Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Mol Carcinog ; 62(9): 1249-1262, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37191369

RESUMEN

Small molecule degraders of small ubiquitin-related modifier 1 (SUMO1) induce SUMO1 degradation in colon cancer cells and inhibits the cancer cell growth; however, it is unclear how SUMO1 degradation leads to the anticancer activity of the degraders. Genome-wide CRISPR-Cas9 knockout screen has identified StAR-related lipid transfer domain containing 7 (StarD7) as a critical gene for the degrader's anticancer activity. Here, we show that both StarD7 mRNA and protein are overexpressed in human colon cancer and its knockout significantly reduces colon cancer cell growth and xenograft progression. The treatment with the SUMO1 degrader lead compound HB007 reduces StarD7 mRNA and protein levels and increases endoplasmic reticulum (ER) stress and reactive oxygen species (ROS) production in colon cancer cells and three-dimensional (3D) organoids. The study further provides a novel mechanism of the compound anticancer activity that SUMO1 degrader-induced decrease of StarD7 occur through degradation of SUMO1, deSUMOylation and degradation of T cell-specific transcription 4 (TCF4) and thereby inhibition of its transcription of StarD7 in colon cancer cells, 3D organoids and patient-derived xenografts (PDX).


Asunto(s)
Proteínas Portadoras , Neoplasias del Colon , Humanos , Proteínas Portadoras/genética , Especies Reactivas de Oxígeno/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/genética , ARN Mensajero , Estrés del Retículo Endoplásmico , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Factor de Transcripción 4/metabolismo
2.
Phytother Res ; 33(3): 610-617, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30537241

RESUMEN

Protopanaxadiol (PPD), a ginseng metabolite generated by the gut bacteria, was shown to induce colorectal cancer cell death and enhance the anticancer effect of chemotherapeutic agent 5-FU. However, the mechanism by which PPD promotes cancer cell death is not clear. In this manuscript, we showed that PPD activated p53 and endoplasmic reticulum (ER) stress and induced expression of BH3-only proteins Puma and Noxa to promote cell death. Induction of Puma by PPD was p53-dependent, whereas induction of Noxa was p53-independent. On the other hand, PPD also induced prosurvival mechanisms including autophagy and expression of Bcl2 family apoptosis regulator Mcl-1. Inhibition of autophagy or knockdown of Mcl-1 significantly enhanced PPD-induced cell death. Interestingly, PPD inhibited expression of genes involved in fatty acid and cholesterol biosynthesis and induced synergistic cancer cell death with fatty acid synthase inhibitor cerulenin. As PPD-induced ER stress was not significantly affected by inhibition of new protein synthesis, we suggest PPD may induce ER stress directly through causing lipid disequilibrium.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Panax/metabolismo , Sapogeninas/farmacología , Autofagia/efectos de los fármacos , Células HCT116 , Humanos , Proteína p53 Supresora de Tumor/fisiología
3.
J Pharmacol Sci ; 118(4): 447-54, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22510965

RESUMEN

The nuclear factor-κB (NF-κB) transcription factors control many physiological processes including inflammation, immunity, and apoptosis. In our search for NF-κB inhibitors from natural resources, we identified yangonin from Piper methysticum as an inhibitor of NF-κB activation. In the present study, we demonstrate that yangonin potently inhibits NF-κB activation through suppression of the transcriptional activity of the RelA/p65 subunit of NF-κB. This compound significantly inhibited the induced expression of the NF-κB-reporter gene. However, this compound did not interfere with tumor necrosis factor-α (TNF-α)-induced inhibitor of κBα (IκBα) degradation, p65 nuclear translocation, and DNA-binding activity of NF-κB. Further analysis revealed that yangonin inhibited not only the induced NF-κB activation by overexpression of RelA/p65, but also transactivation activity of RelA/p65. Moreover, yangonin did not inhibit TNF-α-induced activation of p38, but it significantly impaired activation of extracellular signal-regulated kinase 1/2 and stress-activated protein kinase/c-Jun NH(2)-terminal kinase. We also demonstrated that pretreatment of cells with this compound prevented TNF-α-induced expression of NF-κB target genes, such as interleukin 6, interleukin 8, monocyte chemotactic protein 1, cyclooxygenase-2 and inducible nitric oxide. Taken together, yangonin could be a valuable candidate for the intervention of NF-κB-dependent pathological conditions such as inflammation.


Asunto(s)
FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Pironas/farmacología , Factor de Transcripción ReIA/antagonistas & inhibidores , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Ratones , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/biosíntesis , Subunidades de Proteína/genética , Pironas/química , Factor de Transcripción ReIA/biosíntesis , Factor de Transcripción ReIA/genética , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética
4.
Cancer Biol Ther ; 23(1): 348-357, 2022 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-35438057

RESUMEN

Overexpression of c-myc via increased transcription or decreased protein degradation is common to many cancer etiologies. c-myc protein degradation is mediated by ubiquitin-dependent degradation, and this ubiquitylation is regulated by several E3 ligases. The primary regulator is Fbxw7, which binds to a phospho-degron within c-myc. Here, we identify a new E3 ligase for c-myc, Fbxl8 (F-box and Leucine Rich Repeat Protein 8), as an adaptor component of the SCF (Skp1-Cullin1-F-box protein) ubiquitin ligase complex, for selective c-myc degradation. SCFFbxl8 binds and ubiquitylates c-myc, independent of phosphorylation, revealing that it regulates a pool of c-myc distinct from SCFFbxw7. Loss of Fbxl8 increases c-myc protein levels, protein stability, and cell division, while overexpression of Fbxl8 reduces c-myc protein levels. Concurrent loss of Fbxl8 and Fbxw7 triggers a robust increase in c-myc protein levels consistent with targeting distinct pools of c-myc. This work highlights new mechanisms regulating c-myc degradation.


Asunto(s)
Proteínas F-Box , Ubiquitina-Proteína Ligasas , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Humanos , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
5.
J Biol Chem ; 285(40): 30539-47, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20682767

RESUMEN

The NF-κB transcription factors control many physiological processes, including inflammation, immunity, and apoptosis. Its activity contributes to the development of various cell malignancies. NF-κB-inducing kinase (NIK) plays a pivotal role in NF-κB activation. However, the molecular mechanism to stabilize and activate NIK remains elusive, although it is known that cIAP1/2 (cellular inhibitor of apoptosis 1 and 2) ubiquitinate NIK for degradation. Here, we report a novel NF-κB-related zinc finger protein 91 (ZFP91) that stabilizes and activates NIK in a ubiquitination-dependent manner. We show that ZFP91 interacts with and promotes the Lys(63)-linked ubiquitination of NIK and subsequent processing of p100 to p52. The results of in vitro biochemical assays indicate that ZFP91 functions as an E3 ligase directly to NIK. Remarkably, the ubiquitination of NIK coincides with its Thr(559) phosphorylation. Furthermore, knockdown of ZFP91 expression by RNA interference inhibits the CD40 ligation-induced activation of NIK and p100 processing as well as the expression of noncanonical NF-κB target genes. These data clearly indicate that ZFP91 is an important regulator of the noncanonical NF-κB pathway.


Asunto(s)
FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Línea Celular Tumoral , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , FN-kappa B/genética , Subunidad p52 de NF-kappa B/genética , Subunidad p52 de NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Ubiquitina-Proteína Ligasas/genética , Quinasa de Factor Nuclear kappa B
6.
J Cell Biochem ; 112(6): 1643-50, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21344491

RESUMEN

Cucurbitacin B, a natural triterpenoid is well-known for its strong anticancer activity, and recent studies showed that the compound inhibits JAK/STAT3 pathway. In this study, we demonstrate for the first time that cucurbitacin B is also a potent inhibitor of NF-κB activation. Our results showed that cucurbitacin B inhibited TNF-α-induced expression of NF-κB reporter gene and NF-κB target genes in a dose-dependent manner, however, it did not prevent either stimuli-induced degradation of IκBα or nuclear translocation and DNA-binding activity of NF-κB. On the other hand, cucurbitacin B dose-dependently suppressed not only NF-κB activation induced by overexpression of RelA/p65 but also transactivation activity of RelA/p65 subunit of NF-κB. Consistently, treatment of HeLa cells with the compound significantly suppressed TNF-α-induced activation of Akt and phosphorylation of Ser536 in RelA/p65, which is required for transactivation activity. Consequently, cucurbitacin B inhibited TNF-α-induced expression of NF-κB-dependent anti-apoptotic proteins such as c-IAP1, c-IAP2, XIAP, TRAF1, and TRAF2 and sensitized TNF-α-induced cell death. Taken together, our results demonstrated that cucurbitacin B could be served as a valuable candidate for the intervention of NF-κB-dependent pathological condition such as cancer.


Asunto(s)
Factor de Transcripción ReIA/metabolismo , Triterpenos/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Supervivencia Celular/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética , Células HeLa , Humanos , Reacción en Cadena de la Polimerasa , Factor de Necrosis Tumoral alfa/farmacología
7.
Oncogene ; 40(2): 292-306, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33122824

RESUMEN

Overexpression of D-type cyclins in human cancer frequently occurs as a result of protein stabilization, emphasizing the importance of identification of the machinery that regulates their ubiqutin-dependent degradation. Cyclin D3 is overexpressed in ~50% of Burkitt's lymphoma correlating with a mutation of Thr-283. However, the E3 ligase that regulates phosphorylated cyclin D3 and whether a stabilized, phosphorylation deficient mutant of cyclin D3, has oncogenic activity are undefined. We describe the identification of SCF-Fbxl8 as the E3 ligase for Thr-283 phosphorylated cyclin D3. SCF-Fbxl8 poly-ubiquitylates p-Thr-283 cyclin D3 targeting it to the proteasome. Functional investigation demonstrates that Fbxl8 antagonizes cell cycle progression, hematopoietic cell proliferation, and oncogene-induced transformation through degradation of cyclin D3, which is abolished by expression of cyclin D3T283A, a non-phosphorylatable mutant. Clinically, the expression of cyclin D3 is inversely correlated with the expression of Fbxl8 in lymphomas from human patients implicating Fbxl8 functions as a tumor suppressor.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Linfoma de Burkitt/patología , Ciclina D3/metabolismo , Proteínas F-Box/metabolismo , Regulación Neoplásica de la Expresión Génica , Células Madre Hematopoyéticas/patología , Proteolisis , Animales , Apoptosis , Biomarcadores de Tumor/genética , Linfoma de Burkitt/genética , Linfoma de Burkitt/metabolismo , Ciclo Celular , Proliferación Celular , Ciclina D3/genética , Proteínas F-Box/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Sci Transl Med ; 13(615): eabh1486, 2021 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-34644148

RESUMEN

Discovery of small-molecule degraders that activate ubiquitin ligase­mediated ubiquitination and degradation of targeted oncoproteins in cancer cells has been an elusive therapeutic strategy. Here, we report a cancer cell­based drug screen of the NCI drug-like compounds library that enabled identification of small-molecule degraders of the small ubiquitin-related modifier 1 (SUMO1). Structure-activity relationship studies of analogs of the hit compound CPD1 led to identification of a lead compound HB007 with improved properties and anticancer potency in vitro and in vivo. A genome-scale CRISPR-Cas9 knockout screen identified the substrate receptor F-box protein 42 (FBXO42) of cullin 1 (CUL1) E3 ubiquitin ligase as required for HB007 activity. Using HB007 pull-down proteomics assays, we pinpointed HB007's binding protein as the cytoplasmic activation/proliferation-associated protein 1 (CAPRIN1). Biolayer interferometry and compound competitive immunoblot assays confirmed the selectivity of HB007's binding to CAPRIN1. When bound to CAPRIN1, HB007 induced the interaction of CAPRIN1 with FBXO42. FBXO42 then recruited SUMO1 to the CAPRIN1-CUL1-FBXO42 ubiquitin ligase complex, where SUMO1 was ubiquitinated in several of human cancer cells. HB007 selectively degraded SUMO1 in patient tumor­derived xenografts implanted into mice. Systemic administration of HB007 inhibited the progression of patient-derived brain, breast, colon, and lung cancers in mice and increased survival of the animals. This cancer cell­based screening approach enabled discovery of a small-molecule degrader of SUMO1 and may be useful for identifying other small-molecule degraders of oncoproteins.


Asunto(s)
Neoplasias , Proteína SUMO-1 , Animales , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Proteína SUMO-1/genética , Proteína SUMO-1/metabolismo , Ubiquitinación
9.
Biochem Biophys Res Commun ; 400(4): 581-6, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20804734

RESUMEN

LIGHT is a member of tumor necrosis factor (TNF) superfamily, and its function is mediated through lymphotoxin-ß receptor (LTßR), which is known to play important roles in inflammatory and immune responses through activation of NF-κB signaling pathways. However, molecular mechanism of LTßR ligation-induced NF-κB signaling remains incompletely understood. In this report we demonstrate that a novel zinc-finger protein 91 (ZFP91) is a critical regulator in LIGHT-induced activation of non-canonical NF-κB pathway. ZFP91 appears to be required for NF-κB2 (p100) processing to p52, nuclear translocation of p52 and RelB, and DNA-binding activity of NF-κB in LIGHT-induced activation of non-canonical NF-κB pathway. Furthermore, ZFP91 knock-down by RNA interference blocks the LIGHT-induced accumulation of NIK and p100 processing, as well as the expression of non-canonical NF-κB target genes. These data clearly indicate that ZFP91 is a key regulator in LIGHT-induced activation of non-canonical NF-κB pathway in LTßR signaling.


Asunto(s)
Regulación de la Expresión Génica , FN-kappa B/metabolismo , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Ligando de CD40/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Receptor beta de Linfotoxina/metabolismo , Subunidad p52 de NF-kappa B/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Factor de Transcripción ReIA/metabolismo , Factor de Transcripción ReIB/metabolismo , Ubiquitina-Proteína Ligasas/genética
10.
Cell Death Dis ; 10(4): 311, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952841

RESUMEN

Ginseng is one of the most commonly used herbs that is believed to have a variety of biological activities, including reducing blood sugar and cholesterol levels, anti-cancer, and anti-diabetes activities. However, little is known about the molecular mechanisms involved. In this study, we showed that protopanaxadiol (PPD), a metabolite of the protopanaxadiol group ginsenosides that are the major pharmacological constituents of ginsengs, significantly altered the expression of genes involved in metabolism, elevated Sestrin2 (Sesn2) expression, activated AMPK, and induced autophagy. Using CRISPR/CAS9-mediated gene editing and shRNA-mediated gene silencing, we demonstrated that Sesn2 is required for PPD-induced AMPK activation and autophagy. Interestingly, we showed that PPD-induced Sesn2 expression is mediated redundantly by the GCN2/ATF4 amino acid-sensing pathway and the PERK/ATF4 endoplasmic reticulum (ER) stress pathway. Our results suggest that ginseng metabolite PPD modulates the metabolism of amino acids and lipids, leading to the activation of the stress-sensing kinases GCN2 and PERK to induce Sesn2 expression, which promotes AMPK activation, autophagy, and metabolic health.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteínas Nucleares/metabolismo , Panax/química , Proteínas Serina-Treonina Quinasas/metabolismo , Sapogeninas/farmacología , eIF-2 Quinasa/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Factor de Transcripción Activador 4/genética , Factor de Transcripción Activador 4/metabolismo , Aminoácidos/metabolismo , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Estrés del Retículo Endoplásmico , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Ginsenósidos/farmacología , Células HCT116 , Células HEK293 , Humanos , Ratones , Proteínas Nucleares/genética , Panax/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal/genética , eIF-2 Quinasa/genética
11.
Eur J Pharmacol ; 592(1-3): 41-7, 2008 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-18639543

RESUMEN

Hypoxia-inducible factor-1 (HIF-1) is the central mediator of cellular responses to low oxygen and vital to many aspects of cancer biology. In a search for HIF-1 inhibitors, we identified a quassinoid 6alpha-tigloyloxychaparrinone (TCN) as an inhibitor of HIF-1 activation from Ailantus altissima. We here demonstrated the effect of TCN on HIF-1 activation induced by hypoxia or CoCl2. TCN showed the potent inhibitory activity against HIF-1 activation induced by hypoxia in various human cancer cell lines. This compound markedly decreased the hypoxia-induced accumulation of HIF-1alpha protein dose-dependently, whereas it did not affect the expressions of HIF-1beta and topoisomerase-I. Furthermore, TCN prevented hypoxia-induced expression of HIF-1 target genes for vascular endothelial growth factor (VEGF) and erythropoietin. Further analysis revealed that TCN strongly inhibited HIF-1alpha protein synthesis, without affecting the expression level of HIF-1alpha mRNA or degradation of HIF-1alpha protein. Moreover, the levels of phosphorylation of extracellular signal-regulated kinase-1/2 (ERK1/2), mitogen-activated protein (MAP) kinase-interacting protein kinase-1 (MNK1) and eukaryotic initiation factor 4E (eIF4E) were significantly suppressed by the treatment of TCN, without changing the total levels of these proteins. Our data suggested that TCN may exhibit anticancer activity by inhibiting HIF-1alpha translation through the inhibition of eIF4E phosphorylation pathway and thus provide a novel mechanism for the anticancer activity of quassinoids. TCN could be a new HIF-1-targeted anticancer agent and be effective on mammalian target of rapamycin (mTOR)-targeted cancer therapy, in which mTOR inhibition increases eIF4E phosphorylation.


Asunto(s)
Factor 4E Eucariótico de Iniciación/metabolismo , Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Cuassinas/farmacología , Transducción de Señal/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Ensayo de Inmunoadsorción Enzimática , Factor 4E Eucariótico de Iniciación/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Genes Reporteros/genética , Humanos , Luciferasas/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , Fosforilación/efectos de los fármacos , Proteínas Quinasas/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serina-Treonina Quinasas TOR , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
PLoS One ; 7(6): e40355, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768286

RESUMEN

BACKGROUND: Cryptopleurine, a phenanthroquinolizidine alkaloid, was known to exhibit anticancer activity; however, the underlying mechanism is poorly understood. Because the nuclear factor-κB (NF-κB) transcription factors control many physiological processes including inflammation, immunity, and development and progression of cancer, we investigated the effects of cryptopleurine on tumor necrosis factor alpha (TNF-α)-induced NF-κB activation pathway and on the expression of NF-κB-regulated gene products associated with many pathophysiological processes. METHODOLOGY AND PRINCIPAL FINDING: MDA-MB231, MDA-MB435, MCF-7, HEK293, RAW264.7 and Hep3B cells were used to examine cryptopleurine's effect on the NF-κB activation pathway. Major assays were promoter-reporter gene assay, electrophoretic mobility shift assay (EMSA), in vitro immune complex kinase assay, real-time PCR, Western blot analysis, and Matrigel invasion assay. Experiments documenting cell proliferation and apoptosis were analyzed by MTT method and flow cytometry, respectively. The results indicated that cryptopleurine suppressed the NF-κB activation through the inhibition of IκB kinase (IKK) activation, thereby blocking the phosphorylation and degradation of the inhibitor of NF-κB alpha (IκBα) and the nuclear translocation and DNA-binding activity of p65. The suppression of NF-κB by cryptopleurine led to the down-regulation of gene products involved in inflammation, cell survival, proliferation, invasion, and angiogenesis. CONCLUSIONS AND SIGNIFICANCE: Our results show that cryptopleurine inhibited NF-κB activation pathway, which leads to inhibition of inflammation, proliferation, and invasion, as well as potentiation of apoptosis. Our findings provide a new insight into the molecular mechanisms and a potential application of cryptopleurine for inflammatory diseases as well as certain cancers associated with abnormal NF-κB activation.


Asunto(s)
Alcaloides/farmacología , FN-kappa B/metabolismo , Invasividad Neoplásica/genética , Neovascularización Patológica/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Alcaloides/química , Alcaloides/uso terapéutico , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Citocinas/metabolismo , Expresión Génica/efectos de los fármacos , Genes Reporteros/genética , Humanos , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/metabolismo , Inflamación/genética , Inflamación/patología , Ratones , Inhibidor NF-kappaB alfa , Invasividad Neoplásica/patología , Neovascularización Patológica/tratamiento farmacológico , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
13.
Phytomedicine ; 17(6): 475-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19836221

RESUMEN

The protective effect of a diterpenoid acanthoic acid (AA) isolated from Acanthopanax koreanum Nakai was investigated in acetaminophen (APAP)-induced hepatic toxicity. Drug-induced hepatotoxicity induced by an intraperitoneal (i.p.) injection of 300mg/kg (sub-lethal dose) of APAP. Pretreatment with AA (50 and 100mg/kg) orally 2h before the APAP administration attenuated the APAP-induced acute increase in serum aspartate aminotransferase (AST), and alanine aminotransferase (ALT) activites, replenished the depleted hepatic glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px) activities, decreased malondialdehyde (MDA) level and considerably reduced the histopathological alterations in a manner similar to silymarin (Sily). Immunohistochemical analyses also demonstrated that AA could reduce the appearance of necrosis regions as well as caspase-3 and hypoxia inducible factor-1alpha (HIF-1alpha) expression in liver tissue. Our results indicated that AA protected liver tissue from the oxidative stress elicites by APAP-induced liver damage and suggestes that the hepatic protection mechanism of AA would relate to antioxidation and hypoxia factor on APAP-induced hepatotoxicity.


Asunto(s)
Antioxidantes/uso terapéutico , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Diterpenos/uso terapéutico , Eleutherococcus/química , Hígado/efectos de los fármacos , Fitoterapia , Extractos Vegetales/uso terapéutico , Acetaminofén , Animales , Antioxidantes/farmacología , Caspasa 3/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Diterpenos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hígado/enzimología , Hígado/patología , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Necrosis/prevención & control , Estrés Oxidativo/efectos de los fármacos , Corteza de la Planta , Extractos Vegetales/farmacología , Raíces de Plantas
14.
Biol Pharm Bull ; 31(4): 738-42, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18379074

RESUMEN

The hepatoprotective effects of a diterpenoid acanthoic acid isolated from Acanthopanax koreanum NAKAI were evaluated in a D-galactosamine/lipopolysaccharide-induced fulminant hepatic failure mouse model. Mice were pretreated orally with acanthoic acid 12 and 1 h before intraperitoneal injection of D-galactosamine and lipopolysaccharide. Pretreatment with the compound markedly reduced lethal liver injury in experimental animals. The effects were likely associated with a significant decrease in serum tumor necrosis factor (TNF)-alpha levels, which are correlated not only with those of alanine aminotransferase and aspartate aminotransferase but also with the reduced number of apoptotic hepatocytes in the liver as confirmed using the terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling method and DNA fragmentation assay. These results suggest that acanthoic acid protects against D-galactosamine/lipopolysaccharide-induced fulminant liver failure at least in part by a mechanism associated with the down-regulation of TNF-alpha secretion.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Diterpenos/farmacología , Eleutherococcus/química , Fallo Hepático Agudo/prevención & control , Alanina Transaminasa/sangre , Animales , Antiinflamatorios no Esteroideos/aislamiento & purificación , Antiinflamatorios no Esteroideos/toxicidad , Aspartato Aminotransferasas/sangre , Fragmentación del ADN/efectos de los fármacos , Diterpenos/aislamiento & purificación , Diterpenos/toxicidad , Galactosamina , Etiquetado Corte-Fin in Situ , Lipopolisacáridos , Fallo Hepático Agudo/inducido químicamente , Masculino , Ratones , Ratones Endogámicos C57BL , Raíces de Plantas/química , Factor de Necrosis Tumoral alfa/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA