Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Stem Cells ; 32(12): 3099-111, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25187421

RESUMEN

Pluripotent stem cells have been shown to have unique nuclear properties, for example, hyperdynamic chromatin and large, condensed nucleoli. However, the contribution of the latter unique nucleolar character to pluripotency has not been well understood. Here, we show that fibrillarin (FBL), a critical methyltransferase for ribosomal RNA (rRNA) processing in nucleoli, is one of the proteins highly expressed in pluripotent embryonic stem (ES) cells. Stable expression of FBL in ES cells prolonged the pluripotent state of mouse ES cells cultured in the absence of leukemia inhibitory factor (LIF). Analyses using deletion mutants and a point mutant revealed that the methyltransferase activity of FBL regulates stem cell pluripotency. Knockdown of this gene led to significant delays in rRNA processing, growth inhibition, and apoptosis in mouse ES cells. Interestingly, both partial knockdown of FBL and treatment with actinomycin D, an inhibitor of rRNA synthesis, induced the expression of differentiation markers in the presence of LIF and promoted stem cell differentiation into neuronal lineages. Moreover, we identified p53 signaling as the regulatory pathway for pluripotency and differentiation of ES cells. These results suggest that proper activity of rRNA production in nucleoli is a novel factor for the regulation of pluripotency and differentiation ability of ES cells.


Asunto(s)
Apoptosis/fisiología , Diferenciación Celular/fisiología , Nucléolo Celular/metabolismo , Células Madre Embrionarias de Ratones/citología , Células Madre Pluripotentes/citología , ARN Ribosómico/biosíntesis , Animales , Diferenciación Celular/genética , Células Cultivadas , Factor Inhibidor de Leucemia/metabolismo , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo , Transducción de Señal/fisiología
2.
Proc Natl Acad Sci U S A ; 107(24): 10926-31, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20508149

RESUMEN

Transcription networks composed of various transcriptional factors specifically expressed in undifferentiated embryonic stem (ES) cells have been implicated in the regulation of pluripotency in ES cells. However, the molecular mechanisms responsible for self-renewal, maintenance of pluripotency, and lineage specification during differentiation of ES cells are still unclear. The results of this study demonstrate that a phosphorylation-dependent chromatin relaxation factor, transcriptional intermediary factor-1beta (TIF1beta), is a unique regulator of the pluripotency of ES cells and regulates Oct3/4-dependent transcription in a phosphorylation-dependent manner. TIF1beta is specifically phosphorylated in pluripotent mouse ES cells at the C-terminal serine 824, which has been previously shown to induce chromatin relaxation. Phosphorylated TIF1beta is partially colocalized at the activated chromatin markers, and forms a complex with the pluripotency-specific transcription factor Oct3/4 and subunits of the switching defective/sucrose nonfermenting, ATP-dependent chromatin remodeling complex, Smarcd1 [corrected], Brg-1, and BAF155, all of which are components of an ES-specific chromatin remodeling complex, esBAF. Phosphorylated TIF1beta specifically induces ES cell-specific genes and enables prolonged maintenance of an undifferentiated state in mouse ES cells. Moreover, TIF1beta regulates the reprogramming process of somatic cells in a phosphorylation-dependent manner. Our results suggest that TIF1beta provides a phosphorylation-dependent, bidirectional platform for specific transcriptional factors and chromatin remodeling enzymes that regulate the cell differentiation process and the pluripotency of stem cells.


Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Proteínas Represoras/metabolismo , Sustitución de Aminoácidos , Animales , Diferenciación Celular , Ensamble y Desensamble de Cromatina , Ratones , Mutagénesis Sitio-Dirigida , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Fosforilación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Serina/química , Factores de Transcripción/metabolismo , Proteína 28 que Contiene Motivos Tripartito
3.
Learn Mem ; 17(4): 176-85, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20332189

RESUMEN

A recent study has revealed that fear memory may be vulnerable following retrieval, and is then reconsolidated in a protein synthesis-dependent manner. However, little is known about the molecular mechanisms of these processes. Activin betaA, a member of the TGF-beta superfamily, is increased in activated neuronal circuits and regulates dendritic spine morphology. To clarify the role of activin in the synaptic plasticity of the adult brain, we examined the effect of inhibiting or enhancing activin function on hippocampal long-term potentiation (LTP). We found that follistatin, a specific inhibitor of activin, blocked the maintenance of late LTP (L-LTP) in the hippocampus. In contrast, administration of activin facilitated the maintenance of early LTP (E-LTP). We generated forebrain-specific activin- or follistatin-transgenic mice in which transgene expression is under the control of the Tet-OFF system. Maintenance of hippocampal L-LTP was blocked in the follistatin-transgenic mice. In the contextual fear-conditioning test, we found that follistatin blocked the formation of long-term memory (LTM) without affecting short-term memory (STM). Furthermore, consolidated memory was selectively weakened by the expression of follistatin during retrieval, but not during the maintenance phase. On the other hand, the maintenance of memory was also influenced by activin overexpression during the retrieval phase. Thus, the level of activin in the brain during the retrieval phase plays a key role in the maintenance of long-term memory.


Asunto(s)
Potenciación a Largo Plazo/fisiología , Memoria/fisiología , Animales , Conducta Animal , Biofisica , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/genética , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Giro Dentado/efectos de los fármacos , Giro Dentado/fisiología , Doxiciclina/administración & dosificación , Estimulación Eléctrica/métodos , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Miedo , Folistatina/genética , Folistatina/farmacología , Lateralidad Funcional , Técnicas In Vitro , Subunidades beta de Inhibinas/genética , Subunidades beta de Inhibinas/metabolismo , Potenciación a Largo Plazo/efectos de los fármacos , Potenciación a Largo Plazo/genética , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Prosencéfalo/metabolismo , Ratas , Ratas Wistar
4.
J Alzheimers Dis ; 81(2): 451-458, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33814453

RESUMEN

Accumulating evidence suggests that the adiponectin (APN) paradox might be involved in promoting aging-associated chronic diseases such as Alzheimer's disease (AD). In human brain, APN regulation of the evolvability of amyloidogenic proteins (APs), including amyloid-ß (Aß) and tau, in developmental/reproductive stages, might be paradoxically manifest as APN stimulation of AD through antagonistic pleiotropy in aging. The unique mechanisms underlying APN activity remain unclear, a better understanding of which might provide clues for AD therapy. In this paper, we discuss the possible relevance of activin, a member of transforming growth factor ß (TGFß) superfamily of peptides, to antagonistic pleiotropy effects of APN. Notably, activin, a multiple regulator of cell proliferation and differentiation, as well as an endocrine modulator in reproduction and an organizer in early development, might promote aging-associated disorders, such as inflammation and cancer. Indeed, serum activin, but not serum TGFß increases during aging. Also, activin/TGFß signal through type II and type I receptors, both of which are transmembrane serine/threonine kinases, and the serine/threonine phosphorylation of APs, including Aß42 serine 8 and αS serine 129, may confer pathological significance in neurodegenerative diseases. Moreover, activin expression is induced by APN in monocytes and hepatocytes, suggesting that activin might be situated downstream of the APN paradox. Finally, a meta-analysis of genome-wide association studies demonstrated that two SNPs relevant to the activin/TGFß receptor signaling pathways conferred risk for major aging-associated disease. Collectively, activin might be involved in the APN paradox of AD and could be a significant therapeutic target.


Asunto(s)
Activinas/metabolismo , Envejecimiento/fisiología , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Adiponectina/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Animales , Encéfalo/patología , Humanos
5.
Proteomics ; 9(1): 126-37, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19053146

RESUMEN

Embryonic stem cells (ESCs) are established from the inner cell mass of preimplantation embryos, are capable of self-renewal, and exhibit pluripotency. Given these unique properties, ESCs are expected to have therapeutic potential in regenerative medicine and as a powerful tool for in vitro differentiation studies of stem cells. Various growth factors and extracellular matrix components regulate the pluripotency and differentiation of ESC progenies. Thus, the cell surface receptors that bind these regulatory factors are crucial for the precise regulation of stem cells. To identify membrane proteins that are involved in the regulation of pluripotent stem cells, the membrane proteins of murine ESCs cultured with or without leukemia inhibitory factor (LIF) were purified and analyzed by quantitative proteomics. 2-D PAGE-based analysis using fluorescently labeled proteins and shotgun-based analysis with isotope-labeled peptides identified 338 proteins, including transmembrane, membrane-binding, and extracellular proteins, which were expressed specifically in pluripotent or differentiated murine ESCs. Functions of the identified proteins revealed cell adhesion molecules, channels, and receptors, which are expected to play important roles in the maintenance of murine ESC pluripotency. Membrane proteins that are expressed in pluripotent ESCs but not in differentiated cells such as Slc16a1 and Bsg could be useful for the selection of the stem cells in vitro.


Asunto(s)
Células Madre Embrionarias/química , Células Madre Embrionarias/citología , Proteínas de la Membrana/análisis , Células Madre Pluripotentes/química , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular , Electroforesis en Gel Bidimensional , Células Madre Embrionarias/efectos de los fármacos , Expresión Génica , Marcaje Isotópico , Factor Inhibidor de Leucemia/metabolismo , Ratones , Células Madre Pluripotentes/efectos de los fármacos
6.
FASEB J ; 22(2): 477-87, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17893249

RESUMEN

Myostatin is a potent negative regulator of skeletal muscle growth. Therefore, myostatin inhibition offers a novel therapeutic strategy for muscular dystrophy by restoring skeletal muscle mass and suppressing the progression of muscle degeneration. The known myostatin inhibitors include myostatin propeptide, follistatin, follistatin-related proteins, and myostatin antibodies. Although follistatin shows potent myostatin-inhibiting activities, it also acts as an efficient inhibitor of activins. Because activins are involved in multiple functions in various organs, their blockade by follistatin would affect multiple tissues other than skeletal muscles. In the present study, we report the characterization of a myostatin inhibitor derived from follistatin, which does not affect activin signaling. The dissociation constants (K(d)) of follistatin to activin and myostatin are 1.72 nM and 12.3 nM, respectively. By contrast, the dissociation constants (K(d)) of a follistatin-derived myostatin inhibitor, designated FS I-I, to activin and myostatin are 64.3 microM and 46.8 nM, respectively. Transgenic mice expressing FS I-I, under the control of a skeletal muscle-specific promoter showed increased skeletal muscle mass and strength. Hyperplasia and hypertrophy were both observed. We crossed FS I-I transgenic mice with mdx mice, a model for Duchenne muscular dystrophy. Notably, the skeletal muscles in the mdx/FS I-I mice showed enlargement and reduced cell infiltration. Muscle strength is also recovered in the mdx/FS I-I mice. These results indicate that myostatin blockade by FS I-I has a therapeutic potential for muscular dystrophy.


Asunto(s)
Folistatina/metabolismo , Regulación de la Expresión Génica , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/farmacología , Animales , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Cinética , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético , Distrofias Musculares/genética , Distrofias Musculares/fisiopatología , Mutación/genética , Miostatina , Unión Proteica
7.
Mol Cell Biol ; 26(4): 1318-32, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16449645

RESUMEN

Transforming growth factor beta (TGF-beta) receptors phosphorylate Smad3 and induce its nuclear import so it can regulate gene transcription. Smad3 can return to the cytoplasm to propagate further cycles of signal transduction or to be degraded. We demonstrate that Smad3 is exported by a constitutive mechanism that is insensitive to leptomycin B. The Mad homology 2 (MH2) domain is responsible for Smad3 export, which requires the GTPase Ran. Inactive, GDP-locked RanT24N or nuclear microinjection of Ran GTPase activating protein 1 blocked Smad3 export. Inactivation of the Ran guanine nucleotide exchange factor RCC1 inhibited Smad3 export and led to nuclear accumulation of phosphorylated Smad3. A screen for importin/exportin family members that associate with Smad3 identified exportin 4, which binds a conserved peptide sequence in the MH2 domain of Smad3 in a Ran-dependent manner. Exportin 4 is sufficient for carrying the in vitro nuclear export of Smad3 in cooperation with Ran. Knockdown of endogenous exportin 4 completely abrogates the export of endogenous Smad3. A short peptide representing the minimal interaction domain in Smad3 effectively competes with Smad3 association to exportin 4 and blocks nuclear export of Smad3 in vivo. We thus delineate a novel nuclear export pathway for Smad3.


Asunto(s)
Carioferinas/metabolismo , Proteínas Nucleares/metabolismo , Proteína smad3/metabolismo , Proteína de Unión al GTP ran/metabolismo , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Sitios de Unión/genética , Línea Celular , Células HeLa , Humanos , Técnicas In Vitro , Carioferinas/antagonistas & inhibidores , Carioferinas/genética , Modelos Biológicos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Homología de Secuencia de Aminoácido , Proteína smad3/química , Proteína smad3/genética , Transfección
8.
Clin Cancer Res ; 14(3): 660-7, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-18245525

RESUMEN

PURPOSE: Follistatin (FST), an inhibitor of activin, regulates a variety of biological functions, including cell proliferation, differentiation, and apoptosis. However, the role of FST in cancer metastasis is still unknown. Previous research established a multiple-organ metastasis model of human small cell lung cancer in natural killer cell-depleted SCID mice. In this model, i.v. inoculated tumor cells produced metastatic colonies in multiple organs including the lung, liver, and bone. The purpose of this study is to determine the role of FST in multiple-organ metastasis using this model. EXPERIMENTAL DESIGN: A human FST gene was transfected into the small cell lung cancer cell lines SBC-3 and SBC-5 and established transfectants secreting biologically active FST. The metastatic potential of the transfectants was evaluated using the metastasis model. RESULTS: FST-gene transfection did not affect the cell proliferation, motility, invasion, or adhesion to endothelial cells in vitro. I.v. inoculated SBC-3 or SBC-5 cells produced metastatic colonies into multiple organs, including the lung, liver, and bone in the natural killer cell-depleted SCID mice. FST transfectants produced significantly fewer metastatic colonies in these organs when compared with their parental cells or vector control clones. Immunohistochemical analyses of the liver metastases revealed that the number of proliferating tumor cells and the tumor-associated microvessel density were significantly less in the lesions produced by FST transfectants. CONCLUSIONS: These results suggest that FST plays a critical role in the production of multiple-organ metastasis, predominantly by inhibiting the angiogenesis. This is the first report to show the role of FST in metastases.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Folistatina/fisiología , Células Asesinas Naturales/inmunología , Depleción Linfocítica , Metástasis de la Neoplasia/prevención & control , Animales , Carcinoma de Pulmón de Células no Pequeñas/inmunología , División Celular , Folistatina/genética , Humanos , Masculino , Ratones , Ratones SCID , Ratones Transgénicos , Transfección , Cicatrización de Heridas
9.
Drug Discov Today ; 23(6): 1305-1311, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29747002

RESUMEN

Given the paradigm of anti-insulin resistance in therapies for metabolic syndrome, there has been considerable interest in adiponectin (APN), an adipocyte-derived sensitizer of insulin receptor signaling. In contrast to hypoadiponectinemia in metabolic syndrome, evidence suggests that Alzheimer's disease (AD) and other diseases, including chronic heart failure (CHF) and chronic kidney disease (CKD), are characterized by hyperadiponectinemia as well as the APN/obesity paradoxes, indicating that a decrease in APN might also be beneficial for these diseases. Thus, distinct from metabolic syndrome, it is anticipated that APN receptor antagonists rather than agonists might be effective in therapy for some chronic diseases.


Asunto(s)
Envejecimiento/metabolismo , Receptores de Adiponectina/metabolismo , Adiponectina/deficiencia , Adiponectina/metabolismo , Animales , Enfermedad Crónica , Humanos , Síndrome Metabólico/tratamiento farmacológico , Síndrome Metabólico/metabolismo , Errores Innatos del Metabolismo/tratamiento farmacológico , Errores Innatos del Metabolismo/metabolismo , Obesidad/metabolismo , Receptores de Adiponectina/agonistas , Receptores de Adiponectina/antagonistas & inhibidores , Transducción de Señal
10.
Cell Rep ; 25(5): 1193-1203, 2018 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-30380411

RESUMEN

Brown adipocyte activation or beige adipocyte emergence in white adipose tissue (WAT) increases energy expenditure, leading to a reduction in body fat mass and improved glucose metabolism. We found that activin E functions as a hepatokine that enhances thermogenesis in response to cold exposure through beige adipocyte emergence in inguinal WAT (ingWAT). Hepatic activin E overexpression activated thermogenesis through Ucp1 upregulation in ingWAT and other adipose tissues including interscapular brown adipose tissue and mesenteric WAT. Hepatic activin E-transgenic mice exhibited improved insulin sensitivity. Inhibin ßE gene silencing inhibited cold-induced Ucp1 induction in ingWAT. Furthermore, in vitro experiments suggested that activin E directly stimulated expression of Ucp1 and Fgf21, which was mediated by transforming growth factor-ß or activin type I receptors. We uncovered a function of activin E to stimulate energy expenditure through brown and beige adipocyte activation, suggesting a possible preventive or therapeutic target for obesity.


Asunto(s)
Activinas/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo Energético , Homeostasis , Subunidades beta de Inhibinas/metabolismo , Receptores de Activinas Tipo I/metabolismo , Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Animales , Peso Corporal , Diferenciación Celular , Frío , Factores de Crecimiento de Fibroblastos/metabolismo , Glucosa/metabolismo , Células HEK293 , Humanos , Resistencia a la Insulina , Metabolismo de los Lípidos , Hígado/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Termogénesis , Factor de Crecimiento Transformador beta/metabolismo
11.
Biochim Biophys Acta ; 1763(8): 900-6, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16872693

RESUMEN

Previous studies have demonstrated that treatment with activin A and TGF-beta(1), members of the TGF-beta family, stimulated maturation of mouse bone marrow-derived cultured mast cells (BMMC), which was characterized by morphology and gene expression of mouse mast cell proteases (mmcps). In order to gain a better understanding of activin A- and TGF-beta(1)-induced maturation in mast cells, we investigated the genes that were up-regulated in response to treatment with these two members of the TGF-beta family. The cDNA microarray analyses indicated that in BMMC, five genes were induced by treatment with 4 nM activin A for 2 h. Tocopherol-associated protein (Tap) was one of the induced genes, and the Tap induction in response to activin A treatment was confirmed by real-time RT-PCR analyses. Treatment with TGF-beta(1) at 200 pM but not BMP-2 at 4 nM also increased Tap gene transcript in BMMC. Activin A-induced Tap expression was detected in BMMC but not in RAW264 macrophage-like cells, B16 melanoma cells or P19 embryonic carcinoma cells. Treatment with >1 muM SB431542, an inhibitor of activin and TGF-beta type I receptors ALK4/5, reduced responsiveness of Tap expression to TGF-beta(1), whereas <0.5 microM SB431542 effectively reduced TGF-beta(1)-induced expression of mmcp-1 and mmcp-7. These results suggest that inhibitory effects of SB431542 are different between TGF-beta-induced genes. Reporter assays indicated that Tap expression enhances transcription mediated by the activin/TGF-beta pathway. Thus, the present results suggest that Tap induction in response to activin/TGF-beta occurs predominantly in mast cells and serves as a positive regulator in activin/TGF-beta signaling.


Asunto(s)
Activinas/farmacología , Proteínas Portadoras/genética , Subunidades beta de Inhibinas/farmacología , Lipoproteínas/genética , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Transactivadores/genética , Factor de Crecimiento Transformador beta/farmacología , Receptores de Activinas Tipo I/antagonistas & inhibidores , Activinas/antagonistas & inhibidores , Animales , Benzamidas/farmacología , Línea Celular , Células Cultivadas , ADN Complementario/genética , Dioxoles/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Subunidades beta de Inhibinas/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Transducción de Señal , Proteína smad3/deficiencia , Proteína smad3/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1
12.
Biochim Biophys Acta ; 1759(3-4): 166-70, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16730810

RESUMEN

Mouse mast cell protease-7 (mmcp-7) is a tryptase predominantly expressed in differentiated connective tissue-type mast cells. Previous study revealed that transforming growth factor-beta (TGF-beta) increases gene transcript of mmcp-7 in mast cells. The present study explored molecular mechanism of the up-regulation of mmcp-7 by TGF-beta. Luciferase-based reporter assays using deletion and point mutations of mmcp-7 promoter showed a critical region spanning nt -126 to -122 relative to the transcriptional start site, a Smad-binding element, for transcriptional activation by the TGF-beta pathway. In addition, a region from nt -104 to -98, a TPA-responsive element, was also necessary for the transactivation. Consistent with the current model for the TGF-beta signaling, Smad4 was required for the transcription of mmcp-7 by Smad3, a signal mediator of TGF-beta. Treatment with TGF-beta in mast cells resulted in the differential gene induction of the AP-1 components, i.e., transient induction of c-fos but not of c-jun and junB. Expression of c-fos further enhanced Smad3 and Smad4-induced transcription of mmcp-7, whereas c-jun expression inhibited the transcription. Our results suggest that TGF-beta stimulates mmcp-7 transcription through the Smad3-Smad4 pathway as well as c-fos induction, and that the AP-1 components distinctly related with the TGF-beta pathway.


Asunto(s)
Serina Endopeptidasas/genética , Transcripción Genética/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Quimasas , Mastocitos/efectos de los fármacos , Mastocitos/metabolismo , Ratones , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-fos/genética , Serina Endopeptidasas/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo , Proteína Smad4/genética , Proteína Smad4/metabolismo , Transcripción Genética/genética
13.
Cancer Chemother Pharmacol ; 59(1): 113-26, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16699793

RESUMEN

PURPOSE: We evaluated the antitumor and antiangiogenic activities of human natural interferon-alpha (IFN-alpha) alone or in combination with S-1 against human pancreatic cancer cells. METHODS: Three days after the subcutaneous (s.c.) implantation of tumor cells, mice (n = 12) were received s.c. injection with IFN-alpha alone (10,000 U six times a week), oral administration with S-1 alone (8 mg/kg six times a week), or both with IFN-alpha and S-1 (8, 10, 12 mg/kg six times a week). RESULTS: Administration of IFN-alpha in combination with S-1 significantly decreased progressive growth and angiogenesis of human pancreatic cancer cells. The combination therapy produced more significant inhibition in expression of the representative proangiogenic molecules, vascular endothelial growth factor and basic fibroblast growth factor than individual treatment either IFN-alpha or S-1 alone did. These treatments also decreased the staining of proliferating cell nuclear antigen, induced apoptosis and decreased microvessel density. In order to better understand the precise molecular mechanisms by which IFN-alpha and S-1 exert its effects, we have utilized cDNA microarray including 124 known genes to determine the gene expression profile altered by IFN-alpha and S-1 treatment. We found a total of seven genes which showed a twofold change after IFN-alpha and S-1 treatment in addition to VEGF, bFGF, CD31, MMP-2, MMP-7 and MMP-9. Among these genes, we found down-regulation of six genes and up-regulation of one gene, which are related to angiogenesis, tumor cell invasion and metastasis. CONCLUSIONS: These data suggest that administration of IFN-alpha in combination with S-1 may provide a novel and effective approach to the treatment of human pancreatic cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de la Angiogénesis/administración & dosificación , Animales , Antibióticos Antineoplásicos/administración & dosificación , ADN Complementario/biosíntesis , ADN Complementario/genética , Dihidrouracilo Deshidrogenasa (NADP)/metabolismo , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Humanos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Interferón-alfa/administración & dosificación , Metaloproteinasas de la Matriz/biosíntesis , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas/patología , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/biosíntesis , Antígeno Nuclear de Célula en Proliferación/biosíntesis , Análisis de Supervivencia , Tegafur/administración & dosificación , Factor A de Crecimiento Endotelial Vascular/biosíntesis
14.
Cell Signal ; 18(12): 2154-61, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16750902

RESUMEN

Transforming growth factor-beta (TGF-beta) modulates functions of bone marrow-derived cultured mast cells (BMMCs); cell maturation (up-regulation of mouse mast cell proteases (mmcps)), growth arrest and migration. We investigated the roles of p38 MAP kinase and Smad3 in TGF-beta-mediated cell responses in BMMCs. Treating BMMCs with TGF-beta induced the phosphorylation of p38 within 2 h and persisted for 24 h. The involvement of p38 in TGF-beta-induced cell responses depended upon mast cell functions; it was necessary for up-regulation of mmcp-1 and migration, but not for up-regulation of mmcp-7 and inhibition of metabolic activity. New protein synthesis was required for the up-regulation of mmcp-1 but not mmcp-7 in response to TGF-beta treatment, and stabilization of mRNA was partially responsible for the increase in gene transcript of mmcp-1. The decrease in metabolic activity in response to TGF-beta treatment was smaller in Smad3-deficient BMMCs compared to wild-type BMMCs. Maximal migration was detected at a TGF-beta concentration of 40 fM in wild-type BMMCs, whereas TGF-beta-induced migration was absent in Smad3-deficient BMMCs. Thus, the roles of p38 and Smad3 are different among TGF-beta-mediated cell responses in BMMCs.


Asunto(s)
Mastocitos/efectos de los fármacos , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Antracenos/farmacología , Western Blotting , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Quimasas/genética , Quimasas/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Imidazoles/farmacología , Mastocitos/metabolismo , Mastocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Fosforilación/efectos de los fármacos , Piridinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína smad3/deficiencia , Proteína smad3/genética , Triptasas/genética , Triptasas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
15.
J Med Invest ; 54(3-4): 267-75, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17878676

RESUMEN

Skin grafting has become a basic and established operation technique; however, it is not clear how skin grafts adapt to recipient beds and replace their functions. In this study, we analyzed the origin of cells in adapted transplants by using green fluorescent protein (GFP) transgenic mice, which emits green fluorescence in the whole body. The dorsal skins of GFP transgenic mice were transplanted to the back of wild-type mice. Similarly, wild-type skins were transplanted to the back of GFP transgenic mice. Since transplantation with full thickness back skin was not successful due to severe immunorejection, tail skins, which contain fewer epidermal Langerhans cells, were used for the experiments. Six months after transplantation, immunohistochemical analysis of the grafts revealed that tissues derived from ectodermal origin such as the epidermis, hair follicles, and sebaceous glands survived in transplanted grafts, but that other tissues such as the dermis, nerves and blood vessels are partly replaced by tissues from recipient beds. Our results further demonstrated that transplantation analyses with GFP transgenic mice could be a useful approach to study the origin of cells in transplants.


Asunto(s)
Supervivencia de Injerto , Trasplante de Piel , Animales , Supervivencia de Injerto/fisiología , Proteínas Fluorescentes Verdes/genética , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Recombinantes/genética , Trasplante de Piel/patología , Trasplante de Piel/fisiología
16.
J Med Invest ; 54(3-4): 276-88, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17878677

RESUMEN

Follistatin-related gene (FLRG) encodes a secretory glycoprotein that has characteristic cysteine-rich follistatin domains. FLRG protein binds to and neutralizes several transforming growth factor-beta (TGF-beta) superfamily members, including myostatin (MSTN), which is a potent negative regulator of skeletal muscle mass. We have previously reported that FLRG was abundantly expressed in fetal and adult mouse heart. In this study, we analyzed the expression of FLRG mRNA during mouse heart development. FLRG mRNA was continuously expressed in the embryonic heart, whereas it was very low in skeletal muscles. By contrast, MSTN mRNA was highly expressed in embryonic skeletal muscles, whereas the expression of MSTN mRNA was rather low in the heart. In situ hybridization and immunohistochemical analysis revealed that FLRG expressed in smooth muscle of the aorta and pulmonary artery, valve leaflets of mitral and tricuspid valves, and cardiac muscles in the ventricle of mouse embryonic heart. However, MSTN was expressed in very limited areas, such as valve leaflets of pulmonary and aortic valves, the top of the ventricular and atrial septa. Interestingly, the expression of MSTN was complementary to that of FLRG, especially in the valvular apparatus. Biochemical analyses with surface plasmon resonance biosensor and reporter assays demonstrated that FLRG hardly dissociates from MSTN and activin once it bound to them, and efficiently inhibits these activities. Our results suggest that FLRG could function as a negative regulator of activin family members including MSTN during heart development.


Asunto(s)
Activinas/metabolismo , Corazón Fetal/metabolismo , Proteínas/genética , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN/genética , Femenino , Corazón Fetal/embriología , Proteínas Relacionadas con la Folistatina , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Endogámicos ICR , Miostatina , Embarazo , Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
17.
Hepatogastroenterology ; 54(76): 1216-21, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17629073

RESUMEN

BACKGROUND/AIMS: We performed a long-term assessment of liver regeneration with or without PVL after massive hepatectomy in similar sized remnant livers to evaluate effects of regenerating livers preactivated by PVL following massive hepatectomy. METHODOLOGY: Rats were randomly divided into two groups, PVL-88%Hx and sham 88%Hx. As the initial operation, PVL or sham operation was performed by ligation of the portal vein of the left and median lobes or a similar manipulation without ligation, respectively. Four days after PVL, the volume of the posterior caudate lobe (5%) increased approximately two-fold (12%) and was the same size as the whole caudate lobe (12%) in the sham animals. Subsequently, 88%Hx was performed in the two groups. RESULTS: Survival rates were not significantly different between the two groups. Relative liver weight in PVL-88%Hx group was significantly higher up to 24hr, but after 48hr no significant difference was evident between the two groups. PCNA LI in sham-89%Hx group was significantly higher than that in PVL-88%Hx group after 48hr. The mRNA expression levels of activin A and ActRIIA were significantly higher in PVL-88%Hx group than in sham-88% group at 72 hr. CONCLUSIONS: The regenerating liver preactivated by PVL is restricted late-phase liver regeneration after massive hepatectomy.


Asunto(s)
Regeneración Hepática , Hígado/cirugía , Vena Porta/cirugía , Receptores de Activinas Tipo II/genética , Receptores de Activinas Tipo II/metabolismo , Activinas/genética , Activinas/metabolismo , Animales , Bilirrubina/sangre , Proliferación Celular , Hepatectomía , Hepatocitos/metabolismo , Ligadura , Hígado/irrigación sanguínea , Hígado/citología , Masculino , Tamaño de los Órganos , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas , Albúmina Sérica/análisis
18.
J Med Invest ; 53(3-4): 238-45, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16953060

RESUMEN

Transforming growth factor-beta (TGF-beta) family members regulate a variety of cellular functions and play important roles in cell differentiation. Activin receptor-like kinase 7 (ALK7), a receptor for TGF-beta family members, was initially cloned from rats as an orphan receptor and has been recently shown to be a type I receptor for nodal, activin B and activin AB. ALK7 is expressed not only in neurons, but also in insulin-producing islet beta cells and white and brown adipose tissues; however, the specific functions of ALK7 in these tissues are not known. In order to test whether ALK7 is involved in adipocyte differentiation, we analyzed its expression during adipocyte differentiation. ALK7 expression was detected in the late phase of adipocyte differentiation by reverse transcriptase-polymerase chain reaction (RT-PCR), Western blotting and immunofluorescence staining in 3T3-L1 cells. We also detected the expression of ALK7 by RT-PCR in stromal vascular fraction (SVF) cells. These results indicated that ALK7 is a novel marker specifically expressed during the late phase of adipocyte differentiation. Furthermore, our results suggest the possible involvement of nodal or activin B in adipocyte differentiation.


Asunto(s)
Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Diferenciación Celular/genética , Activinas/fisiología , Adipogénesis/genética , Animales , Línea Celular , Regulación de la Expresión Génica/genética , Marcadores Genéticos , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Neurosci Res ; 53(3): 323-30, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16157407

RESUMEN

To understand the neuronal cell type-specific expression of Ca2+/calmodulin-dependent protein kinase II (CaM kinase II), we investigated binding proteins that specifically activated the promoter of the alpha isoform of CaM kinase II (alpha CaM kinase II). Proteins that bind the promoter sequence were found in rat brain nuclear extract by electrophoretic mobility shift assay. Then, we screened for binding proteins in a mouse brain cDNA library using the yeast one-hybrid system. Zic2, a Zic family zinc finger transcription factor, was identified as one of the binding proteins. To investigate the effect of Zic2 on the promoter activity, Zic2 cDNA was expressed with a luciferase reporter gene containing a neuronal cell type-specific promoter of alpha CaM kinase II in neuronal and non-neuronal cells. The promoter activity of alpha CaM kinase II was enhanced 1.3-5-fold in cultured neuronal cells by Zic2. The activation was varied among neuronal cell types. Zic2 also increased the promoter activity in non-neuronal cells, although the relative luciferase activites in non-neuronal cells were lower than those in neuronal cell lines. These results indicated that Zic2 was one of the proteins binding to, and regulating the activity of, the promoter of alpha CaM kinase II.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Neuronas/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción/genética , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Línea Celular , Línea Celular Tumoral , ADN Complementario/genética , Proteínas de Unión al ADN/genética , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Luciferasas/genética , Ratones , Neuronas/citología , Elementos Reguladores de la Transcripción/genética , Activación Transcripcional/genética , Dedos de Zinc/fisiología
20.
Hepatogastroenterology ; 52(61): 60-6, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15782995

RESUMEN

BACKGROUND/AIMS: Activin A is an autocrine inhibitor of cell growth in the liver. The biological activity of activin A is mediated by a heteromeric receptor complex. Follistatin (FS) binds to activin and inhibits its biological effects, and acts as a negative regulator of muscle cells. The role of activin receptors during liver regeneration following a hepatectomy has not been fully assessed. This study investigates the mechanism underlying how activin receptors regulate hepatocyte growth, and the effects of intravenous administration of FS during liver regeneration. METHODOLOGY: The expression of both activins and activin receptors in the liver after a 70% partial hepatectomy (HT) was assessed by RT-PCR and immunohistochemistry. FS 315 or 288 was infused for different periods of time based on changes in hepatocyte activin receptor expression after HT. RESULTS: Activin receptor expression peaked between 48 and 72 hours after HT. 72 hours after HT, an injection of FS 315 resulted in a more potent stimulation of DNA synthesis and produced a greater increase in body weight compared with the control rats. CONCLUSIONS: The expression of activin receptors after peak DNA synthesis might be a key component in the downregulation of DNA synthesis. Intravenous administration of FS 315 might promote liver regeneration and have anabolic actions.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Folistatina/metabolismo , Subunidades beta de Inhibinas/metabolismo , Regeneración Hepática/fisiología , Proteínas/metabolismo , Receptores de Activinas Tipo II/efectos de los fármacos , Animales , Folistatina/administración & dosificación , Folistatina/farmacología , Hepatectomía , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Subunidades beta de Inhibinas/efectos de los fármacos , Inyecciones Intravenosas , Regeneración Hepática/efectos de los fármacos , Masculino , Proteínas/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA