Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Cancer Res ; 82(16): 2874-2886, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35731926

RESUMEN

Intratumor epigenetic heterogeneity is emerging as a key mechanism underlying tumor evolution and drug resistance. Epigenetic abnormalities frequently occur in medulloblastoma, the most common childhood malignant brain tumor. Medulloblastoma is classified into four subtypes including SHH medulloblastoma, which is characterized by elevated sonic hedgehog (SHH) signaling and a cerebellum granule neuron precursor (CGNP) cell-of-origin. Here, we report that the histone H3K27 methyltransferase polycomb repressor complex 2 (PRC2) is often heterogeneous within individual SHH medulloblastoma tumors. In mouse models, complete deletion of the PRC2 core subunit EED inhibited medulloblastoma growth, while a mosaic deletion of EED significantly enhanced tumor growth. EED is intrinsically required for CGNP maintenance by inhibiting both neural differentiation and cell death. Complete deletion of EED led to CGNP depletion and reduced occurrence of medulloblastoma. Surprisingly, medulloblastomas with mosaic EED levels grew faster than control wild-type tumors and expressed increased levels of oncogenes such as Igf2, which is directly repressed by PRC2 and has been demonstrated to be both necessary and sufficient for SHH medulloblastoma progression. Insulin-like growth factor 2 (IGF2) mediated the oncogenic effects of PRC2 heterogeneity in tumor growth. Assessing clones of a human medulloblastoma cell line with different EED levels confirmed that EEDlow cells can stimulate the growth of EEDhigh cells through paracrine IGF2 signaling. Thus, PRC2 heterogeneity plays an oncogenic role in medulloblastoma through both intrinsic growth competence and non-cell autonomous mechanisms in distinct tumor subclones. SIGNIFICANCE: The identification of an oncogenic function of PRC2 heterogeneity in medulloblastoma provides insights into subclone competition and cooperation during heterogeneous tumor evolution.


Asunto(s)
Neoplasias Cerebelosas , Proteínas de Drosophila , Meduloblastoma , Animales , Neoplasias Cerebelosas/patología , Cerebelo , Niño , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Meduloblastoma/patología , Ratones , Proteínas del Grupo Polycomb/metabolismo , Transducción de Señal/fisiología
2.
Cell Rep ; 36(2): 109357, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34260936

RESUMEN

Neuronal activity-induced enhancers drive gene activation. We demonstrate that BRG1, the core subunit of SWI/SNF-like BAF ATP-dependent chromatin remodeling complexes, regulates neuronal activity-induced enhancers. Upon stimulation, BRG1 is recruited to enhancers in an H3K27Ac-dependent manner. BRG1 regulates enhancer basal activities and inducibility by affecting cohesin binding, enhancer-promoter looping, RNA polymerase II recruitment, and enhancer RNA expression. We identify a serine phosphorylation site in BRG1 that is induced by neuronal stimulations and is sensitive to CaMKII inhibition. BRG1 phosphorylation affects its interaction with several transcription co-factors, including the NuRD repressor complex and cohesin, possibly modulating BRG1-mediated transcription outcomes. Using mice with knockin mutations, we show that non-phosphorylatable BRG1 fails to efficiently induce activity-dependent genes, whereas phosphomimic BRG1 increases enhancer activity and inducibility. These mutant mice display anxiety-like phenotypes and altered responses to stress. Therefore, we reveal a mechanism connecting neuronal signaling to enhancer activities through BRG1 phosphorylation.


Asunto(s)
ADN Helicasas/genética , ADN Helicasas/metabolismo , Elementos de Facilitación Genéticos/genética , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Acetilación , Secuencia de Aminoácidos , Animales , Ansiedad/patología , Conducta Animal , Encéfalo/patología , ADN Helicasas/química , Células HEK293 , Histonas/metabolismo , Humanos , Lisina/metabolismo , Ratones Endogámicos C57BL , Mutación/genética , Proteínas Nucleares/química , Fosforilación , Fosfoserina/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-fos/metabolismo , Estrés Psicológico/complicaciones , Factores de Transcripción/química , alfa-Fetoproteínas/metabolismo
3.
Nat Commun ; 12(1): 2954, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34012010

RESUMEN

How cancer cells cope with high levels of replication stress during rapid proliferation is currently unclear. Here, we show that macrophage migration inhibitory factor (MIF) is a 3' flap nuclease that translocates to the nucleus in S phase. Poly(ADP-ribose) polymerase 1 co-localizes with MIF to the DNA replication fork, where MIF nuclease activity is required to resolve replication stress and facilitates tumor growth. MIF loss in cancer cells leads to mutation frequency increases, cell cycle delays and DNA synthesis and cell growth inhibition, which can be rescued by restoring MIF, but not nuclease-deficient MIF mutant. MIF is significantly upregulated in breast tumors and correlates with poor overall survival in patients. We propose that MIF is a unique 3' nuclease, excises flaps at the immediate 3' end during DNA synthesis and favors cancer cells evading replication stress-induced threat for their growth.


Asunto(s)
Neoplasias de la Mama/metabolismo , Replicación del ADN/fisiología , Endonucleasas de ADN Solapado/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , ADN/química , ADN/metabolismo , Daño del ADN , ADN Polimerasa III/genética , ADN Polimerasa III/metabolismo , Replicación del ADN/genética , Femenino , Endonucleasas de ADN Solapado/deficiencia , Endonucleasas de ADN Solapado/genética , Técnicas de Inactivación de Genes , Inestabilidad Genómica , Células HCT116 , Humanos , Oxidorreductasas Intramoleculares/deficiencia , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/deficiencia , Factores Inhibidores de la Migración de Macrófagos/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Conformación de Ácido Nucleico , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Fase S , Especificidad por Sustrato
4.
J Biol Chem ; 294(14): 5549-5561, 2019 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-30782847

RESUMEN

In Sonic hedgehog (SHH) signaling, GLI family zinc finger (GLI)-mediated diverse gene transcription outcomes are strictly regulated and are important for SHH function in both development and disease. However, how the GLI factors differentially regulate transcription in response to variable SHH activities is incompletely understood. Here, using a newly generated, tagged Gli3 knock-in mouse (Gli3TAP ), we performed proteomic analyses and identified the chromatin-associated SAFB-like transcription modulator (SLTM) as a GLI-interacting protein that context-dependently regulates GLI activities. Using immunoprecipitation and immunoblotting, RT-quantitative PCR, and ChIP assays, we show that SLTM interacts with all three GLI proteins and that its cellular levels are regulated by SHH. We also found that SLTM enhances GLI3 binding to chromatin and increases GLI3 repressor (GLI3R) form protein levels. In a GLI3-dependent manner, SLTM promoted the formation of a repressive chromatin environment and functioned as a GLI3 co-repressor. In the absence of GLI3 or in the presence of low GLI3 levels, SLTM co-activated GLI activator (GLIA)-mediated target gene activation and cell differentiation. Moreover, in vivo Sltm deletion generated through CRISPR/Cas9-mediated gene editing caused perinatal lethality and SHH-related abnormal ventral neural tube phenotypes. We conclude that SLTM regulates GLI factor binding to chromatin and contributes to the transcriptional outcomes of SHH signaling via a novel molecular mechanism.


Asunto(s)
Proteínas Hedgehog/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Transducción de Señal , Proteína Gli3 con Dedos de Zinc/metabolismo , Animales , Sistemas CRISPR-Cas , Cromatina , Edición Génica , Técnicas de Sustitución del Gen , Proteínas Hedgehog/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas Asociadas a Matriz Nuclear/genética , Proteómica , Proteína Gli3 con Dedos de Zinc/genética
5.
BMB Rep ; 52(8): 490-495, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30673584

RESUMEN

Using tunneling nanotubes (TNTs), various pathological molecules and viruses disseminate to adjacent cells intercellularly. Here, we show that the intracellular invasion of Mycoplasma hyorhinis induces the formation of actin- and tubulin-based TNTs in various mammalian cell lines. M. hyorhinis was found in TNTs generated by M. hyorhinis infection in NIH3T3 cells. Because mycoplasma-free recipient cells received mycoplasmas from M. hyorhinis-infected donor cells in a mixed co-culture system and not a spatially separated co-culture system, direct cell-to-cell contact via TNTs was necessary for the intracellular dissemination of M. hyorhinis. The activity of Rac1, which is a small GTP binding protein, was increased by the intracellular invasion of M. hyorhinis, and its pharmacological and genetic inhibition prevented M. hyorhinis infection-induced TNT generation in NIH3T3 cells. The pharmacological and genetic inhibition of Rac1 also reduced the cell-to-cell dissemination of M. hyorhinis. Based on these data, we conclude that intracellular invasion of M. hyorhinis induces the formation of TNTs, which are used for the cell-to-cell dissemination of M. hyorhinis. [BMB Reports 2019; 52(8): 490-495].


Asunto(s)
Mycoplasma hyorhinis/metabolismo , Nanotubos/microbiología , Animales , Comunicación Celular , Ratones , Células 3T3 NIH
6.
Exp Mol Med ; 50(6): 1-12, 2018 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-29884820

RESUMEN

Mitsugumin 53 (MG53) is an E3 ligase that induces insulin receptor substrate-1 (IRS-1) ubiquitination and degradation in skeletal muscle. We previously demonstrated that the pharmaceutical disruption of the MG53-IRS-1 interaction improves insulin sensitivity by abrogating IRS-1 ubiquitination and increasing IRS-1 levels in C2C12 myotubes. Here, we developed a novel MG53-IRS-1 interaction disruptor (MID-00935) that ameliorates insulin resistance in diet-induced obese (DIO) mice. MID-00935 disrupted the molecular interaction of MG53 and IRS-1, abrogated MG53-induced IRS-1 ubiquitination and degradation and improved insulin signaling in C2C12 myotubes. Oral administration of MID-00935 increased insulin-induced IRS-1, Akt, and Erk phosphorylation via increasing IRS-1 levels in the skeletal muscle of DIO mice. In DIO mice, MID-00935 treatment lowered fasting blood glucose levels and improved glucose disposal in glucose and insulin tolerance tests. These results suggest that MID-00935 may be a potential muscle-targeting drug candidate for treating insulin resistance.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Proteínas Sustrato del Receptor de Insulina/antagonistas & inhibidores , Resistencia a la Insulina , Insulina/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Obesidad/metabolismo , Animales , Proteínas Portadoras/metabolismo , Células HEK293 , Humanos , Proteínas Sustrato del Receptor de Insulina/metabolismo , Proteínas de la Membrana , Fibras Musculares Esqueléticas/patología , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico , Obesidad/patología , Transducción de Señal/efectos de los fármacos
7.
Free Radic Biol Med ; 112: 504-514, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28842348

RESUMEN

Although the oxidative phosphorylation (OXPHOS) system has been found in mitochondria and the plasma membrane of various mammalian cell lines, understanding the physiological functions of the plasma membrane OXPHOS system is challenging. Here, we demonstrated that OXPHOS I, II, III, IV and V subunits were expressed in the plasma membrane of HepG2 cells and primary mouse hepatocytes, as determined by non-permeabilized immunofluorescence, total internal reflection fluorescence (TIRF) microscopy, cell surface-biotin labeling and plasma membrane and lipid raft isolation. Next, we demonstrated that NADH administration generated extracellular superoxide and improved insulin signaling in HepG2 cells and primary mouse hepatocytes. The NADH-dependent generation of extracellular superoxide was prevented by knockdown of NDUFV-1, the first subunit of OXPHOS I receiving electrons from NADH and the NADH-improved insulin signaling was abolished by extracellular catalase. Thus, we conclude that the OXPHOS system in the plasma membrane may be required for the generation of extracellular ROS and the regulation of insulin signaling.


Asunto(s)
Membrana Celular/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Insulina/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Superóxidos/metabolismo , Animales , Células COS , Catalasa/metabolismo , Catalasa/farmacología , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Chlorocebus aethiops , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Células HEK293 , Células Hep G2 , Hepatocitos/metabolismo , Humanos , Insulina/metabolismo , Microdominios de Membrana/efectos de los fármacos , Microdominios de Membrana/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , NAD/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal
8.
Science ; 354(6308)2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27846469

RESUMEN

Inhibition or genetic deletion of poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) is protective against toxic insults in many organ systems. The molecular mechanisms underlying PARP-1-dependent cell death involve release of mitochondrial apoptosis-inducing factor (AIF) and its translocation to the nucleus, which results in chromatinolysis. We identified macrophage migration inhibitory factor (MIF) as a PARP-1-dependent AIF-associated nuclease (PAAN). AIF was required for recruitment of MIF to the nucleus, where MIF cleaves genomic DNA into large fragments. Depletion of MIF, disruption of the AIF-MIF interaction, or mutation of glutamic acid at position 22 in the catalytic nuclease domain blocked MIF nuclease activity and inhibited chromatinolysis, cell death induced by glutamate excitotoxicity, and focal stroke. Inhibition of MIF's nuclease activity is a potential therapeutic target for diseases caused by excessive PARP-1 activation.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Apoptosis , División del ADN , Daño del ADN , ADN de Cadena Simple/metabolismo , Desoxirribonucleasas/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Factor Inductor de la Apoptosis/genética , Secuencia de Bases , Dominio Catalítico , Núcleo Celular/enzimología , Cromatina/metabolismo , Daño del ADN/genética , Fragmentación del ADN , Desoxirribonucleasas/química , Desoxirribonucleasas/genética , Ácido Glutámico/química , Ácido Glutámico/genética , Ácido Glutámico/toxicidad , Células HeLa , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/genética , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/genética , Ratones , Ratones Noqueados , Mitocondrias/enzimología , Mutación , Neuronas/enzimología , Conformación de Ácido Nucleico , Estrés Oxidativo , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/genética
9.
J Biol Chem ; 291(52): 26627-26635, 2016 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-27810898

RESUMEN

Mitsugumin 53 (MG53) is an E3 ligase that interacts with and ubiquitinates insulin receptor substrate-1 (IRS-1) in skeletal muscle; thus, an MG53-IRS-1 interaction disruptor (MID), which potentially sensitizes insulin signaling with an elevated level of IRS-1 in skeletal muscle, is an excellent candidate for treating insulin resistance. To screen for an MID, we developed a bimolecular luminescence complementation system using an N-terminal luciferase fragment fused with IRS-1 and a C-terminal luciferase fragment fused with an MG53 C14A mutant that binds to IRS-1 but does not have E3 ligase activity. An MID, which was discovered using the bimolecular luminescence complementation system, disrupted the molecular association of MG53 with IRS-1, thus abolishing MG53-mediated IRS-1 ubiquitination and degradation. Thus, the MID sensitized insulin signaling and increased insulin-elicited glucose uptake with an elevated level of IRS-1 in C2C12 myotubes. These data indicate that this MID holds promise as a drug candidate for treating insulin resistance.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Insulina/metabolismo , Proteínas de Microtúbulos/metabolismo , Músculo Esquelético/metabolismo , Proteínas Nucleares/metabolismo , Mapas de Interacción de Proteínas/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/metabolismo , Células Cultivadas , Humanos , Resistencia a la Insulina , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteolisis , Transducción de Señal/efectos de los fármacos , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
10.
Nat Neurosci ; 19(5): 678-689, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26928066

RESUMEN

Mutations in CHD7, encoding ATP-dependent chromodomain helicase DNA-binding protein 7, in CHARGE syndrome lead to multiple congenital anomalies, including craniofacial malformations, neurological dysfunction and growth delay. Mechanisms underlying the CNS phenotypes remain poorly understood. We found that Chd7 is a direct transcriptional target of oligodendrogenesis-promoting factors Olig2 and Smarca4/Brg1 and is required for proper onset of CNS myelination and remyelination. Genome-occupancy analyses in mice, coupled with transcriptome profiling, revealed that Chd7 interacted with Sox10 and targeted the enhancers of key myelinogenic genes. These analyses identified previously unknown Chd7 targets, including bone formation regulators Osterix (also known as Sp7) and Creb3l2, which are also critical for oligodendrocyte maturation. Thus, Chd7 coordinates with Sox10 to regulate the initiation of myelinogenesis and acts as a molecular nexus of regulatory networks that account for the development of a seemingly diverse array of lineages, including oligodendrocytes and osteoblasts, pointing to previously uncharacterized Chd7 functions in white matter pathogenesis in CHARGE syndrome.


Asunto(s)
Síndrome CHARGE/fisiopatología , Proteínas de Unión al ADN/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Vaina de Mielina/fisiología , Neurogénesis/fisiología , Factores de Transcripción SOXE/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Oligodendroglía/fisiología , Factores de Transcripción SOXE/metabolismo , Factor de Transcripción Sp7 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Ann Dermatol ; 28(1): 22-9, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26848215

RESUMEN

BACKGROUND: Many inflammatory mediators, including various cytokines (e.g. interleukins and tumor necrosis factor [TNF]), inflammatory proteases, and histamine are released following mast cell activation. However, the endogenous modulators for mast cell activation and the underlying mechanism have yet to be elucidated. Endogenous cannabinoids such as palmitoylethanolamide (PEA) and N-arachidonoylethanolamine (anandamide or AEA), were found in peripheral tissues and have been proposed to possess autacoid activity, implying that cannabinoids may downregulate mast cell activation and local inflammation. OBJECTIVE: In order to investigate the effect of cannabinoid receptor-1 (CB1R) agonists on mast cell activation, AEA-derived compounds were newly synthesized and evaluated for their effect on mast cell activation. METHODS: The effects of selected compounds on FcεRI-induced histamine and ß-hexosaminidase release were evaluated in a rat basophilic leukemia cell line (RBL-2H3). To further investigate the inhibitory effects of CB1R agonist in vivo, an oxazolone-induced atopic dermatitis mouse model was exploited. RESULTS: We found that CB1R inhibited the release of inflammatory mediators without causing cytotoxicity in RBL-2H3 cells and that CB1R agonists markedly and dose-dependently suppressed mast cell proliferation indicating that CB1R plays an important role in modulating antigen-dependent immunoglobulin E (IgE)-mediated mast cell activation. We also found that topical application of CB1R agonists suppressed the recruitment of mast cells into the skin and reduced the level of blood histamine. CONCLUSION: Our results indicate that CB1R agonists down-regulate mast cell activation and may be used for relieving inflammatory symptoms mediated by mast cell activation, such as atopic dermatitis, psoriasis, and contact dermatitis.

12.
BMB Rep ; 49(2): 116-21, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26645635

RESUMEN

Although proteomic analyses have revealed the presence of mitochondrial oxidative phosphorylation (OXPHOS) proteins in the plasma membrane, there have been no in-depth evaluations of the presence or function of OXPHOS I-V in the plasma membrane. Here, we demonstrate the in situ localization of OXPHOS I-V complexes to the sarcolemma of skeletal muscle by immunofluorescence and immunohistochemistry. A portion of the OXPHOS I-V complex proteins was not co-stained with MitoTracker but co-localized with caveolin-3 in the sarcolemma of mouse gastrocnemius. Mitochondrial matrix-facing OXPHOS complex subunits were ectopically expressed in the sarcolemma of the non-permeabilized muscle fibers and C2C12 myotubes. The sarcolemmal localization of cytochrome c was also observed from mouse gastrocnemius muscles and C2C12 myotubes, as determined by confocal and total internal resonance fluorescence (TIRF) microscopy. Based on these data, we conclude that a portion of OXPHOS complexes is localized in the sarcolemma of skeletal muscle and may have non-canonical functions. [BMB Reports 2016; 49(2): 116-121].


Asunto(s)
Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Fosforilación Oxidativa , Sarcolema/metabolismo , Animales , Caveolina 3/metabolismo , Membrana Celular/metabolismo , Respiración de la Célula , Citocromos c/metabolismo , Espacio Extracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , NAD/metabolismo , Consumo de Oxígeno
13.
J Cancer Prev ; 20(3): 185-92, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26473157

RESUMEN

BACKGROUND: Withania somnifera (known as Ashwagandha) is a medicinal plant used in the ayurvedic medicines in India. Withaferin-A, a withanolide derived from the leaf extract of W. somnifera, has been reported to exhibit anti-tumor activity against various cancer cells, such as leukemia, breast cancer and colon cancer cells. METHODS: We investigated the anti-cancer effects of withaferin-A on the proliferation and migration of human colorectal cancer (HCT116) cells. And we evaluated the effects of withaferin-A on the transcriptional activity of STAT3 and the growth of HCT116 cells in xenograft mouse tumor model. RESULTS: In the present study, we found that withaferin-A inhibited the proliferation and migration of HCT116 cells in a concentration-dependent manner. Treatment of HCT116 cells with withaferin-A attenuated interleukin-6-induced activation of STAT3, which has been implicated in the development and progression of colon cancer. To examine the effect of withaferin-A on HCT116 cells proliferation in vivo, we generated HCT116 cells xenograft tumors in Balb/c nude mice and treated the tumor bearing mice with or without withaferin-A intraperitoneally. Treatment with withaferin-A exhibited significant decrease in the volume and weight of tumors as compared to untreated controls. CONCLUSIONS: The present study suggests that withaferin-A holds the potential to be developed as a small molecule inhibitor of STAT3 for the treatment of HCT116.

14.
Int J Dermatol ; 54(10): e401-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26095080

RESUMEN

BACKGROUND: Even with the widespread clinical use of cannabinoid receptor (CBR) stimulating compounds, such as palmitoylethanolamine, the role of CBR agonists on inflammatory skin diseases is not yet fully understood. This study was performed to investigate the effects of CBR agonists on skin inflammation, using acute and chronic inflammation animal models. METHODS: The effectiveness of the newly synthesized cannabinoid receptor 1 (CB1R) agonists was determined using in vitro assays. Markers for epidermal permeability barrier function and skin inflammation were measured, and histological assessments were performed for evaluation. RESULTS: Topical application of CB1R-specific agonist significantly accelerated the recovery of epidermal permeability barrier function and showed anti-inflammatory activity in both acute and chronic inflammation models. Histological assessments also confirmed the anti-inflammatory effects, which is consistent with previous reports. CONCLUSIONS: All of the results suggest that topical application of CB1R-specific agonist can be beneficial for alleviating the inflammatory symptoms in chronic skin diseases, including atopic dermatitis.


Asunto(s)
Agonistas de Receptores de Cannabinoides/farmacología , Dermatitis Atópica/tratamiento farmacológico , Dermatitis por Contacto/tratamiento farmacológico , Ácidos Grasos Insaturados/farmacología , Propanolaminas/farmacología , Fenómenos Fisiológicos de la Piel/efectos de los fármacos , Piel/metabolismo , Enfermedad Aguda , Administración Cutánea , Animales , Enfermedad Crónica , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/patología , Dermatitis Atópica/fisiopatología , Dermatitis por Contacto/patología , Modelos Animales de Enfermedad , Femenino , Ratones Endogámicos BALB C , Oxazolona , Permeabilidad/efectos de los fármacos , Receptor Cannabinoide CB1/agonistas , Acetato de Tetradecanoilforbol/análogos & derivados , Pérdida Insensible de Agua
15.
J Dermatol Sci ; 79(3): 229-34, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26113114

RESUMEN

BACKGROUND: The ceramide metabolite, sphingosine-1-phosphate (S1P), regulates multiple cellular functions in keratinocytes (KC). We recently discovered that production of a key innate immune element, cathelicidin antimicrobial peptide (CAMP), is stimulated via a NF-κB-dependent mechanism that is activated by S1P when S1P is generated by sphingosine kinase (SPHK) 1. OBJECTIVE: We investigated whether pharmacological modulation of SPHK1 activity, using a novel synthetic SPHK1 activator, (S)-methyl 2-(hexanamide)-3-(4-hydroxyphenyl) propanoate (MHP), stimulates CAMP expression. METHODS: MHP-mediated changes in both S1P and CAMP downstream mediators were analyzed in normal cultured human KC by qRT-PCR, Western immunoblot, ELISA, confocal microscopy for immunohistochemistry, HPLC and ESI-LC/MS/MS, and microbial pathogen invasion/colonization in a human epidermal organotypic model. RESULTS: Treatment with MHP directly activated SPHK1 and increased cellular S1P content in normal cultured human KC. Because MHP did not inhibit S1P lyase activity, which hydrolyses S1P, augumented S1P levels could be attributed to increased synthesis rather than blockade of S1P degradation. Next, we found that exogenous MHP significantly stimulated CAMP mRNA and protein production in KC, increases that were significantly suppressed by siRNA directed against SPHK1, but not by a scrambled control siRNA. NF-κB activation, assessed by nuclear translocation of NF-κB, occurred in cells following incubation with MHP. Conversely, pretreatment with a specific inhibitor of SPHK1 decreased MHP-induced nuclear translocation of NF-κB, and significantly attenuated the MHP-mediated increase in CAMP production. Finally, topical MHP significantly suppressed invasion of the virulent Staphylococcus aureus into murine skin explants. CONCLUSION: MHP activation of SPHK1, a target enzyme of CAMP production, can stimulate innate immunity.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/biosíntesis , Epidermis/inmunología , Inmunidad Innata , Queratinocitos/enzimología , Lisofosfolípidos/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Esfingosina/análogos & derivados , Animales , Péptidos Catiónicos Antimicrobianos/genética , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Células Cultivadas , Activadores de Enzimas/farmacología , Inhibidores Enzimáticos/farmacología , Epidermis/enzimología , Humanos , Queratinocitos/química , Queratinocitos/inmunología , Ratones , FN-kappa B/metabolismo , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Esfingosina/metabolismo , Staphylococcus aureus/efectos de los fármacos , Staphylococcus aureus/fisiología , Tirosina/análogos & derivados , Tirosina/farmacología , Catelicidinas
16.
BMB Rep ; 48(9): 501-6, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25644636

RESUMEN

Based on the potential beneficial effects of growth hormone releasing peptide (GHRP)-6 on muscle functions, a newly synthesized GHRP-6-biotin conjugate was tested on cultured myoblast cells. Increased expression of myogenic marker proteins was observed in GHRP-6-biotin conjugate-treated cells. Additionally, increased expression levels of insulin-like growth factor-1 and collagen type I were observed. Furthermore, GHRP-6-biotin conjugate-treated cells showed increased metabolic activity, as indicated by increased concentrations of energy metabolites, such as ATP and lactate, and increased enzymatic activity of lactate dehydrogenase and creatine kinase. Finally, binding protein analysis suggested few candidate proteins, including desmin, actin, and zinc finger protein 691 as potential targets for GHRP6-biotin conjugate action. These results suggest that the newly synthesized GHRP-6-biotin conjugate has myogenic stimulating activity through, at least in part, by stimulating collagen type I synthesis and several key proteins. Practical applications of the GHRP-6-biotin conjugate could include improving muscle condition.


Asunto(s)
Biotina/análogos & derivados , Mioblastos/efectos de los fármacos , Oligopéptidos/química , Oligopéptidos/farmacología , Biotina/síntesis química , Biotina/farmacología , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Humanos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Desarrollo de Músculos/efectos de los fármacos , Mioblastos/citología , Mioblastos/metabolismo
17.
Cell Transplant ; 24(8): 1511-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25198120

RESUMEN

Efficient maintenance of the undifferentiated status of embryonic stem cells (ESCs) may be important for preparation of high-quality cell sources that can be successfully used for stem cell research and therapy. Here we tried to identify a compound that can enhance the quality of pluripotent stem cells. Treatment of ESCs and induced pluripotent stem cells (iPSCs) with 3,2'-dihydroxyflavone (3,2'-DHF) led to increases in cell growth, colony formation, and cell proliferation. Treatment with 3,2'-DHF resulted in high expression of pluripotency markers (OCT4, SOX2, and NANOG) and significant activation (STAT3 and AKT) or suppression (GSK3ß and ERK) of self-renewal-related kinases. 3,2'-DHF-treated high-quality pluripotent stem cells also showed enhanced differentiation potential. In particular, treatment of iPSCs with 3,2'-DHF led to elevated expression of ectodermal differentiation markers and improved differentiation into fully matured neurons. Next, we investigated the in vivo effect of 3,2'-DHF-pretreated iPSCs (3,2'-DHF iPSCs) in a peripheral nerve injury model and found that transplantation of 3,2'-DHF iPSCs resulted in more efficient axonal regeneration and functional recovery than in controls. Upon histopathological and gene expression analyses, we found that transplantation of 3,2'-DHF iPSCs stimulated expression of cytokines, such as TNF-α, in the early phase of injury and successfully reduced convalescence time of the injured peripheral nerve, showing an effective neuroprotective property. Taken together, our data suggest that 3,2'-DHF can be used for more efficient maintenance of pluripotent stem cells as well as for further applications in stem cell research and therapy.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Flavonoides/farmacología , Células Madre Pluripotentes/efectos de los fármacos , Animales , Axones/fisiología , Diferenciación Celular/genética , Línea Celular , Linaje de la Célula , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/trasplante , Masculino , Ratones , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Ratas , Ratas Sprague-Dawley , Regeneración , Neuropatía Ciática/terapia , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulación hacia Arriba/efectos de los fármacos
18.
BMB Rep ; 48(2): 68-80, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25413305

RESUMEN

G protein-coupled receptors (GPCRs) are a large class of transmembrane receptors categorized into five distinct families: rhodopsin, secretin, adhesion, glutamate, and frizzled. They bind and regulate 80% of all hormones and account for 20-50% of the pharmaceuticals currently on the market. Hundreds of GPCRs integrate and coordinate the functions of individual cells, mediating signaling between various organs. GPCRs are crucial players in tumor progression, adipogenesis, and inflammation. Several studies have also confirmed their central roles in embryonic development and stem cell maintenance. Recently, GPCRs have emerged as key players in the regulation of cell survival, proliferation, migration, and self-renewal in pluripotent (PSCs) and cancer stem cells (CSCs). Our study and other reports have revealed that the expression of many GPCRs is modulated during the generation of induced PSCs (iPSCs) or CSCs as well as during CSC sphere formation. These GPCRs may have crucial roles in the regulation of selfrenewal and other biological properties of iPSCs and CSCs. This review addresses the current understanding of the role of GPCRs in stem cell maintenance and somatic reprogramming to PSCs or CSCs.


Asunto(s)
Reprogramación Celular , Células Madre Neoplásicas/metabolismo , Células Madre Pluripotentes/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proliferación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Humanos , Células Madre Neoplásicas/citología , Células Madre Pluripotentes/citología , Transducción de Señal
19.
PLoS One ; 9(6): e99421, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24922551

RESUMEN

Placenta, as a reservoir of nutrients, has been widely used in medical and cosmetic materials. Here, we focused on the antioxidant properties of placental extract and attempted to isolate and identify the main antioxidant factors. Porcine placental extracts were prepared through homogenization or acid hydrolysis, and their antioxidant activity was investigated in the human keratinocyte HaCaT cell line. Treatment with homogenized placental extract (H-PE) increased the cell viability of H2O2-treated HaCaT cells more than two-fold. H-PE treatment suppressed H2O2-induced apoptotic and necrotic cell death and decreased intracellular ROS levels in H2O2-treated HaCaT cells. The antioxidant factors in H-PE were found to be thermo-unstable and were thus expected to include proteins. The candidate antioxidant proteins were fractionated with cation-exchange, anion-exchange, and size-exclusion chromatography, and the antioxidant properties of the chromatographic fractions were investigated. We obtained specific antioxidant fractions that suppressed ROS generation and ROS-induced DNA strand breaks. From silver staining and MALDI-TOF analyses, alpha-fetoprotein (AFP) precursor was identified as a main marker for the antioxidant effect of H-PE. Purified AFP or ectopically expressed AFP exhibited synergistic antioxidant activity in the presence of estradiol. Taken together, our data suggest that AFP, a serum glycoprotein produced at high levels during fetal development, is a novel marker protein for the antioxidant effect of the placenta that exhibits synergistic antioxidant activity in the presence of estradiol.


Asunto(s)
Antioxidantes/farmacología , Estradiol/farmacología , Extractos Placentarios/farmacología , alfa-Fetoproteínas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Femenino , Calor , Humanos , Peróxido de Hidrógeno/farmacología , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Necrosis , Estrés Oxidativo/efectos de los fármacos , Sus scrofa
20.
J Biol Chem ; 289(29): 20012-25, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24895128

RESUMEN

To address whether mitochondrial biogenesis is essential for skeletal myogenesis, C2C12 myogenesis was investigated after knockdown of NADH dehydrogenase (ubiquintone) flavoprotein 1 (NDUFV1), which is an oxidative phosphorylation complex I subunit that is the first subunit to accept electrons from NADH. The NDUFVI knockdown enhanced C2C12 myogenesis by decreasing the NAD(+)/NADH ratio and subsequently inactivating SIRT1 and SIRT1 activators (pyruvate, SRT1720, and resveratrol) abolished the NDUFV1 knockdown-induced myogenesis enhancement. However, the insulin-elicited activation of insulin receptor ß (IRß) and insulin receptor substrate-1 (IRS-1) was reduced with elevated levels of protein-tyrosine phosphatase 1B after NDUFV1 knockdown in C2C12 myotubes. The NDUFV1 knockdown-induced blockage of insulin signaling was released by protein-tyrosine phosphatase 1B knockdown in C2C12 myotubes, and we found that NDUFV1 or SIRT1 knockdown did not affect mitochondria biogenesis during C2C12 myogenesis. Based on these data, we can conclude that complex I dysfunction-induced SIRT1 inactivation leads to myogenesis enhancement but blocks insulin signaling without affecting mitochondria biogenesis.


Asunto(s)
Complejo I de Transporte de Electrón/deficiencia , Complejo I de Transporte de Electrón/metabolismo , Insulina/metabolismo , Enfermedades Mitocondriales/metabolismo , Desarrollo de Músculos/fisiología , Músculo Esquelético/crecimiento & desarrollo , Músculo Esquelético/metabolismo , Sirtuina 1/antagonistas & inhibidores , Animales , Línea Celular , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Complejo I de Transporte de Electrón/genética , Técnicas de Silenciamiento del Gen , Resistencia a la Insulina/fisiología , Ratones , Modelos Biológicos , Desarrollo de Músculos/genética , Fibras Musculares Esqueléticas/metabolismo , NAD/metabolismo , Fosforilación Oxidativa , Proteína Tirosina Fosfatasa no Receptora Tipo 1/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Sirtuina 1/genética , Sirtuina 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA