Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Dev Biol ; 494: 60-70, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36509125

RESUMEN

Neuroepithelial cells balance tissue growth requirement with the morphogenetic imperative of closing the neural tube. They apically constrict to generate mechanical forces which elevate the neural folds, but are thought to apically dilate during mitosis. However, we previously reported that mitotic neuroepithelial cells in the mouse posterior neuropore have smaller apical surfaces than non-mitotic cells. Here, we document progressive apical enrichment of non-muscle myosin-II in mitotic, but not non-mitotic, neuroepithelial cells with smaller apical areas. Live-imaging of the chick posterior neuropore confirms apical constriction synchronised with mitosis, reaching maximal constriction by anaphase, before division and re-dilation. Mitotic apical constriction amplitude is significantly greater than interphase constrictions. To investigate conservation in humans, we characterised early stages of iPSC differentiation through dual SMAD-inhibition to robustly produce pseudostratified neuroepithelia with apically enriched actomyosin. These cultured neuroepithelial cells achieve an equivalent apical area to those in mouse embryos. iPSC-derived neuroepithelial cells have large apical areas in G2 which constrict in M phase and retain this constriction in G1/S. Given that this differentiation method produces anterior neural identities, we studied the anterior neuroepithelium of the elevating mouse mid-brain neural tube. Instead of constricting, mid-brain mitotic neuroepithelial cells have larger apical areas than interphase cells. Tissue geometry differs between the apically convex early midbrain and flat posterior neuropore. Culturing human neuroepithelia on equivalently convex surfaces prevents mitotic apical constriction. Thus, neuroepithelial cells undergo high-amplitude apical constriction synchronised with cell cycle progression but the timing of their constriction if influenced by tissue geometry.


Asunto(s)
Mitosis , Sistema Nervioso , Humanos , Animales , Ratones , Constricción , Ciclo Celular , Diferenciación Celular/fisiología
2.
Proc Natl Acad Sci U S A ; 113(1): E91-100, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26699507

RESUMEN

Optogenetics provides new ways to activate gene transcription; however, no attempts have been made as yet to modulate mammalian transcription factors. We report the light-mediated regulation of the repressor element 1 (RE1)-silencing transcription factor (REST), a master regulator of neural genes. To tune REST activity, we selected two protein domains that impair REST-DNA binding or recruitment of the cofactor mSin3a. Computational modeling guided the fusion of the inhibitory domains to the light-sensitive Avena sativa light-oxygen-voltage-sensing (LOV) 2-phototrophin 1 (AsLOV2). By expressing AsLOV2 chimeras in Neuro2a cells, we achieved light-dependent modulation of REST target genes that was associated with an improved neural differentiation. In primary neurons, light-mediated REST inhibition increased Na(+)-channel 1.2 and brain-derived neurotrophic factor transcription and boosted Na(+) currents and neuronal firing. This optogenetic approach allows the coordinated expression of a cluster of genes impinging on neuronal activity, providing a tool for studying neuronal physiology and correcting gene expression changes taking place in brain diseases.


Asunto(s)
Regulación de la Expresión Génica , Neuronas/metabolismo , Optogenética/métodos , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/genética , Animales , Avena/genética , Línea Celular Tumoral , Cromatina/metabolismo , ADN/química , Ensayo de Cambio de Movilidad Electroforética , Ratones , Proteínas de Plantas/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/química , Transcripción Genética , Transducción Genética
3.
EMBO J ; 32(22): 2994-3007, 2013 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-24149584

RESUMEN

Intrinsic homeostasis enables neuronal circuits to maintain activity levels within an appropriate range by modulating neuronal voltage-gated conductances, but the signalling pathways involved in this process are largely unknown. We characterized the process of intrinsic homeostasis induced by sustained electrical activity in cultured hippocampal neurons based on the activation of the Repressor Element-1 Silencing Transcription Factor/Neuron-Restrictive Silencer Factor (REST/NRSF). We showed that 4-aminopyridine-induced hyperactivity enhances the expression of REST/NRSF, which in turn, reduces the expression of voltage-gated Na(+) channels, thereby decreasing the neuronal Na(+) current density. This mechanism plays an important role in the downregulation of the firing activity at the single-cell level, re-establishing a physiological spiking activity in the entire neuronal network. Conversely, interfering with REST/NRSF expression impaired this homeostatic response. Our results identify REST/NRSF as a critical factor linking neuronal activity to the activation of intrinsic homeostasis and restoring a physiological level of activity in the entire neuronal network.


Asunto(s)
Homeostasis/fisiología , Proteínas Represoras/fisiología , 4-Aminopiridina/farmacología , Animales , Células Cultivadas , Hipocampo/citología , Hipocampo/fisiología , Homeostasis/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Red Nerviosa , Neuronas/fisiología
4.
Hum Mol Genet ; 22(11): 2186-99, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23406870

RESUMEN

Synapsin I (SynI) is a synaptic vesicle (SV) phosphoprotein playing multiple roles in synaptic transmission and plasticity by differentially affecting crucial steps of SV trafficking in excitatory and inhibitory synapses. SynI knockout (KO) mice are epileptic, and nonsense and missense mutations in the human SYN1 gene have a causal role in idiopathic epilepsy and autism. To get insights into the mechanisms of epileptogenesis linked to SYN1 mutations, we analyzed the effects of the recently identified Q555X mutation on neurotransmitter release dynamics and short-term plasticity (STP) in excitatory and inhibitory synapses. We used patch-clamp electrophysiology coupled to electron microscopy and multi-electrode arrays to dissect synaptic transmission of primary SynI KO hippocampal neurons in which the human wild-type and mutant SynI were expressed by lentiviral transduction. A parallel decrease in the SV readily releasable pool in inhibitory synapses and in the release probability in excitatory synapses caused a marked reduction in the evoked synchronous release. This effect was accompanied by an increase in asynchronous release that was much more intense in excitatory synapses and associated with an increased total charge transfer. Q555X-hSynI induced larger facilitation and post-tetanic potentiation in excitatory synapses and stronger depression after long trains in inhibitory synapses. These changes were associated with higher network excitability and firing/bursting activity. Our data indicate that imbalances in STP and release dynamics of inhibitory and excitatory synapses trigger network hyperexcitability potentially leading to epilepsy/autism manifestations.


Asunto(s)
Epilepsia/genética , Epilepsia/metabolismo , Plasticidad Neuronal/genética , Sinapsis/metabolismo , Sinapsinas/genética , Sinapsinas/metabolismo , Animales , Femenino , Expresión Génica , Hipocampo/metabolismo , Humanos , Espacio Intracelular/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Técnicas de Placa-Clamp , Fenotipo , Multimerización de Proteína , Transporte de Proteínas , Sinapsinas/química , Potenciales Sinápticos , Vesículas Sinápticas/metabolismo
5.
J Biol Chem ; 288(5): 3227-39, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23250796

RESUMEN

The development and function of the nervous system are directly dependent on a well defined pattern of gene expression. Indeed, perturbation of transcriptional activity or epigenetic modifications of chromatin can dramatically influence neuronal phenotypes. The phosphoprotein synapsin I (Syn I) plays a crucial role during axonogenesis and synaptogenesis as well as in synaptic transmission and plasticity of mature neurons. Abnormalities in SYN1 gene expression have been linked to important neuropsychiatric disorders, such as epilepsy and autism. SYN1 gene transcription is suppressed in non-neural tissues by the RE1-silencing transcription factor (REST); however, the molecular mechanisms that allow the constitutive expression of this genetic region in neurons have not been clarified yet. Herein we demonstrate that a conserved region of human and mouse SYN1 promoters contains cis-sites for the transcriptional activator Sp1 in close proximity to REST binding motifs. Through a series of functional assays, we demonstrate a physical interaction of Sp1 on the SYN1 promoter and show that REST directly inhibits Sp1-mediated transcription, resulting in SYN1 down-regulation. Upon differentiation of neuroblastoma Neuro2a cells, we observe a decrease in endogenous REST and a higher stability of Sp1 on target GC boxes, resulting in an increase of SYN1 transcription. Moreover, methylation of Sp1 cis-sites in the SYN1 promoter region could provide an additional level of transcriptional regulation. Our results introduce Sp1 as a fundamental activator of basal SYN1 gene expression, whose activity is modulated by the neural master regulator REST and CpG methylation.


Asunto(s)
Islas de CpG/genética , Metilación de ADN/genética , Proteínas Represoras/metabolismo , Factor de Transcripción Sp1/metabolismo , Sinapsinas/genética , Activación Transcripcional/genética , Animales , Secuencia de Bases , Diferenciación Celular/genética , Línea Celular , Secuencia Conservada/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Genéticos , Datos de Secuencia Molecular , Neuronas/citología , Neuronas/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Estabilidad Proteica , Transcripción Genética
6.
Circ Res ; 110(3): 394-405, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22207709

RESUMEN

RATIONALE: The nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) is an important regulator of gene transcription in vascular cells and mediates the vascular protection observed with antidiabetic glitazones. OBJECTIVE: To determine the molecular mechanism of ligand-dependent transrepression in vascular smooth muscle cells and their impact on the vascular protective actions of PPARγ. METHODS AND RESULTS: Here, we report a molecular pathway in vascular smooth muscle cells by which ligand-activated PPARγ represses transcriptional activation of the matrix-degrading matrix metalloproteinase-9 (MMP-9) gene, a crucial mediator of vascular injury. PPARγ-mediated transrepression of the MMP-9 gene was dependent on the presence of the high-mobility group A1 (HMGA1) protein, a gene highly expressed in vascular smooth muscle cells, newly identified by oligonucleotide array expression analysis. Transrepression of MMP-9 by PPARγ and regulation by HMGA1 required PPARγ SUMOylation at K367. This process was associated with formation of a complex between PPARγ, HMGA1, and the SUMO E2 ligase Ubc9 (ubiquitin-like protein SUMO-1 conjugating enzyme). After PPARγ ligand stimulation, HMGA1 and PPARγ were recruited to the MMP-9 promoter, which facilitated binding of SMRT (silencing mediator of retinoic acid and thyroid hormone receptor), a nuclear corepressor involved in transrepression. The relevance of HMGA1 for vascular PPARγ signaling was underlined by the complete absence of vascular protection through a PPARγ ligand in HMGA1(-/-) mice after arterial wire injury. CONCLUSIONS: The present data suggest that ligand-dependent formation of HMGA1-Ubc9-PPARγ complexes facilitates PPARγ SUMOylation, which results in the prevention of SMRT corepressor clearance and induction of MMP-9 transrepression. These data provide new information on PPARγ-dependent vascular transcriptional regulation and help us to understand the molecular consequences of therapeutic interventions with PPARγ ligands in the vasculature.


Asunto(s)
Proteína HMGA1a/metabolismo , Músculo Liso Vascular/metabolismo , PPAR gamma/metabolismo , Transducción de Señal/fisiología , Transcripción Genética/fisiología , Animales , Endotelina-1/metabolismo , Arteria Femoral/efectos de los fármacos , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Proteína HMGA1a/deficiencia , Proteína HMGA1a/genética , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Modelos Animales , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/lesiones , FN-kappa B/metabolismo , Tiazolidinedionas/farmacología , Enzimas Ubiquitina-Conjugadoras/metabolismo
7.
Nat Med ; 11(7): 765-73, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15924147

RESUMEN

Type 2 diabetes mellitus is a widespread disease, affecting millions of people globally. Although genetics and environmental factors seem to have a role, the cause of this metabolic disorder is largely unknown. Here we report a genetic flaw that markedly reduced the intracellular expression of the high mobility group A1 (HMGA1) protein, and adversely affected insulin receptor expression in cells and tissues from four subjects with insulin resistance and type 2 diabetes. Restoration of HMGA1 protein expression in subjects' cells enhanced INSR gene transcription, and restored cell-surface insulin receptor protein expression and insulin-binding capacity. Loss of Hmga1 expression, induced in mice by disrupting the Hmga1 gene, considerably decreased insulin receptor expression in the major targets of insulin action, largely impaired insulin signaling and severely reduced insulin secretion, causing a phenotype characteristic of human type 2 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Proteína HMGA1a/genética , Resistencia a la Insulina/genética , Regiones no Traducidas 3' , Adolescente , Adulto , Animales , Antígenos CD , Células Cultivadas , Niño , Femenino , Regulación de la Expresión Génica , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4 , Proteína HMGA1a/metabolismo , Homeostasis/genética , Humanos , Insulina/metabolismo , Secreción de Insulina , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/genética , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Mutantes , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Mutación , Páncreas/metabolismo , Páncreas/patología , Linaje , Tomografía de Emisión de Positrones , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Estabilidad del ARN , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Transducción de Señal
8.
JAMA ; 305(9): 903-12, 2011 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-21364139

RESUMEN

CONTEXT: High-mobility group A1 (HMGA1) protein is a key regulator of insulin receptor (INSR) gene expression. We previously identified a functional HMGA1 gene variant in 2 insulin-resistant patients with decreased INSR expression and type 2 diabetes mellitus (DM). OBJECTIVE: To examine the association of HMGA1 gene variants with type 2 DM. DESIGN, SETTINGS, AND PARTICIPANTS: Case-control study that analyzed the HMGA1 gene in patients with type 2 DM and controls from 3 populations of white European ancestry. Italian patients with type 2 DM (n = 3278) and 2 groups of controls (n = 3328) were attending the University of Catanzaro outpatient clinics and other health care sites in Calabria, Italy, during 2003-2009; US patients with type 2 DM (n = 970) were recruited in Northern California clinics between 1994 and 2005 and controls (n = 958) were senior athletes without DM collected in 2004 and 2009; and French patients with type 2 DM (n = 354) and healthy controls (n = 50) were enrolled at the University of Reims in 1992. Genomic DNA was either directly sequenced or analyzed for specific HMGA1 mutations. Messenger RNA and protein expression for HMGA1 and INSR were measured in both peripheral lymphomonocytes and cultured Epstein-Barr virus-transformed lymphoblasts from patients with type 2 DM and controls. MAIN OUTCOME MEASURES: The frequency of HMGA1 gene variants among cases and controls. Odds ratios (ORs) for type 2 DM were estimated by logistic regression analysis. RESULTS: The most frequent functional HMGA1 variant, IVS5-13insC, was present in 7% to 8% of patients with type 2 DM in all 3 populations. The prevalence of IVS5-13insC variant was higher among patients with type 2 DM than among controls in the Italian population (7.23% vs 0.43% in one control group; OR, 15.77 [95% confidence interval {CI}, 8.57-29.03]; P < .001 and 7.23% vs 3.32% in the other control group; OR, 2.03 [95% CI, 1.51-3.43]; P < .001). In the US population, the prevalence of IVS5-13insC variant was 7.7% among patients with type 2 DM vs 4.7% among controls (OR, 1.64 [95% CI, 1.05-2.57]; P = .03). In the French population, the prevalence of IVS5-13insC variant was 7.6% among patients with type 2 DM and 0% among controls (P = .046). In the Italian population, 3 other functional variants were observed. When all 4 variants were analyzed, HMGA1 defects were present in 9.8% of Italian patients with type 2 DM and 0.6% of controls. In addition to the IVS5 C-insertion, the c.310G>T (p.E104X) variant was found in 14 patients and no controls (Bonferroni-adjusted P = .01); the c.*82G>A variant (rs2780219) was found in 46 patients and 5 controls (Bonferroni-adjusted P < .001); the c.*369del variant was found in 24 patients and no controls (Bonferroni-adjusted P < .001). In circulating monocytes and Epstein-Barr virus-transformed lymphoblasts from patients with type 2 DM and the IVS5-13insC variant, the messenger RNA levels and protein content of both HMGA1 and the INSR were decreased by 40% to 50%, and these defects were corrected by transfection with HMGA1 complementary DNA. CONCLUSIONS: Compared with healthy controls, the presence of functional HMGA1 gene variants in individuals of white European ancestry was associated with type 2 DM.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Predisposición Genética a la Enfermedad , Proteínas HMGA/genética , Regiones no Traducidas 3'/genética , Anciano , Alelos , Estudios de Casos y Controles , Exones/genética , Femenino , Francia , Variación Genética , Heterocigoto , Humanos , Italia , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Regiones Promotoras Genéticas/genética , Sitios de Empalme de ARN/genética , Estados Unidos , Población Blanca/genética
9.
BMC Biol ; 7: 24, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19460132

RESUMEN

BACKGROUND: We previously showed that mice lacking the high mobility group A1 gene (Hmga1-knockout mice) developed a type 2-like diabetic phenotype, in which cell-surface insulin receptors were dramatically reduced (below 10% of those in the controls) in the major targets of insulin action, and glucose intolerance was associated with increased peripheral insulin sensitivity. This particular phenotype supports the existence of compensatory mechanisms of insulin resistance that promote glucose uptake and disposal in peripheral tissues by either insulin-dependent or insulin-independent mechanisms. We explored the role of these mechanisms in the regulation of glucose homeostasis by studying the Hmga1-knockout mouse model. Also, the hypothesis that increased insulin sensitivity in Hmga1-deficient mice could be related to the deficit of an insulin resistance factor is discussed. RESULTS: We first show that HMGA1 is needed for basal and cAMP-induced retinol-binding protein 4 (RBP4) gene and protein expression in living cells of both human and mouse origin. Then, by employing the Hmga1-knockout mouse model, we provide evidence for the identification of a novel biochemical pathway involving HMGA1 and the RBP4, whose activation by the cAMP-signaling pathway may play an essential role for maintaining glucose metabolism homeostasis in vivo, in certain adverse metabolic conditions in which insulin action is precluded. In comparative studies of normal and mutant mice, glucagon administration caused a considerable upregulation of HMGA1 and RBP4 expression both at the mRNA and protein level in wild-type animals. Conversely, in Hmga1-knockout mice, basal and glucagon-mediated expression of RBP4 was severely attenuated and correlated inversely with increased Glut4 mRNA and protein abundance in skeletal muscle and fat, in which the activation state of the protein kinase Akt, an important downstream mediator of the metabolic effects of insulin on Glut4 translocation and carbohydrate metabolism, was simultaneously increased. CONCLUSION: These results indicate that HMGA1 is an important modulator of RBP4 gene expression in vivo. Further, they provide evidence for the identification of a novel biochemical pathway involving the cAMP-HMGA1-RBP4 system, whose activation may play a role in glucose homeostasis in both rodents and humans. Elucidating these mechanisms has importance for both fundamental biology and therapeutic implications.


Asunto(s)
AMP Cíclico/metabolismo , Glucosa/metabolismo , Proteína HMGA1a/metabolismo , Homeostasis , Redes y Vías Metabólicas , Proteínas Plasmáticas de Unión al Retinol/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , AMP Cíclico/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Glucagón/administración & dosificación , Glucagón/farmacología , Transportador de Glucosa de Tipo 4/metabolismo , Proteína HMGA1a/deficiencia , Proteína HMGA1a/genética , Homeostasis/efectos de los fármacos , Humanos , Inyecciones Intraperitoneales , Insulina/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Proteínas Plasmáticas de Unión al Retinol/genética , Transducción de Señal/efectos de los fármacos , Transcripción Genética/efectos de los fármacos
10.
Cell Rep ; 26(3): 582-593.e5, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650353

RESUMEN

The neuronal microtubule-associated protein tau, MAPT, is central to the pathogenesis of many dementias. Autosomal-dominant mutations in MAPT cause inherited frontotemporal dementia (FTD), but the underlying pathogenic mechanisms are unclear. Using human stem cell models of FTD due to MAPT mutations, we find that tau becomes hyperphosphorylated and mislocalizes to cell bodies and dendrites in cortical neurons, recapitulating a key early event in FTD. Mislocalized tau in the cell body leads to abnormal microtubule movements in FTD-MAPT neurons that grossly deform the nuclear membrane. This results in defective nucleocytoplasmic transport, which is corrected by microtubule depolymerization. Neurons in the post-mortem human FTD-MAPT cortex have a high incidence of nuclear invaginations, indicating that tau-mediated nuclear membrane dysfunction is an important pathogenic process in FTD. Defects in nucleocytoplasmic transport in FTD point to important commonalities in the pathogenic mechanisms of tau-mediated dementias and ALS-FTD due to TDP-43 and C9orf72 mutations.


Asunto(s)
Transporte Activo de Núcleo Celular/genética , Demencia Frontotemporal/genética , Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Demencia Frontotemporal/patología , Humanos
11.
Cancer Res ; 66(10): 5085-93, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707431

RESUMEN

Various studies have shown that the insulin receptor (IR) is increased in most human breast cancers, and both ligand-dependent malignant transformation and increased cell growth occur in cultured breast cells overexpressing the IR. However, although numerous in vivo and in vitro observations have indicated an important contributory role for the IR in breast cancer cell biology, the molecular mechanisms accounting for increased IR expression in breast tumors have not previously been elucidated. Herein, we did immunoblot analyses of nuclear protein from cultured breast cancer cells and normal and tumoral tissues from breast cancer patients combined with promoter studies by using a series of human wild-type and mutant IR promoter constructs. We provide evidence that IR overexpression in breast cancer is dependent on the assembly of a transcriptionally active multiprotein-DNA complex, which includes the high-mobility group A1 (HMGA1) protein, the developmentally regulated activator protein-2 (AP-2) transcription factor and the ubiquitously expressed transcription factor Sp1. In cultured breast cancer cells and human breast cancer specimens, the expression of AP-2 was significantly higher than that observed in cells and tissues derived from normal breast, and this overexpression paralleled the increase in IR expression. However, AP-2 DNA-binding activity was undetectable with the IR gene promoter, suggesting that transactivation of this gene by AP-2 might occur indirectly through physical and functional cooperation with HMGA1 and Sp1. Our findings support this hypothesis and suggest that in affected individuals, hyperactivation of the AP-2 gene through the overexpression of IR may play a key role in breast carcinogenesis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor de Insulina/biosíntesis , Factor de Transcripción AP-2/biosíntesis , Neoplasias de la Mama/genética , Línea Celular Tumoral , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína HMGA1a/genética , Proteína HMGA1a/metabolismo , Células HeLa , Humanos , Regiones Promotoras Genéticas , Receptor de Insulina/genética , Factor de Transcripción Sp1/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
12.
Arthritis Res Ther ; 14(5): R207, 2012 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-23036517

RESUMEN

INTRODUCTION: Insulin-like growth factor I (IGF-I) regulates articular cartilage homeostasis. During osteoarthritis (OA), the anabolic responses of chondrocytes to IGF-I are likely to be prevented by the enhanced production of IGF-binding proteins (IGFBPs), especially IGFBP-3. The aim of this study is to evaluate whether the architectural transcription factor high mobility group A1 (HMGA1) influences IGFBP-3 overexpression in vitro, in cultured chondrocytic cell lines, and ex vivo, in human osteoarthritic cartilage compared to healthy human cartilage controls. METHODS: Quantitative real-time reverse transcription-PCR (qRT-PCR) was performed to assess the relative transcript levels of HMGA1 and IGFBP-3 in vitro, in the human chondrocytic cell lines T/C-28a4 and C-28/I2. An electrophoretic mobility shift assay (EMSA), chromatin immunoprecipitation (ChIP) and transient transfection assays were performed to investigate the HMGA1-IGFBP-3 gene interaction. Samples of articular cartilage were harvested from osteoarthritic patients and controls and analyzed by qRT-PCR for HMGA1 and IGFBP-3 mRNA levels. RESULTS: A parallelism between HMGA1 protein levels and IGFBP-3 gene expression has been observed in T/C-28a4 and C-28/I2 cells. The interaction of HMGA1 with the IGFBP-3 gene promoter has been demonstrated by EMSA and ChIP. In transient transfections, IGFBP-3 promoter activity increased in cells overexpressing HMGA1 and decreased in cells pretreated with siRNA detected against HMGA1. IGFBP-3 mRNA expression was higher in cartilage from patients with OA, where the increased expression of IGFBP-3 closely paralleled the increased expression of HMGA1 mRNA. CONCLUSIONS: Our observations indicate that increased HMGA1 expression in human chondrocytes is associated with increased expression of IGFBP-3. It is tempting to speculate that, through the regulation of IGFBP3 expression, HMGA1 may act as a pathogenetic factor for OA.


Asunto(s)
Condrocitos/metabolismo , Proteína HMGA1a/metabolismo , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Osteoartritis/metabolismo , Anciano , Cartílago Articular/metabolismo , Cartílago Articular/patología , Estudios de Casos y Controles , Línea Celular , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/patología , Femenino , Regulación de la Expresión Génica , Proteína HMGA1a/efectos de los fármacos , Homeostasis/fisiología , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Osteoartritis/patología , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología
13.
Sci Rep ; 2: 251, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22355763

RESUMEN

High-mobility group AT-hook 1 (HMGA1) protein is an important nuclear factor that activates gene transcription by binding to AT-rich sequences in the promoter region of DNA. We previously demonstrated that HMGA1 is a key regulator of the insulin receptor (INSR) gene and individuals with defects in HMGA1 have decreased INSR expression and increased susceptibility to type 2 diabetes mellitus. In addition, there is evidence that intracellular regulatory molecules that are employed by the INSR signaling system are involved in post-translational modifications of HMGA1, including protein phosphorylation. It is known that phosphorylation of HMGA1 reduces DNA-binding affinity and transcriptional activation. In the present study, we investigated whether activation of the INSR by insulin affected HMGA1 protein phosphorylation and its regulation of gene transcription. Collectively, our findings indicate that HMGA1 is a novel downstream target of the INSR signaling pathway, thus representing a new critical nuclear mediator of insulin action and function.

14.
ACS Appl Mater Interfaces ; 4(6): 2903-11, 2012 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-22583790

RESUMEN

Porous silicon (PSi) is a promising material in several biomedical applications because of its biocompatibility and biodegradability. Despite the plethora of studies focusing on the interaction of cells with micrometer and submicro geometrical features, limited information is available on the response of cells to substrates with a quasi-regular distribution of nanoscopic pores. Here, the behavior of four different cell types is analyzed on two mesoporous (MeP) silicon substrates, with an average pore size of ∼5 (MeP1) and ∼20 nm (MeP2), respectively. On both MeP substrates, cells are observed to spread and adhere in a larger number as compared to flat silicon wafers. At all considered time points, the surface density of the adhering cells nd is larger on the PSi substrate with the smaller average pore size (MeP1). At 60 h, nd is from ∼1.5 to 5 times larger on MeP1 than on MeP2 substrates, depending on the cell type. The higher rates of proliferation are observed for the two neuronal cell types, the mouse neuroblastoma cells (N2A) and the immortalized human cortical neuronal cells (HCN1A). It is speculated that the higher adhesion on MeP1 could be attributed to a preferential matching of the substrate topography with the recently observed multiscale molecular architecture of focal adhesions. These results have implications in the rational development of PSi substrates for supporting cell adhesion and controlling drug release in implants and scaffolds for tissue engineering applications.


Asunto(s)
Silicio/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Adhesión Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Microscopía Confocal , Porosidad , Ingeniería de Tejidos , Vinculina/metabolismo
15.
Nat Commun ; 1: 40, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20975707

RESUMEN

Processed pseudogenes are non-functional copies of normal genes that arise by a process of mRNA retrotransposition. The human genome contains thousands of pseudogenes; however, knowledge regarding their biological role is limited. Previously, we demonstrated that high mobility group A1 (HMGA1) protein regulates the insulin receptor (INSR) gene and that two diabetic patients demonstrated a marked destabilization of HMGA1 mRNA. In this paper we report that this destabilization of HMGA1 mRNA is triggered by enhanced expression of RNA from an HMGA1 pseudogene, HMGA1-p. Targeted knockdown of HMGA1-p mRNA in patient cells results in a reciprocal increase in HMGA1 mRNA stability and expression levels with a parallel correction in cell-surface INSR expression and insulin binding. These data provide evidence for a regulatory role of an expressed pseudogene in humans and establishes a novel mechanistic linkage between pseudogene HMGA1-p expression and type 2 diabetes mellitus.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Proteínas HMGA/metabolismo , Resistencia a la Insulina/genética , Seudogenes/genética , Regiones no Traducidas 3'/genética , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Northern Blotting , Línea Celular , Células Cultivadas , Ensayo de Cambio de Movilidad Electroforética , Silenciador del Gen , Células HEK293 , Proteínas HMGA/genética , Células HeLa , Humanos , Resistencia a la Insulina/fisiología , Ratones , Modelos Biológicos , Datos de Secuencia Molecular , Receptor de Insulina/genética , Receptor de Insulina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
PPAR Res ; 2009: 571365, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19587804

RESUMEN

The insulin receptor (IR) plays a crucial role in mediating the metabolic and proliferative functions triggered by the peptide hormone insulin. There is considerable evidence that abnormalities in both IR expression and function may account for malignant transformation and tumour progression in some human neoplasias, including breast cancer. PPARγ is a ligand-activated, nuclear hormone receptor implicated in many pleiotropic biological functions related to cell survival and proliferation. In the last decade, PPARγ agonists-besides their known action and clinical use as insulin sensitizers-have proved to display a wide range of antineoplastic effects in cells and tissues expressing PPARγ, leading to intensive preclinical research in oncology. PPARγ and activators affect tumours by different mechanisms, involving cell proliferation and differentiation, apoptosis, antiinflammatory, and antiangiogenic effects. We recently provided evidence that PPARγ and agonists inhibit IR by non canonical, DNA-independent mechanisms affecting IR gene transcription. We conclude that IR may be considered a new PPARγ "target" gene, supporting a potential use of PPARγ agonists as antiproliferative agents in selected neoplastic tissues that overexpress the IR.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA