Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMB Rep ; 55(7): 348-353, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35725010

RESUMEN

Gastrointestinal cancer is associated with a high mortality rate. Here, we report that the splice variant of NRP/B contributes to tumorigenic activity in highly malignant gastric cancer through dissociation from the tumor repressor, HDAC5. NRP/B mRNA expression is significantly higher in the human gastric cancer tissues than in the normal tissues. Further, high levels of both the NRP/B splice variant and Lgr5, but not the full-length protein, are found in highly tumorigenic gastric tumor cells, but not in non-tumorigenic cells. The loss of NRP/B markedly inhibits cell migration and invasion, which reduces tumor formation in vivo. Importantly, the inhibition of alternative splicing increases the levels of NRP/B-1 mRNA and protein in AGS cells. The ectopic expression of full-length NRP/B exhibits tumor-suppressive activity, whereas NRP/B-2 induces the noninvasive human gastric cancer cells tumorigenesis. The splice variant NRP/B-2 which loses the capacity to interact with tumor repressors promoted oncogenic activity, suggesting that the BTB/POZ domain in the N-terminus has a crucial role in the suppression of gastric cancer. Therefore, the regulation of alternative splicing of the NRP/B gene is a potential novel target for the treatment of gastrointestinal cancer. [BMB Reports 2022; 55(7): 348-353].


Asunto(s)
Neoplasias Gastrointestinales , Neoplasias Gástricas , Empalme Alternativo/genética , Carcinogénesis/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , ARN Mensajero , Neoplasias Gástricas/genética
2.
Cell Prolif ; 53(10): e12893, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32865873

RESUMEN

OBJECTIVES: Huntington's disease (HD) is a devastating neurodegenerative disease caused by polyglutamine (polyQ) expansion in the huntingtin (HTT) gene. Mutant huntingtin (mHTT) is the main cause of HD and is associated with impaired mitochondrial dynamics, ubiquitin-proteasome system and autophagy, as well as tauopathy. In this study, we aimed to establish a new neural stem cell line for HD studies. MATERIALS AND METHODS: YAC128 mice are a yeast artificial chromosome (YAC)-based transgenic mouse model of HD. These mice express a full-length human mutant HTT gene with 128 CAG repeats and exhibit various pathophysiological features of HD. In this study, we isolated a new neural stem cell line from the forebrains of YAC128 mouse embryos (E12.5) and analysed its characteristics using cellular and biochemical methods. RESULTS: Compared to wild-type (WT) NSCs, the YAC128 NSC line exhibited greater proliferation and migration capacity. In addition to mHTT expression, increased intracellular Ca2+ levels and dysfunctional mitochondrial membrane potential were observed in the YAC128 NSCs. YAC128 NSCs had defects in mitochondrial dynamics, including a deficit in mitochondrial axonal transport and unbalanced fusion and fission processes. YAC128 NSCs also displayed decreased voltage response variability and Na+ current amplitude. Additionally, the ubiquitin-proteasome and autophagy systems were impaired in the YAC128 NSCs. CONCLUSIONS: We have established a new neural stem line from YAC128 transgenic mice, which may serve as a useful resource for studying HD pathogenesis and drug screening.


Asunto(s)
Enfermedad de Huntington/patología , Células-Madre Neurales/metabolismo , Prosencéfalo/citología , Animales , Autofagia , Calcio/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Células-Madre Neurales/citología , Técnicas de Placa-Clamp , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo
3.
Molecules ; 25(8)2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32340245

RESUMEN

A concise and scalable synthetic route for optically pure (4S) and (4R)-5-(3',4'-dihydroxyphenyl)-γ-valerolactones (DHPVs), catechin metabolites, has been developed via the efficient construction of a γ-valerolactone moiety from hexenol. Noticeably, the different skin wrinkle-reducing activities of each metabolite were revealed via our unique syntheses of DHPVs in an enantiomerically pure form.


Asunto(s)
Catequina/síntesis química , Catequina/farmacología , Lactonas/síntesis química , Lactonas/farmacología , Piel/efectos de los fármacos , Humanos , Estructura Molecular , Oxidación-Reducción , Envejecimiento de la Piel/efectos de los fármacos
4.
Acta Neuropathol ; 132(4): 577-92, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27221146

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder of the central nervous system (CNS) that is defined by a CAG expansion in exon 1 of the huntingtin gene leading to the production of mutant huntingtin (mHtt). To date, the disease pathophysiology has been thought to be primarily driven by cell-autonomous mechanisms, but, here, we demonstrate that fibroblasts derived from HD patients carrying either 72, 143 and 180 CAG repeats as well as induced pluripotent stem cells (iPSCs) also characterized by 143 CAG repeats can transmit protein aggregates to genetically unrelated and healthy host tissue following implantation into the cerebral ventricles of neonatal mice in a non-cell-autonomous fashion. Transmitted mHtt aggregates gave rise to both motor and cognitive impairments, loss of striatal medium spiny neurons, increased inflammation and gliosis in associated brain regions, thereby recapitulating the behavioural and pathological phenotypes which characterizes HD. In addition, both in vitro work using co-cultures of mouse neural stem cells with 143 CAG fibroblasts and the SH-SY5Y human neuroblastoma cell line as well as in vivo experiments conducted in newborn wild-type mice suggest that exosomes can cargo mHtt between cells triggering the manifestation of HD-related behaviour and pathology. This is the first evidence of human-to-mouse prion-like propagation of mHtt in the mammalian brain; a finding which will help unravel the molecular bases of HD pathology as well as to lead to the development of a whole new range of therapies for neurodegenerative diseases of the CNS.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Células Madre Pluripotentes Inducidas/citología , Proteínas Mutantes/metabolismo , Neuronas/citología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Niño , Modelos Animales de Enfermedad , Humanos , Enfermedad de Huntington/terapia , Células Madre Pluripotentes Inducidas/patología , Masculino , Ratones , Neuronas/patología
5.
J Invest Dermatol ; 136(5): 1012-1021, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26854493

RESUMEN

Cacao beans contain various bioactive phytochemicals that could modify the pathogeneses of certain diseases. Here, we report that oral administration of cacao powder (CP) attenuates UVB-induced skin wrinkling by the regulation of genes involved in dermal matrix production and maintenance. Transcriptome analysis revealed that 788 genes are down- or upregulated in the CP supplemented group, compared with the UVB-irradiated mouse skin controls. Among the differentially expressed genes, cathepsin G and serpin B6c play important roles in UVB-induced skin wrinkle formation. Gene regulatory network analysis also identified several candidate regulators responsible for the protective effects of CP supplementation against UVB-induced skin damage. CP also elicited antiwrinkle effects via inhibition of UVB-induced matrix metalloproteinases-1 expression in both the human skin equivalent model and human dermal fibroblasts. Inhibition of UVB-induced activator protein-1 via CP supplementation is likely to affect the expression of matrix metalloproteinases-1. CP supplementation also downregulates the expression of cathepsin G in human dermal fibroblasts. 5-(3',4'-Dihydroxyphenyl)-γ-valerolactone, a major in vivo metabolite of CP, showed effects similar to CP supplementation. These results suggest that cacao extract may offer a protective effect against photoaging by inhibiting the breakdown of dermal matrix, which leads to an overall reduction in wrinkle formation.


Asunto(s)
Cacao , Colágeno/efectos de los fármacos , Suplementos Dietéticos , Envejecimiento de la Piel/genética , Rayos Ultravioleta/efectos adversos , Administración Oral , Análisis de Varianza , Animales , Colágeno/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Metaloproteinasa 1 de la Matriz/genética , Ratones , Ratones Pelados , Extractos Vegetales/farmacología , Distribución Aleatoria , Sensibilidad y Especificidad , Factor de Transcripción AP-1/genética , Regulación hacia Arriba
6.
J Cancer Prev ; 19(3): 187-98, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25337588

RESUMEN

BACKGROUND: NRP/B, a family member of the BTB/Kelch repeat proteins, is implicated in neuronal and cancer development, as well as the regulation of oxidative stress responses in breast and brain cancer. Our previous studies indicate that the NRP/B-BTB/POZ domain is involved in the dimerization of NRP/B and in a complex formation with the tumor suppressor, retinoblastoma protein. Although much evidence supports the potential role of NRP/B as a tumor suppressor, the molecular mechanisms of NRP/B action on E2F transcription factors have not been elucidated. METHODS: Three-dimensional modeling of NRP/B was used to generate point mutations in the BTB/Kelch domains. Tet-on inducible NRP/B expression was established. The NRP/B deficient breast cancer cell line, MDA-MB-231, was generated using lentiviral shNRP/B to evaluate the effect of NRP/B on cell proliferation, invasion and migration. Immunoprecipitation was performed to verify the interaction of NRP/B with E2F and histone deacetylase (HDAC-1), and the expression level of NRP/B protein was analyzed by Western blot analysis. Changes in cell cycle were determined by flow cytometry. Transcriptional activities of E2F transcription factors were measured by chloramphenicol acetyltransferase (CAT) activity. RESULTS: Ectopic overexpression of NRP/B demonstrated that the NRP/B-BTB/POZ domain plays a critical role in E2F-mediated transcriptional activity. Point mutations within the BTB/POZ domain restored E2-promoter activity inhibited by NRP/B. Loss of NRP/B enhanced the proliferation and migration of breast cancer cells. Endogenous NRP/B interacted with E2F and HDAC1. Treatement with an HDAC inhibitor, trichostatin A (TSA), abolished the NRP/B-mediated suppression of E2-promoter activity. Gain or loss of NRP/B in HeLa cells confirmed the transcriptional repressive capability of NRP/B on the E2F target genes, Cyclin E and HsORC (Homo sapiens Origin Recognition Complex). CONCLUSIONS: The present study shows that NRP/B acts as a transcriptional repressor by interacting with the co-repressors, HDAC1, providing new insight into the molecular mechanisms of NRP/B on tumor suppression.

7.
Cancer Res ; 74(21): 6139-49, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25205100

RESUMEN

Smad3, a major intracellular mediator of TGFß signaling, functions as both a positive and negative regulator in carcinogenesis. In response to TGFß, the TGFß receptor phosphorylates serine residues at the Smad3 C-tail. Cancer cells often contain high levels of the MAPK and CDK activities, which can lead to the Smad3 linker region becoming highly phosphorylated. Here, we report, for the first time, that mutation of the Smad3 linker phosphorylation sites markedly inhibited primary tumor growth, but significantly increased lung metastasis of breast cancer cell lines. In contrast, mutation of the Smad3 C-tail phosphorylation sites had the opposite effect. We show that mutation of the Smad3 linker phosphorylation sites greatly intensifies all TGFß-induced responses, including growth arrest, apoptosis, reduction in the size of putative cancer stem cell population, epithelial-mesenchymal transition, and invasive activity. Moreover, all TGFß responses were completely lost on mutation of the Smad3 C-tail phosphorylation sites. Our results demonstrate a critical role of the counterbalance between the Smad3 C-tail and linker phosphorylation in tumorigenesis and metastasis. Our findings have important implications for therapeutic intervention of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/genética , Proteína smad3/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Fosforilación/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Cell Sci ; 127(Pt 2): 411-21, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24259667

RESUMEN

In most human cancers the Myc proto-oncogene is highly activated. Dysregulation of Myc oncoprotein contributes to tumorigenesis in numerous tissues and organs. Thus, targeting Myc stability could be a crucial step for cancer therapy. Here we report Smad7 as a key molecule regulating Myc stability and activity by recruiting the F-box protein, Skp2. Ectopic expression of Smad7 downregulated the protein level of Myc without affecting the transcription level, and significantly repressed its transcriptional activity, leading to inhibition of cell proliferation and tumorigenic activity. Furthermore, Smad7 enhanced ubiquitylation of Myc through direct interaction with Myc and recruitment of Skp2. Ablation of Smad7 resulted in less sensitivity to the growth inhibitory effect of TGF-ß by inducing stable Myc expression. In conclusion, these findings that Smad7 functions in Myc oncoprotein degradation and enhances the cytostatic effect of TGF-ß signaling provide a possible new therapeutic approach for cancer treatment.


Asunto(s)
Citostáticos/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proto-Oncogenes Mas , Transcripción Genética/efectos de los fármacos , Ubiquitina/metabolismo
9.
PLoS One ; 7(3): e32705, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22427868

RESUMEN

Transforming growth factor-ß1 (TGF-ß1) is an important anti-inflammatory cytokine that modulates and resolves inflammatory responses. Recent studies have demonstrated that inflammation enhances neoplastic risk and potentiates tumor progression. In the evolution of cancer, pro-inflammatory cytokines such as IL-1ß must overcome the anti-inflammatory effects of TGF-ß to boost pro-inflammatory responses in epithelial cells. Here we show that IL-1ß or Lipopolysaccharide (LPS) suppresses TGF-ß-induced anti-inflammatory signaling in a NF-κB-independent manner. TRAF6, a key molecule in IL-1ß signaling, mediates this suppressive effect through interaction with the type III TGF-ß receptor (TßRIII), which is TGF-ß-dependent and requires type I TGF-ß receptor (TßRI) kinase activity. TßRI phosphorylates TßRIII at residue S829, which promotes the TRAF6/TßRIII interaction and consequent sequestration of TßRIII from the TßRII/TßRI complex. Our data indicate that IL-1ß enhances the pro-inflammatory response by suppressing TGF-ß signaling through TRAF6-mediated sequestration of TßRIII, which may be an important contributor to the early stages of tumor progression.


Asunto(s)
Inflamación/inmunología , Interleucina-1beta/metabolismo , Proteoglicanos/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Fraccionamiento Celular , Fragmentación del ADN , Técnicas de Silenciamiento del Gen , Etiquetado Corte-Fin in Situ , Lipopolisacáridos , Ratones , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , ARN Interferente Pequeño/genética
10.
Proc Natl Acad Sci U S A ; 108(50): 20048-53, 2011 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-22123967

RESUMEN

Human artificial chromosome (HAC)-based vectors offer a promising system for delivery and expression of full-length human genes of any size. HACs avoid the limited cloning capacity, lack of copy number control, and insertional mutagenesis caused by integration into host chromosomes that plague viral vectors. We previously described a synthetic HAC that can be easily eliminated from cell populations by inactivation of its conditional kinetochore. Here, we demonstrate the utility of this HAC, which has a unique gene acceptor site, for delivery of full-length genes and correction of genetic deficiencies in human cells. A battery of functional tests was performed to demonstrate expression of NBS1 and VHL genes from the HAC at physiological levels. We also show that phenotypes arising from stable gene expression can be reversed when cells are "cured" of the HAC by inactivating its kinetochore in proliferating cell populations, a feature that provides a control for phenotypic changes attributed to expression of HAC-encoded genes. This generation of human artificial chromosomes should be suitable for studies of gene function and therapeutic applications.


Asunto(s)
Centrómero/genética , Cromosomas Artificiales Humanos/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Animales , Autoantígenos/metabolismo , Células CHO , Proteínas de Ciclo Celular/genética , Proteína A Centromérica , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas Artificiales de Levadura/genética , Clonación Molecular , Cricetinae , Cricetulus , Expresión Génica , Prueba de Complementación Genética , Genoma Humano/genética , Humanos , Hibridación Fluorescente in Situ , Integrasas/metabolismo , Mutagénesis Insercional/genética , Proteínas Nucleares/genética , Recombinación Genética/genética , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
11.
J Dermatol Sci ; 50(1): 15-23, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18078741

RESUMEN

BACKGROUND: Increasing evidence shows persistent phenotypic alterations in fibroblasts from non-healing human chronic wounds, which may result in faulty extracellular matrix deposition and keratinocyte migration. We have previously shown that these cells are characterized by morphological changes, low proliferative potential and unresponsiveness to TGF-beta1, and down regulated phosphorylation of Smad 2/3 and p42/44 MAPK from decreased expression of the TGF-beta type II receptor. OBJECTIVE: To identify genes and proteins that may be differentially expressed in chronic wounds and their cultured fibroblasts. METHODS: Differential display analysis with 120 random primer sets was used in fibroblasts from human venous ulcers and acute wounds created on the ipsilateral thighs of the same patients. Positive differential results were confirmed by RT-PCR. Immunohistochemistry of cultured fibroblasts and tissues was used to determine the expression of differentially expressed proteins. RESULTS: A total of 16 differentially expressed genes were identified and cloned. The only candidate gene that was differentially expressed in all patients and confirmed by repeated differential display testing and RT-PCR was beta ig-h3, a TGF-beta-induced gene involved in cell adhesion, migration, and proliferation. Decreased expression of beta ig-h3 in chronic wounds and their fibroblasts was further confirmed by Western blot and immunostaining. CONCLUSION: These findings point to beta ig-h3 as an important gene characterizing the abnormal phenotype of chronic wound fibroblasts. Corrective measures to increase the expression of this protein might have therapeutic potential.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Fibroblastos/metabolismo , Factor de Crecimiento Transformador beta/genética , Heridas y Lesiones/metabolismo , Enfermedad Crónica , Proteínas de la Matriz Extracelular/análisis , Humanos , Inmunohistoquímica , Reacción en Cadena de la Polimerasa , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta1/fisiología
12.
Cell Signal ; 18(9): 1439-46, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16413169

RESUMEN

Dynamin-1 is a GTP-hydrolyzing protein and a key element in the clathrin-mediated endocytosis of secretory granules and neurovesicles at the plasma membrane. The unique receptor-like protein tyrosine phosphatase, PTP-NP/Phogrin/IAR/IA-2, is associated with neuroendocrine secretory granules and is highly expressed in the brain. Here, we show by confocal microscopy and biochemical studies that PTP-NP rapidly associates with Dynamin-1 in a depolarization-dependent manner and regulates Dynamin-1 GTPase activity upon KCl depolarization of rat primary hippocampal neurons. Depolarization of primary neurons induced direct association of PTP-NP with Dynamin-1 within 30 s. This association resulted in significant inhibition of Dynamin-1 GTPase activity (approximately 75% inhibition). Mutation within the phosphatase domain of PTP-NP (PTP-NP(D947A)) abolished the direct interaction of PTP-NP with Dynamin-1 and failed to inhibit Dynamin-1 GTPase activity. To further confirm the endogenous interaction of Dynamin-1 with wild-type PTP-NP, Dynamin-1 was purified biochemically from rat brain and its interaction with purified PTP-NP was analyzed. Highly purified Dynamin-1 specifically associated with wild-type PTP-NP and not with mutated PTP-NP, resulting in significant inhibition (approximately 70%) of Dynamin-1 GTPase activity. This is the first report to suggest a novel function of this unique receptor-type tyrosine phosphatase as a potential regulator of Dynamin-1 GTPase activity upon neuronal depolarization.


Asunto(s)
Autoantígenos/metabolismo , Dinamina I , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/metabolismo , Neuronas/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Alanina/metabolismo , Animales , Autoantígenos/genética , Dinamina I/antagonistas & inhibidores , Dinamina I/metabolismo , Endocitosis/fisiología , Hipocampo/citología , Proteínas de la Membrana/genética , Neuronas/citología , Proteínas Tirosina Fosfatasas/genética , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Proteínas Tirosina Fosfatasas Clase 8 Similares a Receptores
13.
J Cell Sci ; 118(Pt 23): 5537-48, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16306221

RESUMEN

The neuronal nuclear matrix protein, NRP/B, contains a BTB domain and kelch repeats and is expressed in primary neurons but not in primary glial cells. To examine the function of NRP/B in neurons, we analyzed the structure/function of the NRP/B-BTB domain and its role in neurite outgrowth. Based on three-dimensional modeling of NRP/B, we generated an NRP/B-BTB mutant containing three mutations in the conserved amino acids D47A, H60A and R61D that was termed BTB mutant A. BTB mutant A significantly reduced the dimerization of NRP/B compared to wild-type NRP/B. The NRP/B-BTB domain was required for nuclear localization and mediated the association of NRP/B with p110RB through the TR subdomain within the B pocket of p110RB. Overexpression of wild-type NRP/B and NRP/B-BTB domain significantly induced neurite outgrowth in PC12 cells and enhanced the G0-G1 cell population by approximately 23% compared to the control cells, whereas NRP/B-BTB mutant A reduced neurite outgrowth by 70-80%, and inhibited NRP/B-p110RB association. Single cell microinjection of NRP/B-specific antibodies also blocked the neurite outgrowth of PC12 cells upon NGF stimulation. Interference of NRP/B expression by small interfering RNA (NRP/B-siRNA) inhibited neurite outgrowth and suppressed the NGF-induced outgrowth of neurites in PC12 cells. Additionally, p110RB phosphorylation at serine residue 795 was significantly reduced in PC12 cells treated with NRP/B siRNA compared to those treated with control GFP-siRNA, indicating that p110RB is a downstream target of NRP/B. Thus, the BTB domain of NRP/B regulates neurite outgrowth through its interaction with the TR subdomain within the B pocket of p110RB, and the conserved amino acids D47A, H60A and R61D within this domain of NRP/B are crucial residues for neurite extension in neuronal cells. These findings support a role for the BTB-domain of NRP/B as an important regulator of neuronal differentiation.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Microfilamentos/genética , Neuritas/fisiología , Neuropéptidos/genética , Proteínas Nucleares/genética , Animales , Anticuerpos/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/farmacología , Regulación de la Expresión Génica , Humanos , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/farmacología , Mutación , Neuritas/efectos de los fármacos , Neuritas/ultraestructura , Neuronas/citología , Neuronas/fisiología , Neuropéptidos/metabolismo , Neuropéptidos/farmacología , Proteínas Nucleares/metabolismo , Proteínas Nucleares/farmacología , Células PC12 , Fosforilación , Conformación Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño/farmacología , Ratas
14.
J Neurochem ; 92(5): 1191-203, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15715669

RESUMEN

Rearrangement of the cytoskeleton leading to the extension of cellular processes is essential for the myelination of axons by oligodendrocytes. We observed that the actin-binding protein, Mayven, is expressed during all stages of the oligodendrocyte lineage, and that its expression is up-regulated during oligodendrocyte differentiation. Mayven is localized in the cytoplasm and along the cell processes. Mayven also binds actin, and is involved in the cytoskeletal reorganization in oligodendrocyte precursor cells (O-2A cells) that leads to process elongation. Mayven overexpression resulted in an increase in the process outgrowth of O-2A cells and in the lengths of the processes, while microinjection of Mayven-specific antibodies inhibited process extension in these cells. Furthermore, O-2A cells transduced with recombinant retroviral sense Mayven (pMIG-W-Mayven) showed an increase in the number of oligodendrocyte processes with outgrowth, while recombinant retroviral antisense Mayven (pMIG-W-Mayven-AS) blocked O-2A process extension. Interestingly, co-localization and association of Mayven with Fyn kinase were found in O-2A cells, and these interactions were increased during the outgrowth of oligodendrocyte processes. This association was mediated via the SH3 domain ligand (a.a. 1-45) of Mayven and the SH3 domain of Fyn, suggesting that Mayven may act as a linker to bind Fyn, via its N-terminus. Thus, Mayven plays a role in the dynamics of cytoskeletal rearrangement leading to the process extension of oligodendrocytes.


Asunto(s)
Extensiones de la Superficie Celular/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Microfilamentos/farmacología , Proteínas del Tejido Nervioso/farmacología , Oligodendroglía/efectos de los fármacos , Prosencéfalo/citología , Actinas/metabolismo , Animales , Anticuerpos/farmacología , Sitios de Unión/fisiología , Northern Blotting/métodos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Tamaño de la Célula/efectos de los fármacos , Extensiones de la Superficie Celular/fisiología , Extensiones de la Superficie Celular/virología , Células Cultivadas , Interacciones Farmacológicas , Embrión de Mamíferos , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica/métodos , Inmunoprecipitación/métodos , Proteínas de Microfilamentos/inmunología , Microinyecciones/métodos , Microscopía Confocal/métodos , Proteína Básica de Mielina/metabolismo , Proteínas del Tejido Nervioso/inmunología , Antígenos O/metabolismo , Oligodendroglía/metabolismo , Oligodendroglía/virología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-fyn , Ratas , Proteínas Recombinantes/metabolismo , Médula Espinal/citología , Médula Espinal/metabolismo , Fracciones Subcelulares/metabolismo , Transfección/métodos , Familia-src Quinasas/metabolismo
15.
J Biol Chem ; 279(27): 28458-65, 2004 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-15107418

RESUMEN

Activin receptor-like kinase (ALK)7 is a type I serine/threonine kinase receptor of the transforming growth factor (TGF)-beta family of proteins that has similar properties to other type I receptors when activated. To see whether ALK7 can induce apoptosis as can some of the other ALK proteins, we infected the FaO rat hepatoma cell line with adenovirus expressing a constitutively active form of the ALK7. Cells infected with active ALK7 adenovirus showed an apoptotic-positive phenotype, as opposed to those that were infected with a control protein. DNA fragmentation assays and fluorescence-activated cell sorter analysis also indicated that ALK7 infection induced apoptosis in FaO cells. We also confirmed this finding in Hep3B human hepatoma cells by transiently transfecting the constitutively active form of ALK7, ALK7(T194D). Investigation into the downstream targets and mechanisms involved in ALK7-induced apoptosis revealed that the TGF-beta signaling intermediates, Smad2 and -3, were activated, as well as the MAPKs JNK and p38. In addition, caspase-3 and -9 were also activated, and cytochrome c release from the mitochondria was observed. Short interfering RNA-mediated inhibition of Smad3 markedly suppressed ALK7-induced caspase-3 activation. Treatment with protein synthesis inhibitors or the expression of the dominant-negative form of the stress-activated protein/extracellular signal-regulated kinase 1 abolished not only JNK activation but apoptosis as well. Taken together, these results suggest that ALK7 induces apoptosis through activation of the traditional TGF-beta pathway components, thus resulting in new gene transcription and JNK and p38 activation that initiates cross-talk with the cellular stress death pathway and ultimately leads to apoptosis.


Asunto(s)
Receptores de Activinas Tipo I/metabolismo , Apoptosis , Carcinoma Hepatocelular/patología , Proteínas de Unión al ADN/metabolismo , Sistema de Señalización de MAP Quinasas , Transactivadores/metabolismo , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Western Blotting , Carcinoma Hepatocelular/metabolismo , Caspasa 3 , Caspasa 9 , Caspasas/metabolismo , Línea Celular , Línea Celular Tumoral , Separación Celular , Supervivencia Celular , Citocromos c/metabolismo , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Genes Dominantes , Vectores Genéticos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Plásmidos/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Proteína Smad2 , Proteína smad3 , Transfección , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1 , Proteínas Quinasas p38 Activadas por Mitógenos
16.
J Biol Chem ; 278(38): 36661-8, 2003 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-12844492

RESUMEN

Vascular endothelial growth factor (VEGF) plays a significant role in blood-brain barrier breakdown and angiogenesis after brain injury. VEGF-induced endothelial cell migration is a key step in the angiogenic response and is mediated by an accelerated rate of focal adhesion complex assembly and disassembly. In this study, we identified the signaling mechanisms by which VEGF regulates human brain microvascular endothelial cell (HBMEC) integrity and assembly of focal adhesions, complexes comprised of scaffolding and signaling proteins organized by adhesion to the extracellular matrix. We found that VEGF treatment of HBMECs plated on laminin or fibronectin stimulated cytoskeletal organization and increased focal adhesion sites. Pretreating cells with VEGF antibodies or with the specific inhibitor SU-1498, which inhibits Flk-1/KDR receptor phosphorylation, blocked the ability of VEGF to stimulate focal adhesion assembly. VEGF induced the coupling of focal adhesion kinase (FAK) to integrin alphavbeta5 and tyrosine phosphorylation of the cytoskeletal components paxillin and p130cas. Additionally, FAK and related adhesion focal tyrosine kinase (RAFTK)/Pyk2 kinases were tyrosine-phosphorylated by VEGF and found to be important for focal adhesion sites. Overexpression of wild type RAFTK/Pyk2 increased cell spreading and the migration of HBMECs, whereas overexpression of catalytically inactive mutant RAFTK/Pyk2 markedly suppressed HBMEC spreading ( approximately 70%), adhesion ( approximately 82%), and migration ( approximately 65%). Furthermore, blocking of FAK by the dominant-interfering mutant FRNK (FAK-related non-kinase) significantly inhibited HBMEC spreading and migration and also disrupted focal adhesions. Thus, these studies define a mechanism for the regulatory role of VEGF in focal adhesion complex assembly in HBMECs via activation of FAK and RAFTK/Pyk2.


Asunto(s)
Encéfalo/irrigación sanguínea , Endotelio Vascular/citología , Adhesiones Focales/metabolismo , Microcirculación/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas , Factor A de Crecimiento Endotelial Vascular/fisiología , Adenoviridae/genética , Western Blotting , Línea Celular , Movimiento Celular , Células Cultivadas , Proteína Sustrato Asociada a CrK , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Activación Enzimática , Matriz Extracelular/metabolismo , Fibronectinas/metabolismo , Quinasa 1 de Adhesión Focal , Quinasa 2 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Genes Dominantes , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Immunoblotting , Inmunohistoquímica , Integrinas/metabolismo , Laminina/metabolismo , Proteínas Luminiscentes/metabolismo , Microscopía Confocal , Modelos Biológicos , Mutación , Paxillin , Fosfoproteínas/metabolismo , Fosforilación , Pruebas de Precipitina , Unión Proteica , Receptores de Vitronectina/metabolismo , Proteínas Recombinantes/metabolismo , Proteína p130 Similar a la del Retinoblastoma , Transducción de Señal , Tirosina/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
J Immunol ; 170(5): 2629-37, 2003 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-12594291

RESUMEN

The integrity of the blood-brain barrier (BBB) is critical for normal brain function. Neuropathological abnormalities in AIDS patients have been associated with perivascular HIV-infected macrophages, gliosis, and abnormalities in the permeability of the BBB. The processes by which HIV causes these pathological conditions are not well understood. To characterize the mechanism by which HIV-1 Tat protein modulates human brain microvascular endothelial cell (HBMEC) functions, we studied the effects of HIV-1 Tat in modulating HBMEC apoptosis and permeability. Treatment of HBMEC with HIV-1 Tat led to Flk-1/KDR and Flt-4 receptor activation and the release of NO. The protein levels of endothelial NO synthase (NOS) and inducible NOS were increased by HIV-1 Tat stimulation. Importantly, HIV-1 Tat caused apoptosis of HBMEC, as evidenced by changes in the cleavage of poly(A)DP-ribose polymerase, DNA laddering, and incorporation of fluorescein into the nicked chromosomal DNA (TUNEL assay). HIV-1 Tat-mediated apoptosis in HBMEC was significantly inhibited in the presence of N-nitro-L-arginine methyl ester (an inhibitor of NOS) and wortmannin (a phosphoinositol 3-kinase inhibitor). Furthermore, HIV-1 Tat treatment significantly increased HBMEC permeability, and pretreatment with both N-nitro-L-arginine methyl ester and wortmannin inhibited the Tat-induced permeability. Taken together, these results indicate that dysregulated production of NO by HIV-1 Tat plays a pivotal role in brain endothelial injury, resulting in the irreversible loss of BBB integrity, which may lead to enhanced infiltration of virus-carrying cells across the BBB.


Asunto(s)
Apoptosis/fisiología , Endotelio Vascular/fisiología , Endotelio Vascular/virología , Productos del Gen tat/fisiología , VIH-1/fisiología , Apoptosis/efectos de los fármacos , Permeabilidad de la Membrana Celular/efectos de los fármacos , Permeabilidad de la Membrana Celular/fisiología , Tamaño de la Célula/efectos de los fármacos , Tamaño de la Célula/fisiología , Células Cultivadas , Fragmentación del ADN/efectos de los fármacos , Fragmentación del ADN/fisiología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Productos del Gen tat/farmacología , Humanos , Microcirculación/citología , Microcirculación/metabolismo , Microcirculación/fisiología , Microcirculación/virología , Óxido Nítrico/metabolismo , Óxido Nítrico/fisiología , Óxido Nítrico Sintasa/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Recombinantes/farmacología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...