Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
J Clin Invest ; 134(2)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-37988169

RESUMEN

Alzheimer's disease is characterized by the accumulation of amyloid-ß plaques, aggregation of hyperphosphorylated tau (pTau), and microglia activation. Galectin-3 (Gal3) is a ß-galactoside-binding protein that has been implicated in amyloid pathology. Its role in tauopathy remains enigmatic. Here, we showed that Gal3 was upregulated in the microglia of humans and mice with tauopathy. pTau triggered the release of Gal3 from human induced pluripotent stem cell-derived microglia in both its free and extracellular vesicular-associated (EV-associated) forms. Both forms of Gal3 increased the accumulation of pathogenic tau in recipient cells. Binding of Gal3 to pTau greatly enhanced tau fibrillation. Besides Gal3, pTau was sorted into EVs for transmission. Moreover, pTau markedly enhanced the number of EVs released by iMGL in a Gal3-dependent manner, suggesting a role of Gal3 in biogenesis of EVs. Single-cell RNA-Seq analysis of the hippocampus of a mouse model of tauopathy (THY-Tau22) revealed a group of pathogenic tau-evoked, Gal3-associated microglia with altered cellular machineries implicated in neurodegeneration, including enhanced immune and inflammatory responses. Genetic removal of Gal3 in THY-Tau22 mice suppressed microglia activation, reduced the level of pTau and synaptic loss in neurons, and rescued memory impairment. Collectively, Gal3 is a potential therapeutic target for tauopathy.


Asunto(s)
Galectina 3 , Tauopatías , Proteínas tau , Animales , Humanos , Ratones , Enfermedad de Alzheimer/patología , Modelos Animales de Enfermedad , Galectina 3/genética , Galectina 3/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Ratones Transgénicos , Microglía/patología , Proteínas tau/genética , Proteínas tau/metabolismo , Tauopatías/genética , Tauopatías/metabolismo
4.
Commun Biol ; 6(1): 767, 2023 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-37479809

RESUMEN

Abnormal polyglutamine (polyQ) expansion and fibrillization occur in Huntington's disease (HD). Amyloid modifier SERF enhances amyloid formation, but the underlying mechanism is not revealed. Here, the fibrillization and toxicity effect of SERF1a on Htt-exon1 are examined. SERF1a enhances the fibrillization of and interacts with mutant thioredoxin (Trx)-fused Httex1. NMR studies with Htt peptides show that TrxHttex1-39Q interacts with the helical regions in SERF1a and SERF1a preferentially interacts with the N-terminal 17 residues of Htt. Time-course analysis shows that SERF1a induces mutant TrxHttex1 to a single conformation enriched of ß-sheet. Co-expression of SERF1a and Httex1-polyQ in neuroblastoma and lentiviral infection of SERF1a in HD-induced polypotent stem cell (iPSC)-derived neurons demonstrates the detrimental effect of SERF1a in HD. Higher level of SERF1a transcript or protein is detected in HD iPSC, transgenic mice, and HD plasma. Overall, this study provides molecular mechanism for SERF1a and mutant Httex1 to facilitate therapeutic development for HD.


Asunto(s)
Proteínas Amiloidogénicas , Enfermedad de Huntington , Animales , Ratones , Péptidos/genética , Factores de Transcripción , Exones , Enfermedad de Huntington/genética , Ratones Transgénicos
5.
Mov Disord ; 37(10): 2008-2020, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35997316

RESUMEN

BACKGROUND: Huntington's disease (HD) is a neurodegenerative disease caused by CAG-repeat expansions (>36) in exon 1 of HTT, which dysregulates multiple cellular machineries. Translin-associated protein X (TRAX) is a scaffold protein with diverse functions, including suppressing the microRNA (miRNA)-mediated silencing by degrading pre-miRNA. To date, the role of TRAX in neurodegenerative diseases remains unknown. OBJECTIVES: We delineated the role of TRAX upregulation during HD progression. METHODS: Expression of TRAX in the brains of humans and three mouse models with HD were analyzed by immunohistochemistry staining, western blot, and quantitative reverse transcription-polymerase chain reaction. Adeno-associated viruses harboring TRAX short hairpin RNA were intrastriatally injected into HD mice to downregulate TRAX. HD-like symptoms were analyzed by behavioral and biochemical assessments. The miRNA-sequencing and RNA-sequencing analyses were used to identify the TRAX- regulated miRNA-messenger RNA (mRNA) axis during HD progression. The identified gene targets were validated biochemically in mouse and human striatal cells. RESULTS: We discovered that TRAX was upregulated in the brains of HD patients and three HD mouse models. Downregulation of TRAX enhanced 83 miRNAs (including miR-330-3p, miR-496a-3p) and subsequently changed the corresponding mRNA networks critical for HD pathogenesis (eg, DARPP-32 and brain-derived neurotrophic factor). Disruption of the TRAX-mediated miRNA-mRNA axis accelerated the progression of HD-like symptoms, including the degeneration of motor function, accumulation of mHTT aggregates, and shortened neurite outgrowth. CONCLUSIONS: We demonstrated that TRAX upregulation is authentic and protective in HD. Our study provides a novel layer of regulation for HD pathogenesis and may lead to the development of new therapeutic strategies for HD. © 2022 International Parkinson and Movement Disorder Society.


Asunto(s)
Enfermedad de Huntington , MicroARNs , Enfermedades Neurodegenerativas , Animales , Humanos , Ratones , Factor Neurotrófico Derivado del Encéfalo , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Enfermedad de Huntington/metabolismo , MicroARNs/genética , Neuroprotección , ARN Mensajero , ARN Interferente Pequeño
6.
Open Biol ; 9(1): 180177, 2019 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-30958120

RESUMEN

Adult-onset neurodegenerative diseases are among the most difficult human health conditions to model for drug development. Most genetic or toxin-induced cell and animal models cannot faithfully recapitulate pathology in disease-relevant cells, making it excessively challenging to explore the potential mechanisms underlying sporadic disease. Patient-derived induced pluripotent stem cells (iPSCs) can be differentiated into disease-relevant neurons, providing an unparalleled platform for in vitro modelling and development of therapeutic strategies. Here, we review recent progress in generating Alzheimer's, Parkinson's and Huntington's disease models from patient-derived iPSCs. We also describe novel discoveries of pathological mechanisms and drug evaluations that have used these patient iPSC-derived neuronal models. Additionally, current human iPSC technology allows researchers to model diseases with 3D brain organoids, which are more representative of tissue architecture than traditional neuronal cultures. We discuss remaining challenges and emerging opportunities for the use of three-dimensional brain organoids in modelling brain development and neurodegeneration.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas/citología , Modelos Neurológicos , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Enfermedad de Alzheimer/patología , Animales , Humanos , Enfermedad de Huntington/patología , Enfermedad de Parkinson/patología
7.
PLoS Pathog ; 14(7): e1007166, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30016363

RESUMEN

CCCH-type zinc-finger antiviral protein (ZAP) is a host factor that restricts the infection of many viruses mainly through RNA degradation, translation inhibition and innate immune responses. So far, only one flavivirus, yellow fever virus, has been reported to be ZAP-resistant. Here, we investigated the antiviral potential of human ZAP (isoform ZAP-L and ZAP-S) against three flaviviruses, Japanese encephalitis virus (JEV), dengue virus (DENV) and Zika virus (ZIKV). Infection of JEV but not DENV or ZIKV was blocked by ZAP overexpression, and depletion of endogenous ZAP enhanced JEV replication. ZAP hampered JEV translation and targeted viral RNA for 3'-5' RNA exosome-mediated degradation. The zinc-finger motifs of ZAP were essential for RNA targeting and anti-JEV activity. JEV 3'-UTR, especially in the region with dumbbell structures and high content of CG dinucleotide, was mapped to bind ZAP and confer sensitivity to ZAP. In summary, we identified JEV as the first ZAP-sensitive flavivirus. ZAP may act as an intrinsic antiviral factor through specific RNA binding to fight against JEV infection.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie)/inmunología , Encefalitis Japonesa/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/inmunología , Proteínas de Unión al ARN/inmunología , Humanos
8.
Mol Psychiatry ; 23(12): 2375-2390, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-29298990

RESUMEN

Translin-associated protein X (TRAX) is a scaffold protein with various functions and has been associated with mental illnesses, including schizophrenia. We have previously demonstrated that TRAX interacts with a Gsα protein-coupled receptor, the A2A adenosine receptor (A2AR), and mediates the function of this receptor in neuritogenesis. In addition, stimulation of the A2AR markedly ameliorates DNA damage evoked by elevated oxidative stress in neurons derived from induced pluripotent stem cells (iPSCs). Here, we report that glycogen synthase kinase 3 beta (GSK3ß) and disrupted-in-schizophrenia 1 (DISC1) are two novel interacting proteins of TRAX. We present evidence to suggest that the stimulation of A2AR markedly facilitated DNA repair through the TRAX/DISC1/GSK3ß complex in a rat neuronal cell line (PC12), primary mouse neurons, and human medium spiny neurons derived from iPSCs. A2AR stimulation led to the inhibition of GSK3ß, thus dissociating the TRAX/DISC1/GSK3ß complex and facilitating the non-homologous end-joining pathway (NHEJ) by enhancing the activation of a DNA-dependent protein kinase via phosphorylation at Thr2609. Similarly, pharmacological inhibition of GSK3ß by SB216763 also facilitated the TRAX-mediated repair of oxidative DNA damage. Collectively, GSK3ß binds with TRAX and negatively affects its ability to facilitate NHEJ repair. The suppression of GSK3ß by A2AR activation or a GSK3ß inhibitor releases TRAX for the repair of oxidative DNA damage. Our findings shed new light on the molecular mechanisms underlying diseases associated with DNA damage and provides a novel target (i.e., the TRAX/DISC1/GSK3ß complex) for future therapeutic development for mental disorders.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Receptor de Adenosina A2A/metabolismo , Animales , Proteínas Portadoras/genética , Reparación del ADN , Proteínas de Unión al ADN/genética , Glucógeno Sintasa Quinasa 3 beta/genética , Glucógeno Sintasa Quinasa 3 beta/fisiología , Hipocampo/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuritas , Neuronas/metabolismo , Células PC12 , Fosforilación , Ratas , Receptor de Adenosina A2A/genética , Transducción de Señal
9.
Hum Mol Genet ; 24(21): 6066-79, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26264576

RESUMEN

Huntington's disease (HD) is an autosomal-dominant degenerative disease caused by a cytosine-adenine-guanine trinucleotide expansion in the Huntingtin (htt) gene. The most vulnerable brain areas to mutant HTT-evoked toxicity are the striatum and cortex. In spite of the extensive efforts that have been devoted to the characterization of HD pathogenesis, no disease-modifying therapy for HD is currently available. The A2A adenosine receptor (A2AR) is widely distributed in the brain, with the highest level observed in the striatum. We previously reported that stimulation of the A2AR triggers an anti-apoptotic effect in a rat neuron-like cell line (PC12). Using a transgenic mouse model (R6/2) of HD, we demonstrated that A2AR-selective agonists effectively ameliorate several major symptoms of HD. In the present study, we show that human iPSCs can be successfully induced to differentiate into DARPP32-positive, GABAergic neurons which express the A2AR in a similar manner to striatal medium spiny neurons. When compared with those derived from control subjects (CON-iPSCs), these HD-iPSC-derived neurons exhibited a higher DNA damage response, based on the observed expression of γH2AX and elevated oxidative stress. This is a critical observation, because oxidative damage and abnormal DNA damage/repair have been reported in HD patients. Most importantly, stimulation of the A2AR using selective agonists reduced DNA damage and oxidative stress-induced apoptosis in HD-iPSC-derived neurons through a cAMP/PKA-dependent pathway. These findings support our hypothesis that human neurons derived from diseased iPSCs might serve as an important platform to investigate the beneficial effects and underlying mechanisms of A2AR drugs.


Asunto(s)
Neuronas GABAérgicas/patología , Enfermedad de Huntington/patología , Degeneración Nerviosa , Células Madre Pluripotentes/patología , Receptor de Adenosina A2A/metabolismo , Adulto , Apoptosis , Caspasa 3/metabolismo , Línea Celular , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Daño del ADN , Fosfoproteína 32 Regulada por Dopamina y AMPc/biosíntesis , Femenino , Neuronas GABAérgicas/metabolismo , Humanos , Proteína Huntingtina , Enfermedad de Huntington/genética , Peróxido de Hidrógeno , Recién Nacido , Masculino , Persona de Mediana Edad , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Células Madre Pluripotentes/metabolismo , Adulto Joven
10.
Ann Neurol ; 78(2): 178-92, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25914140

RESUMEN

OBJECTIVE: Huntington disease (HD) is an inherited neurodegenerative disease caused by the mutant huntingtin gene (mHTT), which harbors expanded CAG repeats. We previously reported that the brain vessel density is higher in mice and patients with HD than in controls. The present study determines whether vascular function is altered in HD and characterizes the underlying mechanism. METHODS: The brain vessel density and vascular reactivity (VR) to carbogen challenge of HD mice were monitored by 3D ΔR2 -mMRA and blood oxygenation level-dependent (BOLD)/flow-sensitive alternating inversion recovery (FAIR) magnetic resonance imaging (MRI), respectively. The amount of vascular endothelial growth factor (VEGF)-A and the pericyte coverage were determined by immunohistochemistry and enzyme-linked immunosorbent assay in human and mouse brain sections, primary mouse astrocytes and pericytes, and human astrocytes derived from induced pluripotent stem cells. RESULTS: Expression of mHTT in astrocytes and neurons is sufficient to increase the brain vessel density in HD mice. BOLD and FAIR MRI revealed gradually impaired VR to carbogen in HD mice. Astrocytes from HD mice and patients contained more VEGF-A, which triggers proliferation of endothelial cells and may be responsible for the augmented neurovascular changes. Moreover, an astrocytic inflammatory response, which reduces the survival of pericytes through an IκB kinase-dependent pathway, mediates the low pericyte coverage of blood vessels in HD brains. INTERPRETATION: Our findings suggest that the inflammation-prone HD astrocytes provide less pericyte coverage by promoting angiogenesis and reducing the number of pericytes and that these changes can explain the inferior VR in HD mice. The resultant impaired VR might hinder cerebral hemodynamics and increase brain atrophy during HD progression.


Asunto(s)
Astrocitos/metabolismo , Vasos Sanguíneos/metabolismo , Encéfalo/irrigación sanguínea , Enfermedad de Huntington/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Nucleares/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Adulto , Anciano , Animales , Astrocitos/patología , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Femenino , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Enfermedad de Huntington/fisiopatología , Células Madre Pluripotentes Inducidas/metabolismo , Angiografía por Resonancia Magnética , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Pericitos/patología
11.
Hum Mol Genet ; 23(16): 4328-44, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24698979

RESUMEN

Neuroinflammation is a common feature of many neurodegenerative diseases, including Huntington's disease (HD). HD is an autosomal dominant genetic disease caused by an expanded CAG repeat in exon 1 of the huntingtin (HTT) gene. Previous studies demonstrated that levels of several proinflammatory cytokines, including tumor necrosis factor (TNF)-α, were higher in the plasma and brain tissues of mice and patients with HD, suggesting that inflammation may contribute to HD progression. To evaluate the pathological role of TNF-α in HD pathogenesis, we blocked TNF-α signaling using a dominant negative inhibitor of soluble TNF-α (XPro1595). XPro1595 effectively suppressed the inflammatory responses of primary astrocytes-enriched culture isolated from a transgenic mouse model (R6/2) and human astrocytes-enriched culture derived from induced pluripotent stem cells (iPSCs) of HD patients evoked by lipopolysaccharide and cytokines, respectively. Moreover, XPro1595 protected the cytokine-induced toxicity of primary R6/2 neurons and human neurons derived from iPSCs of HD patients. To assess the beneficial effect of XPro1595 in vivo, an intracerebroventricular (i.c.v.) infusion was provided with an osmotic minipump. ELISA analyses showed that i.c.v. infusion of XPro1595 decreased elevated levels of TNFα in the cortex and striatum, improved motor function, reduced caspase activation, diminished the amount of mutant HTT aggregates, increased neuronal density and decreased gliosis in brains of R6/2 mice. Moreover, reducing the peripheral inflammatory response by a systemic injection of XPro1595 improved the impaired motor function of R6/2 mice but did not affect caspase activation. Collectively, our findings suggest that an effective and selective anti-inflammatory treatment targeting the abnormal brain inflammatory response is a potential therapeutic strategy for HD.


Asunto(s)
Antiinflamatorios/uso terapéutico , Enfermedad de Huntington/tratamiento farmacológico , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Animales Recién Nacidos , Antiinflamatorios/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Supervivencia Celular/efectos de los fármacos , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Humanos , Enfermedad de Huntington/metabolismo , Inflamación/tratamiento farmacológico , Infusiones Intraventriculares , Ratones , Ratones Transgénicos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Células Madre Pluripotentes , Cultivo Primario de Células , Factor de Necrosis Tumoral alfa/farmacología , Factor de Necrosis Tumoral alfa/uso terapéutico
12.
Stem Cell Reports ; 2(2): 189-204, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24527393

RESUMEN

The mechanisms of transcriptional regulation underlying human primordial germ cell (PGC) differentiation are largely unknown. The transcriptional repressor Prdm1/Blimp-1 is known to play a critical role in controlling germ cell specification in mice. Here, we show that PRDM1 is expressed in developing human gonads and contributes to the determination of germline versus neural fate in early development. We show that knockdown of PRDM1 in human embryonic stem cells (hESCs) impairs germline potential and upregulates neural genes. Conversely, ectopic expression of PRDM1 in hESCs promotes the generation of cells that exhibit phenotypic and transcriptomic features of early PGCs. Furthermore, PRDM1 suppresses transcription of SOX2. Overexpression of SOX2 in hESCs under conditions favoring germline differentiation skews cell fate from the germline to the neural lineage. Collectively, our results demonstrate that PRDM1 serves as a molecular switch to modulate the divergence of neural or germline fates through repression of SOX2 during human development.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Células Germinativas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción SOXB1/genética , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular/genética , Feto/embriología , Feto/metabolismo , Gónadas/embriología , Gónadas/metabolismo , Humanos , Modelos Biológicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Represoras/genética , Proteína Wnt3A/metabolismo
13.
FEBS Lett ; 582(16): 2407-12, 2008 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-18544347

RESUMEN

We use the LPS-stimulated macrophage as a model of inflammation to investigate the anti-inflammatory effects of tomatidine and solasodine, whose structures resemble glucocorticoids. We found that tomatidine exhibited a more potent anti-inflammatory effect than solasodine. Tomatidine could decrease inducible nitric oxide synthase and cyclooxygenase-2 expression through suppression of I-kappaBalpha phosphorylation, NF-kappaB nuclear translocation and JNK activation, which in turn inhibits c-jun phosphorylation and Oct-2 expression. Here, we demonstrate that tomatidine acts as an anti-inflammatory agent by blocking NF-kappaB and JNK signaling, and may possibly be developed as a useful agent for the chemoprevention of cancer or inflammatory diseases.


Asunto(s)
Antiinflamatorios/farmacología , Ciclooxigenasa 2/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Macrófagos/inmunología , FN-kappa B/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo II/biosíntesis , Tomatina/análogos & derivados , Animales , Antiinflamatorios/química , Línea Celular , Ciclooxigenasa 2/genética , Dinoprostona/biosíntesis , Diosgenina/química , Diosgenina/farmacología , Proteínas I-kappa B/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Inhibidor NF-kappaB alfa , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/genética , Factor 2 de Transcripción de Unión a Octámeros/biosíntesis , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal , Alcaloides Solanáceos/química , Alcaloides Solanáceos/farmacología , Tomatina/química , Tomatina/farmacología
14.
Prostate ; 68(1): 61-71, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18008333

RESUMEN

BACKGROUND: Androgen receptor (ARs) play a crucial role in the development and progression of prostate cancer. Recent studies have suggested that prostate cancer cell proliferation is inhibited by AR downregulation. Our aim was to investigate how luteolin, a natural flavonoid, affects cell growth and AR expression in prostate cancer cells and xenografts. METHODS: We assessed prostate cancer cell (LNCaP, DU145, and PC-3) proliferation and apoptosis by MTT assay, flow cytometric analysis, and Western analysis. AR function was measured by evaluating the AR target molecule, prostate-specific antigen (PSA), by RT-PCR, Western blotting, and enzyme-linked immunosorbent assay. We determined the mechanism of AR downregulation with cycloheximide chase assays, proteasome inhibitor, and coimmunoprecipitation experiments. The effects of luteolin on growth inhibition in vivo were examined by LNCaP xenografts in SCID mice. RESULTS: Luteolin significantly repressed prostate cancer cell proliferation and induced apoptosis in LNCaP cells. PC-3 and DU145 cells were less susceptible to luteolin-mediated growth inhibition. Luteolin simultaneously suppressed intracellular and secreted PSA levels and repressed AR mRNA and protein expression in a dose- and time-dependent manner. Luteolin reduced the association between AR and heat-shock protein 90, causing AR degradation through a proteasome-mediated pathway in a ligand-independent manner. Luteolin also suppressed LNCaP xenograft tumor growth in SCID mice. CONCLUSION: Luteolin-mediated AR downregulation contributes to the inhibition of cell proliferation and the induction of apoptosis in LNCaP human prostate cancer cells, suggesting that AR is a molecular target for luteolin-mediated anticancer activity. Luteolin may act as a chemopreventive or chemotherapeutic agent for prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Luteolina/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Animales , División Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Fase G1/efectos de los fármacos , Fase G2/efectos de los fármacos , Humanos , Masculino , Ratones , Ratones SCID , Receptores Androgénicos/metabolismo , Fase S/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Agric Food Chem ; 53(18): 7035-42, 2005 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-16131108

RESUMEN

(-)-Epigallocatechin-3-gallate (EGCG), a major polyphenol of green tea, undergoes substantial biotransformation to species that includes the methylated compounds. Recent studies have demonstrated that the methylated EGCG has many biological activities. In this study, we have investigated the composition of the three O-methylated EGCG derivatives, (-)-epigallocatechin-3-O-(3-O-methyl)gallate (3' '-Me-EGCG), (-)-epigallocatechin-3-O-(4-O-methyl)gallate (4' '-Me-EGCG) and (-)-4'-methyl epigallocatechin-3-O-(4-O-methyl)gallate (4',4' '-di-Me-EGCG) in tea leaves which were picked from various species and at various seasons, ages of leaves, locations, and fermentation levels. Higher levels of 3' '-Me-EGCG and 4' '-Me-EGCG were detected in Chinshin-Kanzai (a species of Camellia sinensis) cultivated in the mountain area of Sansia, Taipei County, Taiwan. Also, these O-methylated EGCG levels were found to be higher in autumn and winter than in spring and summer. The young leaves were found to be richer in the O-methylated compounds than old leaves and the amount of O-methylated EGCG was higher in unfermented longjin green tea than in semifermented oolong tea. However, the fermented black tea and puerh tea did not contain these compounds. 4',4' '-diMe-EGCG could not be detected in either fresh tea leaves or commercial tea leaves. We also found that 3' '-Me-EGCG has a higher inhibitory effect on the nitric oxide generation and inducible nitric oxide synthase (iNOS) expression as compared with EGCG, while 4' '-Me-EGCG and 4',4' '-di-Me-EGCG were less effective.


Asunto(s)
Camellia sinensis/química , Catequina/análogos & derivados , Hojas de la Planta/química , Catequina/análisis , Fermentación , Metilación , Estaciones del Año , Taiwán
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...