases and regulates cellular processes such as cell proliferation, differentiation, and apoptosis. In this study, we firstly examined the functional evaluation of Glepp1 in tooth development and morphogenesis. The precise expression level and developmental function of Glepp1 were examined by RT-qPCR, in situ hybridization, and loss and gain of functional study using a range of in vitro organ cultivation methods. Expression of Glepp1 was detected in the developing tooth germs in cap and bell stage of tooth development. Knocking down Glepp1 at E13 for 2 days showed the altered expression levels of tooth development-related signaling molecules, including Bmps, Dspp, Fgf4, Lef1, and Shh. Moreover, transient knock down of Glepp1 revealed alterations in cellular physiology, examined by the localization patterns of Ki67 and E-cadherin. Similarly, knocking down of Glepp1 showed disrupted enamel rod and interrod formation in 3-week renal transplanted teeth. In addition, due to attrition of odontoblastic layers, the expression signals of Dspp and the localization of NESTIN were almost not detected after knock down of Glepp1; however, their expressions were increased after Glepp1 overexpression. Thus, our results suggested that Glepp1 plays modulating roles during odontogenesis by regulating the expression levels of signaling molecules and cellular events to achieve the proper structural formation of hard tissue matrices in mice molar development.


Asunto(s)
Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores , Diente , Animales , Ratones , Regulación del Desarrollo de la Expresión Génica , Morfogénesis , Odontogénesis , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/metabolismo , Transducción de Señal , Diente/metabolismo
2.
Eur J Oral Sci ; 129(1): e12750, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33503283

RESUMEN

Patients with oral squamous cell carcinoma (OSCC) bone invasion are surgically treated with bone resection, which results in severe physical and psychological damage. Here, we investigated the potential of fractalkine (CX3CL1), which is regulated by transforming growth factor (TGF-ß), as a novel biomarker for correct prediction and early detection of OSCC-associated bone invasion. TGF-ß knockdown and treatment with a TGF-ß-neutralizing antibody decreased the level of fractalkine in the culture media of HSC-2 and YD10B OSCC cells. Treatment with a fractalkine-neutralizing antibody reduced TGF-ß-stimulated invasion by HSC-2 and YD10B cells. Fractalkine treatment increased the viability, invasion, and uPA secretion of both OSCC cell lines. Furthermore, OSCC cell bone invasion was assessed following subcutaneous inoculation of wild-type or TGF-ß knockdown OSCC cells in mouse calvaria. TGF-ß knockdown prevented erosive bone invasion, reduced the number of osteoclasts at the tumor-bone interface, and downregulated fractalkine expression in mouse tumor tissues. Our results indicate that the production of fractalkine is stimulated by TGF-ß and mediates TGF-ß-induced cell invasion in several OSCC cell lines showing an erosive pattern of bone invasion. Fractalkine may be a useful predictive marker and therapeutic target for OSCC-induced bone destruction.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Animales , Biomarcadores , Línea Celular Tumoral , Quimiocina CX3CL1 , Humanos , Ratones , Invasividad Neoplásica , Carcinoma de Células Escamosas de Cabeza y Cuello , Factor de Crecimiento Transformador beta , Factor de Crecimiento Transformador beta1
3.
Cell Physiol Biochem ; 49(4): 1460-1475, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30205376

RESUMEN

BACKGROUND/AIMS: Bone metastasis of cancer cells decreases patient survival and quality of life. Hybridization via the covalent coupling of two bioactive natural products is a useful strategy for developing more potent anticancer agents by enhancing their bioavailability and avoiding drug resistance. METHODS: The in vivo activities of artemisinin-daumone hybrid 15 (ARTD) were estimated in cancer cell-inoculated mice and ovariectomized mice. The viability, migration, and invasion of cancer cells were measured via MTT, wound-healing, and transwell invasion assays. ARTD-regulated transcription factors were detected with an RT2 profiler PCR array kit and Western blotting. Osteoclastogenesis and osteoclast activity were detected with tartrate-resistant acid phosphatase staining, a pit formation assay, gelatin zymography, and a cathepsin K ELISA assay. RESULTS: ARTD blocked cancer-associated osteolysis more potently than artemisinin in mice with intratibially inoculated breast cancer or lung cancer cells. ARTD inhibited the viability, migration, and invasion of breast and lung cancer cells in the absence or presence of transforming growth factor-ß1. ARTD treatment induced the expression of tumor suppressive activating transcription factor 3 and inhibited oncogenic E2F transcription factor 1 expression at the mRNA and protein levels. ARTD inhibited receptor activator of nuclear factor kappa-B ligand-induced osteoclast formation and bone resorbing activity by reducing the secreted levels of matrix metalloproteinase-9 and cathepsin K. Furthermore, ARTD prevented estrogen deficiency-induced bone loss in ovariectomized mice. CONCLUSION: ARTD may be a promising candidate for inhibiting cancer-induced bone destruction. The application of ARTD may be extended to patients with chemotherapy-induced ovarian failure or postmenopausal osteoporosis.


Asunto(s)
Artemisininas/química , Conservadores de la Densidad Ósea/uso terapéutico , Ácidos Grasos/química , Osteólisis/prevención & control , Feromonas/química , Animales , Conservadores de la Densidad Ósea/química , Conservadores de la Densidad Ósea/farmacología , Huesos/diagnóstico por imagen , Huesos/patología , Catepsina K/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteólisis/etiología , Factor de Transcripción STAT3/metabolismo , Factor de Crecimiento Transformador beta1/farmacología
4.
J Pathol ; 237(4): 520-31, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26239696

RESUMEN

Non-small cell lung cancer (NSCLC) frequently metastasizes to bone, which is associated with significant morbidity and a dismal prognosis. RUNX3 functions as a tumour suppressor in lung cancer and loss of expression occurs more frequently in invasive lung adenocarcinoma than in pre-invasive lesions. Here, we show that RUNX3 and RUNX3-regulated chemokines are linked to NSCLC-mediated bone resorption. Notably, the receptor activator of nuclear factor-κB ligand (RANKL)/osteoprotegerin (OPG) ratio, an index of osteoclastogenic stimulation, was significantly increased in human osteoblastic cells treated with conditioned media derived from RUNX3-knockdown NSCLC cells. We aimed to identify RUNX3-regulated factors that modify the osteoblastic RANKL/OPG ratio and found that RUNX3 knockdown led to CCL5 up-regulation and down-regulation of CCL19 and CXCL11 in NSCLC cells. Tumour size was noticeably increased and more severe osteolytic lesions were induced in the calvaria and tibiae of mice that received RUNX3-knockdown cells. In response to RUNX3 knockdown, serum and tissue levels of CCL5 increased, whereas CCL19 and CXCL11 decreased. Furthermore, CCL5 increased the proliferation, migration, and invasion of lung cancer cells in a dose-dependent manner; however, CCL19 and CXCL11 did not show any significant effects. The RANKL/OPG ratio in osteoblastic cells was increased by CCL5 but reduced by CCL19 and CXCL11. CCL5 promoted osteoclast differentiation, but CCL19 and CXCL11 reduced osteoclastogenesis in RANKL-treated bone marrow macrophages. These findings suggest that RUNX3 and related chemokines are useful markers for the prediction and/or treatment of NSCLC-induced bone destruction.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias Óseas/secundario , Carcinoma de Pulmón de Células no Pequeñas/secundario , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Neoplasias Pulmonares/patología , Animales , Western Blotting , Resorción Ósea , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Quimiocina CCL19/biosíntesis , Quimiocina CCL5/biosíntesis , Quimiocina CXCL11/biosíntesis , Ensayo de Inmunoadsorción Enzimática , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Inmunohistoquímica , Neoplasias Pulmonares/metabolismo , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Microtomografía por Rayos X
5.
Cell Physiol Biochem ; 36(5): 1809-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26184636

RESUMEN

BACKGROUND: Metastatic breast cancer cells are frequently associated with osteoclast-mediated bone resorption, resulting in severe bone destruction and increased mortality in patients. Platycodin D (PD) isolated from Platycodon grandiflorum is a triterpenoid saponin with anti-cancer and anti-angiogenic potential. METHODS: The in vivo activity was determined in mice with the intratibial injection of human metastatic breast cancer cells. Osteoclast formation and activity were detected using tartrate-resistant acid phosphatase staining and calcium phosphate-coated plates. The expression of osteoclastogenesis-inducing molecules was detected by RT-PCR and western blotting in RANKL-treated bone marrow macrophages (BMMs). Cell viability and DNA synthesis were measured with MTT and BrdU incorporation assays. The induction of apoptosis was estimated using TUNEL staining and a caspase-3 activity assay. RESULTS: The oral administration of PD inhibited MDA-MB-231 cell-induced osteolysis in an intratibial mouse model. PD treatment blocked RANKL-induced osteoclast formation by inhibiting the expression and nuclear translocation of NFATc1 and c-Fos in BMMs and consequently reduced osteoclast-mediated bone resorption. Furthermore, PD treatment induced apoptosis in osteoclasts and inhibited the growth of MDA-MB-231 cells. CONCLUSION: PD may block breast cancer-induced bone loss by suppressing the formation, activity, and survival of osteoclasts, as well as the growth of metastatic breast cancer cells.


Asunto(s)
Huesos/patología , Neoplasias de la Mama/patología , División Celular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Saponinas/farmacología , Triterpenos/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Osteoclastos/patología , Factores de Transcripción/antagonistas & inhibidores
6.
Chem Pharm Bull (Tokyo) ; 63(9): 669-77, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26329860

RESUMEN

Novel saponins that retain a free carboxyl group at the C-17 position and various sugars linked at the C-3 position of hederagenin aglycone were synthesized via stereospecific glycosylation. Since these natural products represented by Pulsatilla saponin D (PSD) were obtained in very small amounts, the total synthesis developed in this paper will resolve this problem of scarcity. The two types of synthesized arabinose- and rhamnose-cored saponins showed potent anticancer activity against a human lung cancer cell line (A549), and most disaccharide moiety saponins possessed more potent anti-lung cancer activity. Among the novel PSD analogues containing disaccharide saponins, compound 10i showed anti-lung cancer activity (6.6 µM) that was four-fold more potent than the clinical agent Iressa (26.08 µM).


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Saponinas/farmacología , Antineoplásicos Fitogénicos/síntesis química , Antineoplásicos Fitogénicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Células MCF-7 , Estructura Molecular , Saponinas/síntesis química , Saponinas/química , Relación Estructura-Actividad
7.
Invest New Drugs ; 32(1): 1-13, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23673814

RESUMEN

Breast cancer frequently spreads to bone. The interaction between bone metastases and microenvironment, referred as the "vicious cycle", increases both tumor burden and bone destruction. Therefore, inhibition at any point in this "vicious cycle" can reduce malignant osteolytic lesions in patients with advanced breast cancer. In this study, we evaluated whether tetrahydrofurofuran-type lignans derived from Magnoliae Flos, commonly used in traditional Asian medicine to treat inflammatory diseases, could block breast cancer-mediated bone loss. Aschatin, fargesin, lirioresinol B dimethyl ether, and magnolin at noncytotoxic concentrations suppressed mRNA expression and secretion of osteolytic factor PTHrP in MDA-MB-231 metastatic human breast cancer cells. Fargesin inhibited TGF-ß-stimulated cell viability, migration, and invasion and decreased TGF-ß-induced PTHrP production in MDA-MB-231 cells. In addition, these lignans reduced RANKL/OPG ratio in PTHrP-treated hFOB1.19 human osteoblastic cells and inhibited RANKL-mediated osteoclast differentiation in mouse bone marrow macrophages. Aschatin, fargesin, lirioresinol B dimethyl ether, and magnolin substantially reduced bone-resorbing activity of osteoclasts by inhibiting MMP-9 and cathepsin K activities. Furthermore, orally administered fargesin inhibited tumor growth and cancer-mediated bone destruction in mice with MDA-MB-231 cells injected into calvarial tissues. Aschatin, fargesin, lirioresinol B dimethyl ether, and magnolin blocked initiation and progression of the "vicious cycle" between breast cancer metastases and bone microenvironment by inhibiting PTHrP production in breast cancer cells and osteoclastic bone resorption. Therefore, these tetrahydrofurofuran-type lignans have the potential to serve as beneficial agents to prevent and treat cancer-induced bone destruction in breast cancer patients.


Asunto(s)
Resorción Ósea/tratamiento farmacológico , Resorción Ósea/etiología , Neoplasias de la Mama/patología , Furanos/uso terapéutico , Lignina/uso terapéutico , Osteoblastos/patología , Osteoclastos/patología , Animales , Benzodioxoles/química , Benzodioxoles/farmacología , Benzodioxoles/uso terapéutico , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Femenino , Furanos/química , Furanos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Lignanos/química , Lignanos/farmacología , Lignanos/uso terapéutico , Lignina/química , Lignina/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Metástasis de la Neoplasia , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/biosíntesis , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Toxicol Appl Pharmacol ; 275(2): 152-62, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24463094

RESUMEN

Many breast cancer patients experience bone metastases and suffer skeletal complications. The present study provides evidence on the protective and therapeutic potential of betulinic acid on cancer-associated bone diseases. Betulinic acid is a naturally occurring triterpenoid with the beneficial activity to limit the progression and severity of cancer, diabetes, cardiovascular diseases, atherosclerosis, and obesity. We first investigated its effect on breast cancer cells, osteoblastic cells, and osteoclasts in the vicious cycle of osteolytic bone metastasis. Betulinic acid reduced cell viability and the production of parathyroid hormone-related protein (PTHrP), a major osteolytic factor, in MDA-MB-231 human metastatic breast cancer cells stimulated with or without tumor growth factor-ß. Betulinic acid blocked an increase in the receptor activator of nuclear factor-kappa B ligand (RANKL)/osteoprotegerin ratio by downregulating RANKL protein expression in PTHrP-treated human osteoblastic cells. In addition, betulinic acid inhibited RANKL-induced osteoclastogenesis in murine bone marrow macrophages and decreased the production of resorbed area in plates with a bone biomimetic synthetic surface by suppressing the secretion of matrix metalloproteinase (MMP)-2, MMP-9, and cathepsin K in RANKL-induced osteoclasts. Furthermore, oral administration of betulinic acid inhibited bone loss in mice intra-tibially inoculated with breast cancer cells and in ovariectomized mice causing estrogen deprivation, as supported by the restored bone morphometric parameters and serum bone turnover markers. Taken together, these findings suggest that betulinic acid may have the potential to prevent bone loss in patients with bone metastases and cancer treatment-induced estrogen deficiency.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Triterpenos/administración & dosificación , Administración Oral , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Resorción Ósea/prevención & control , Neoplasias de la Mama/patología , Catepsina K/genética , Catepsina K/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Estrógenos/deficiencia , Estrógenos/metabolismo , Femenino , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos ICR , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteoprotegerina/antagonistas & inhibidores , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/antagonistas & inhibidores , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Triterpenos Pentacíclicos , Ligando RANK/antagonistas & inhibidores , Ligando RANK/genética , Ligando RANK/metabolismo , Ácido Betulínico
9.
J Bone Miner Metab ; 32(2): 110-22, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23624721

RESUMEN

Bone is continuously remodeled by bone formation and resorption, and cooperative bone metabolism is precisely regulated to maintain homeostasis. Osteoclasts, which are responsible for bone resorption, are differentiated through multiple steps that include cell fusion at the last step of differentiation, yielding multinuclear cells. However, the factors involved in and the precise mechanism of cell fusion are still unknown. To determine the molecules involved in osteoclast fusion, we examined the effect of lysophosphatidic acid (LPA), which has been reported to participate in the progression of cancer bone metastasis. LPA had no effect on osteoclast formation and bone resorption under receptor activator of nuclear factor kappa B ligand (RANKL) conditions, whereas LPA stimulated osteoclast fusion, thereby causing increased osteoclast diameter and bone resorptive capacity under a RANKL-limited condition. This result encouraged us to assess what molecules are needed for LPA-stimulated osteoclast fusion. Interestingly, LPA stimulated osteoclast stimulatory transmembrane protein (OC-STAMP) and P2X7 receptor mRNA expression during osteoclast fusion under a RANKL limiting condition. siRNA-induced OC-STAMP or P2X7 receptor knockdown significantly suppressed the LPA-stimulated increase in osteoclast diameter and bone resorptive capacity in differentiating cultures. Using cyclosporin A as an inhibitor, we revealed that NF-ATc1 directly regulates OC-STAMP and P2X7 receptor expression during LPA-stimulated osteoclast fusion. These results suggest that LPA is a critical regulator of osteoclast fusion by inducing the OC-STAMP and P2X7 receptor. Therefore, LPA signaling might be useful to help understand their effects on osteoclast formation and as a therapeutic target for patients with pathologically increased osteoclast formation.


Asunto(s)
Lisofosfolípidos/farmacología , Proteínas de la Membrana/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Receptores Purinérgicos P2X7/metabolismo , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Células Cultivadas , Proteínas de la Membrana/genética , Ratones , Microscopía Confocal , Reacción en Cadena de la Polimerasa , Regulación hacia Arriba/efectos de los fármacos
10.
Chem Pharm Bull (Tokyo) ; 62(5): 446-53, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24614158

RESUMEN

The practical synthesis and anticancer activity of novel deoxoartemisinin-glycolipid hybrids, which incorporate two drugs into a single molecule and can impact multiple targets simultaneously are presented. These hybrids exhibited potent in vitro anticancer activity against several human cancer cell lines. The deoxoartemisinin-glycolipid hybrids generally demonstrated better anticancer activity than either artemisinin or daumone alone and cisplatin.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Artemisininas/farmacología , Glucolípidos/farmacología , Antineoplásicos/química , Artemisininas/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Glucolípidos/química , Humanos , Células MCF-7 , Estructura Molecular , Relación Estructura-Actividad
11.
Phytother Res ; 27(12): 1776-83, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23401151

RESUMEN

The aim of this study was to determine whether the ethanol extract of roasted licorice (rLE) could inhibit breast cancer-mediated bone destruction. rLE treatment reduced the viability of MDA-MB-231 human metastatic breast cancer cells but did not show any cytotoxicity in hFOB1.19 human osteoblastic cells and murine bone marrow-derived macrophages (BMMs). rLE inhibited expression and secretion of receptor activator of nuclear factor κB ligand (RANKL) as well as the mRNA and protein expression of cyclooxygenase-2 in osteoblastic cells exposed to the conditioned medium of breast cancer cells. rLE dramatically inhibited RANKL-induced osteoclastogenesis in BMMs, thereby reducing osteoclast-mediated pit formation. Moreover, treatment with licochalcone A and isoliquiritigenin as the active components, whose contents are increased by the roasting process, remarkably suppressed RANKL-induced osteoclast formation in BMMs, respectively. Furthermore, orally administered rLE substantially blocked tumor growth and bone destruction in mice inoculated with breast cancer cells in the tibiae. Serum levels of tartrate-resistant acid phosphatase and C-terminal cross-linking telopeptide of type I collagen and trabecular bone morphometric parameters were reversed to almost the same levels as the control mice by the rLE treatment. In conclusion, rLE may be a beneficial agent for preventing and treating bone destruction in patients with breast cancer.


Asunto(s)
Resorción Ósea/patología , Huesos/patología , Neoplasias de la Mama/patología , Glycyrrhiza/química , Osteoblastos/efectos de los fármacos , Extractos Vegetales/farmacología , Fosfatasa Ácida/sangre , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Resorción Ósea/tratamiento farmacológico , Huesos/efectos de los fármacos , Línea Celular Tumoral , Chalconas/farmacología , Ciclooxigenasa 2/metabolismo , Femenino , Humanos , Isoenzimas/sangre , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Metástasis de la Neoplasia , Osteoclastos/efectos de los fármacos , Ligando RANK/metabolismo , Fosfatasa Ácida Tartratorresistente
12.
Phytother Res ; 27(4): 493-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22627996

RESUMEN

Xanthorrhizol, a natural sesquiterpenoid compound isolated from Curcuma xanthorrhiza Roxb, has been known to inhibit the growth of human colon, breast, liver and cervical cancer cells. In this study, xanthorrhizol decreased cell viability, induced apoptosis and decreased the level of full-length PARP in SCC-15 oral squamous cell carcinoma (OSCC) cells. A decrease in cell viability and PARP degradation was not prevented by treatment with the caspase inhibitor Z-VAD-fmk in xanthorrhizol-treated cells. Xanthorrhizol treatment elevated intracellular Ca(2+) and ROS levels in SCC-15 cells. Treatment with a Ca(2+) chelator, EGTA/AM, did not affect xanthorrhizol- induced cytotoxicity, but cell viability was partly recovered by treatment with endogenous antioxidant, GSH, or hydroxy radical trapper, MCI-186. Furthermore, the viability of xanthorrhizol-treated SCC-15 cells was significantly restored by treatment with SB203580 and/or SP600125 but not significantly by PD98059 treatment. Xanthorrhizol-induced activation of p38 MAPK and JNK was blocked by MCI-186. Finally, xanthorrhizol suppressed the number of tumors in buccal pouches and increased the survival rate in hamsters treated with 7,12-dimethylbenz[a]anthracene. In conclusion, xanthorrhizol may induce caspase-independent apoptosis through ROS-mediated p38 MAPK and JNK activation in SCC-15 OSCC cells and prevent chemical-induced oral carcinogenesis. Therefore, xanthorrhizol seems to be a promising chemopreventive agent.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias de la Boca/patología , Fenoles/farmacología , 9,10-Dimetil-1,2-benzantraceno , Animales , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Inhibidores de Caspasas/farmacología , Línea Celular Tumoral , Supervivencia Celular , Cricetinae , Humanos , Masculino , Neoplasias de la Boca/metabolismo , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Especies Reactivas de Oxígeno/metabolismo
13.
Carcinogenesis ; 33(11): 2135-46, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22859271

RESUMEN

We previously reported that insulin-like growth factor-II mRNA-binding protein-3 (IMP-3) depletion (IMP-3(Δ)) was shown to inhibit invadopodia formation and extracellular matrix degradation capacity in oral squamous cell carcinoma (OSCC) cells. In this study, we found that IMP-3(Δ) cells significantly downregulated the podoplanin (PDPN) level, which resulted in a loss of extracellular matrix degradation activity, although invadopodia was still thriving. From RNA in situ hybridization using a digoxigenin-labeled 3'UTR recognition probe of PDPN and reporter assay with 3'UTR of the PDPN gene cloned downstream from the luciferase reporter gene, we revealed that IMP-3 depletion was shown to be downregulated, which most probably lowered PDPN gene expression by reducing mRNA stabilization. In a xenograft model, PDPN depletion was the cause of a decrease in tumor volume and regional infiltration into nearby stroma. Taken together, transforming growth factor beta 1 increased PDPN expression, which potentiated cancer invasion through increased invadopodia formation and extracellular matrix degradation in the low invasive OSCC cell line. Reciprocally, interleukin-1 beta secreted by OSCC cells, stimulated transforming growth factor beta 1 secretion from stromal fibroblasts to induce PDPN expression in OSCC cells. In addition, a retrospective investigation of OSCC patients found that IMP-3 and PDPN expression significantly correlated with lymph node metastasis of OSCC patients. Moreover, co-expression of IMP-3 and PDPN were frequently detected both in primary and lymph nodes metastatic OSCC cells using immunohistochemical dual staining. Thus, the IMP-3-PDPN axis may be a sensitive target molecule in anti-invadopodia therapy for the treatment of metastatic cancers.


Asunto(s)
Carcinoma de Células Escamosas/secundario , Comunicación Celular , Extensiones de la Superficie Celular/fisiología , Glicoproteínas de Membrana/genética , Neoplasias de la Boca/patología , Proteínas de Unión al ARN/metabolismo , Células del Estroma/metabolismo , Regiones no Traducidas 3' , Animales , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Adhesión Celular , Movimiento Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Matriz Extracelular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Encía/citología , Encía/metabolismo , Humanos , Técnicas para Inmunoenzimas , Hibridación in Situ , Interleucina-1beta/metabolismo , Metástasis Linfática , Masculino , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Invasividad Neoplásica , Pronóstico , Estabilidad del ARN , ARN Mensajero/química , ARN Mensajero/genética , Proteínas de Unión al ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/patología , Factor de Crecimiento Transformador beta1/metabolismo
14.
Biol Pharm Bull ; 35(3): 289-300, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22382313

RESUMEN

An inability to control cancer cell invasion and metastasis is the leading cause of death in patients with cancer. The present study was performed to determine the anti-invasive effect of Kalopanaxsaponin A (KPS-A) on matrix metalloproteinase-9 (MMP-9)-meidated invasion in phorbol 12-myristate 13-acetate (PMA)-stimulated human oral squamous cell carcinoma (OSCC) cells and a murine xenograft model of human OSCC. KPS-A, isolated from Kalopanax pictus, inhibited PMA-induced proliferation and invasion as well as PMA-induced MMP-9 expression and secretion at non-cytotoxic doses. KPS-A treatment reduced the stability of PMA-induced MMP-9 mRNA and inhibited the PMA-induced cytoplasmic translocation of HuR. In PMA-treated cells, KPS-A treatment resulted in the intracellular accumulation of MMP-9 and suppressed Ras-associated binding 1A (Rab1A) expression. KPS-A treatment suppressed PMA-induced phosphorylation of extracellular signal regulated kinase (ERK)1/2 and Akt. Furthermore, the oral administration of KPS-A led to substantial inhibition of tumor growth and the expression of proliferating cell nuclear antigen (PCNA), MMP-9, tissue inhibitor of metalloproteinase-1 (TIMP-1), HuR, and Rab1A in the tumor tissues of mice inoculated with YD-10B OSCC cells. Collectively, KPS-A inhibits the invasiveness of oral cancer by reducing HuR-mediated MMP-9 mRNA stability and Rab1A-mediated MMP-9 secretion via ERK1/2 and phosphatidylinositide 3-kinase (PI3K)/Akt. Therefore, KPS-A is a promising anti-invasive agent.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Ácido Oleanólico/análogos & derivados , Saponinas/uso terapéutico , Animales , Antineoplásicos Fitogénicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Metaloproteinasa 9 de la Matriz/genética , Ratones , Ratones Desnudos , Neoplasias de la Boca/patología , Invasividad Neoplásica , Ácido Oleanólico/farmacología , Ácido Oleanólico/uso terapéutico , Transporte de Proteínas/efectos de los fármacos , ARN Mensajero/metabolismo , Saponinas/farmacología , Acetato de Tetradecanoilforbol/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Am J Chin Med ; 50(6): 1645-1661, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35848124

RESUMEN

Platycodin D is a major constituent in the root of Platycodon grandiflorum and has diverse pharmacologic activities, including anti-inflammatory, anti-allergic, and antitumor activities. Vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) are potent angiogenic factors and contribute to tumor angiogenesis by directly and indirectly promoting angiogenic processes, including the proliferation, adhesion, migration, and tube formation of endothelial cells. Here, we found that platycodin D at noncytotoxic concentrations inhibited VEGF-induced proliferation, adhesion to the extracellular matrix proteins fibronectin and vitronectin, chemotactic motility, and tube formation of human umbilical vein endothelial cells (HUVECs). Platycodin D reduced the phosphorylation of extracellular signal-regulated kinase (ERK), p38, and c-Jun N-terminal kinase (JNK) and the secretion of IL-8 in VEGF-stimulated HUVECs. Moreover, platycodin D inhibited tube formation and the phosphorylation of ERK and p38 in IL-8-stimulated HUVECs. The in vitro anti-angiogenic activity of platycodin D was confirmed by in vivo experimental models. Platycodin D inhibited the formation of new blood vessels into mouse Matrigel plugs with VEGF or IL-8. In mice injected with MDA-MB-231 human breast cancer cells, orally administered platycodin D inhibited tumor growth, the number of CD34 [Formula: see text]vessels, and the expression of VEGF and IL-8. Taken together, platycodin D directly and indirectly prevents VEGF-induced and IL-8-induced angiogenesis by blocking the activation of mitogen-activated protein kinases (MAPKs). Platycodin D may be beneficial for the prevention or treatment of tumor angiogenesis and angiogenesis-related human diseases.


Asunto(s)
Interleucina-8 , Factor A de Crecimiento Endotelial Vascular , Inhibidores de la Angiogénesis/farmacología , Animales , Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Interleucina-8/metabolismo , Ratones , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/prevención & control , Saponinas , Triterpenos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/farmacología
16.
Phytother Res ; 25(10): 1503-10, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21394802

RESUMEN

Buddlejasaponin IV (BS-IV), a major component of Pleurospermum kamtschaticum, exerts antiinflammatory and cytotoxic effects against cancer cells. The study investigated whether BS-IV could prevent oral carcinogenesis by inhibiting the growth of immortalized human oral keratinocytes (IHOKs). BS-IV reduced cell viability and induced cell cycle arrest at G2/M phase and apoptotic morphological changes in IHOKs. BS-IV inhibited the levels of cyclin B1, Cdc2 and Cdc25C, but enhanced Chk2 phosphorylation. The increased levels of pRb and p21 protein and the activation of p53 were also noted in BS-IV-treated IHOKs. In addition, BS-IV induced cytochrome c release from mitochondria by reducing antiapoptotic Bcl-2 levels and increasing pro-apoptotic Bax levels. BS-IV treatment resulted in the activation of caspase-9 and caspase-3. PARP cleavage was also clearly observed in the BS-IV-treated IHOKs. Furthermore, the expression of the Fas death receptor and Fas ligand was induced and procaspase-8 level was suppressed by BS-IV treatment. Taken together, BS-IV treatment inhibited the growth of IHOK cells via the induction of p53-dependent cell cycle arrest at the G2/M phase and apoptosis via both mitochondrial-dependent and death receptor-mediated pathways. Thus, BS-IV can be considered an excellent candidate for a chemopreventive agent to block the progression of HPV-induced oral carcinogenesis.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apiaceae/química , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Queratinocitos/efectos de los fármacos , Mucosa Bucal/efectos de los fármacos , Saponinas/farmacología , Triterpenos/farmacología , Alphapapillomavirus , Antineoplásicos Fitogénicos/uso terapéutico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/prevención & control , Carcinoma de Células Escamosas/virología , Caspasas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Queratinocitos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mucosa Bucal/metabolismo , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/prevención & control , Neoplasias de la Boca/virología , Proteínas Nucleares/metabolismo , Fosforilación , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Salivales Ricas en Prolina/metabolismo , Saponinas/uso terapéutico , Triterpenos/uso terapéutico , Factores de Necrosis Tumoral/metabolismo , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/metabolismo
17.
Phytother Res ; 25(7): 959-64, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21170925

RESUMEN

The principal objective of the present study was to evaluate the antimetastatic activity of decursin and decursinol isolated from Angelica gigas. Decursin and decursinol inhibited the proliferation and invasion of CT-26 colon carcinoma cells. The expressions of matrix metalloproteinase (MMP)-2 and MMP-9 in cells and the activities in the culture medium were also reduced by decursin and decursinol treatment. In CT-26 cells, the extracellular signal-regulated kinase (ERK) inhibitor inhibited cell proliferation, invasion and MMP-9 expression, and the c-Jun N-terminal kinase (JNK) inhibitor suppressed the expression of both MMPs, as well as cell proliferation and cell invasion. The phosphatidylinositol-3 kinase (PI3K) inhibitor reduced only the expression of MMP-2. In addition, the invasion of CT-26 cells was inhibited by the treatment with anti-MMP-9 antibody, rather than anti-MMP-2 antibody. These results indicate that MMP-9 expression via ERK and JNK plays a critical role for the invasion of CT26 cells. Decursin and decursinol downregulated ERK and JNK phosphorylation. Moreover, oral administration of decursin and decursinol reduced the formation of tumor nodules in the lungs and the increase in lung weight caused by CT-26 metastases. Therefore, both decursin and decursinol may be beneficial antimetastatic agents, targeting MMPs and their upstream signaling molecules.


Asunto(s)
Angelica/química , Antineoplásicos Fitogénicos/farmacología , Benzopiranos/farmacología , Butiratos/farmacología , Neoplasias del Colon/patología , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Neoplasias Pulmonares/secundario , Masculino , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia/tratamiento farmacológico
18.
Chem Pharm Bull (Tokyo) ; 59(12): 1471-5, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22130368

RESUMEN

Novel artemisinin-glycolipid hybrids were directly synthesized from 12ß (C-C)-type deoxoartemisinin and glycolipid and exhibited exceptional in vitro anticancer activity, particularly against the oral carcinoma cancer cell lines, respectively. The artemisinin-glycolipid hybrids, with effective concentrations under 20 µM, demonstrated better anticancer activity than either artemisinin or glycolipid alone and showed five times more anti-oral cancer activity than either cisplatin or paclitaxel.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Artemisininas/química , Artemisininas/farmacología , Glucolípidos/química , Glucolípidos/farmacología , Antineoplásicos/síntesis química , Artemisia/química , Artemisininas/síntesis química , Carcinoma/tratamiento farmacológico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Glucolípidos/síntesis química , Humanos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias/tratamiento farmacológico
19.
Am J Chin Med ; 49(3): 737-751, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33683188

RESUMEN

Angiogenesis plays a crucial role in tumor growth and metastasis. Vascular endothelial growth factor (VEGF)-stimulated endothelial cell proliferation and migration are critical steps in tumor angiogenesis. Here, we investigated the anti-angiogenic activity of xanthorrhizol, a sesquiterpenoid isolated from the Indonesian medicinal plant Curcuma xanthorrhiza. Xanthorrhizol at noncytotoxic concentrations inhibited the proliferation, migration, and formation of capillary-like tubes in VEGF-treated human umbilical vein endothelial cells (HUVECs). Xanthorrhizol inhibited the phosphorylation of Akt and endothelial nitric oxide synthase (eNOS) and the expression of vascular cell adhesion molecule (VCAM)-1 and E-selectin in VEGF-treated HUVECs. The expression and transcriptional activity of NF-[Formula: see text]B were downregulated by xanthorrhizol in VEGF-treated HUVECs. Furthermore, xanthorrhizol significantly inhibited VEGF-induced angiogenesis in the chorioallantoic membrane of fertilized eggs and Matrigel plugs subcutaneously injected into mice. Xanthorrhizol inhibited tumor volume and tumor-derived angiogenesis in mice inoculated with breast cancer cells. The in vitro and in vivo anti-angiogenic activities of xanthorrhizol were as potent as those of curcumin, a well-known anticancer agent derived from C. longa. Taken together, xanthorrhizol inhibits VEGF-induced angiogenesis of endothelial cells by blocking the activation of the PI3K/Akt/eNOS axis and subsequent upregulation of adhesion molecules induced by the transcriptional activation of NF-[Formula: see text]B. Xanthorrhizol is a promising anti-angiogenic agent and can serve as a beneficial agent to enhance anticancer treatments.


Asunto(s)
Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , FN-kappa B/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/prevención & control , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fenoles/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular/efectos adversos , Animales , Curcuma/química , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Neovascularización Patológica/inducido químicamente , Fenoles/aislamiento & purificación , Fenoles/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Fitoterapia , Células Tumorales Cultivadas
20.
Cancers (Basel) ; 13(9)2021 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-33924899

RESUMEN

It is well-known that microbiota dysbiosis is closely associated with numerous diseases in the human body. The oral cavity and gut are the two largest microbial habitats, playing a major role in microbiome-associated diseases. Even though the oral cavity and gut are continuous regions connected through the gastrointestinal tract, the oral and gut microbiome profiles are well-segregated due to the oral-gut barrier. However, the oral microbiota can translocate to the intestinal mucosa in conditions of the oral-gut barrier dysfunction. Inversely, the gut-to-oral microbial transmission occurs as well in inter- and intrapersonal manners. Recently, it has been reported that oral and gut microbiomes interdependently regulate physiological functions and pathological processes. Oral-to-gut and gut-to-oral microbial transmissions can shape and/or reshape the microbial ecosystem in both habitats, eventually modulating pathogenesis of disease. However, the oral-gut microbial interaction in pathogenesis has been underappreciated to date. Here, we will highlight the oral-gut microbiome crosstalk and its implications in the pathogenesis of the gastrointestinal disease and cancer. Better understanding the role of the oral-gut microbiome axis in pathogenesis will be advantageous for precise diagnosis/prognosis and effective treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA