Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Transplant ; 29: 963689720971204, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33150791

RESUMEN

Since the 1970s, rodent and human insulin-secreting pancreatic beta-cell lines have been developed and found useful for studying beta-cell biology. Surprisingly, although the dog has been widely used as a translational model for diabetes, no canine insulin-secreting beta cells have ever been produced. Here, a targeted oncogenesis protocol previously described by some of us for generating human beta cells was adapted to produce canine beta cells. Canine fetal pancreata were obtained by cesarean section between 42 and 55 days of gestation, and fragments of fetal glands were transduced with a lentiviral vector expressing SV40LT under the control of the insulin promoter. Two Lox P sites flanking the sequence allowed subsequent transgene excision by Cre recombinase expression. When grafted into SCID mice, these transduced pancreata formed insulinomas. ACT-164 is the cell line described in this report. Insulin mRNA expression and protein content were lower than reported with adult cells, but the ACT-164 cells were functional, and their insulin production in vitro increased under glucose stimulation. Transgene excision upon Cre expression arrested proliferation and enhanced insulin expression and production. When grafted in SCID mice, intact and excised cells reversed chemically induced diabetes. We have thus produced an excisable canine beta-cell line. These cells may play an important role in the study of several aspects of the cell transplantation procedure including the encapsulation process, which is difficult to investigate in rodents. Although much more work is needed to improve the excision procedure and achieve 100% removal of large T antigen expression, we have shown that functional cells can be obtained and might in the future be used for replacement therapy in diabetic dogs.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Páncreas/enzimología , Páncreas/metabolismo , Animales , Proliferación Celular/genética , Proliferación Celular/fisiología , Perros , Femenino , Insulina/metabolismo , Insulinoma/metabolismo , Integrasas/genética , Integrasas/metabolismo , Ratones SCID , Embarazo , Regiones Promotoras Genéticas/genética , ARN Mensajero/metabolismo
2.
Cell Transplant ; 28(12): 1641-1651, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31450972

RESUMEN

We evaluated the cell composition and function of canine pancreatic pseudoislets (PIs) produced from 42- to 55-day-old fetuses, 1- to 21-day-old pups, and an adult dog pancreas. After mild collagenase treatment, partially digested tissues were cultured for 2-3 weeks. PI production started on culture day 3, was marked for 6 to 9 days, and then stopped. PI production was greatest with the neonatal specimens, reaching about 12 million aggregates per litter (55-day-old fetus) or per pancreas (1-day-old pup). Cell composition at all stages was similar to that in adult pancreatic islets, with predominant ß cells, scant α cells and, most importantly, presence of δ cells. Among pancreatic markers assessed by quantitative real-time PCR (qRT-PCR) mRNA assay, insulin showed the highest expression levels in PIs from newborn and adult pancreas, although these were more than 1000 times lower than in adult islets. Pdx1 mRNA expression was high in PIs from 55-day-old pancreases and was lower at later stages. Consistent with the qRT-PCR results, the insulin content was far lower than reported in adult dog pancreatic islets. However, insulin release by PIs from 1-day-old pups was demonstrated and was stimulated by a high-glucose medium. PIs were transplanted into euglycemic and diabetic SCID mice. In euglycemic animals, the transplant cell composition underwent maturation and transplants were still viable after 6 months. In diabetic mice, the PI transplants produced insulin and partially controlled the hyperglycemia. These data indicate that PIs can be produced ex vivo from canine fetal or postnatal pancreases. Although functional PIs can be obtained, the production yield is most likely insufficient to meet the requirements for diabetic dog transplantation without further innovation in cell culture amplification.


Asunto(s)
Diabetes Mellitus Experimental , Feto/metabolismo , Regulación de la Expresión Génica , Insulina/biosíntesis , Trasplante de Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Organoides , Animales , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/terapia , Perros , Feto/patología , Xenoinjertos , Islotes Pancreáticos/patología , Ratones , Ratones SCID , Organoides/metabolismo , Organoides/patología , Organoides/trasplante
3.
Sci Rep ; 9(1): 1409, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30723217

RESUMEN

Midbrain dopaminergic (DA) neurons are involved in diverse neurological functions, including control of movements, emotions or reward. In turn, their dysfunctions cause severe clinical manifestations in humans, such as the appearance of motor and cognitive symptoms in Parkinson's Disease. The physiology and pathophysiology of these neurons are widely studied, mostly with respect to molecular mechanisms implicating protein-coding genes. In contrast, the contribution of non-coding elements of the genome to DA neuron function is poorly investigated. In this study, we isolated DA neurons from E14.5 ventral mesencephalons in mice, and used RNA-seq and ATAC-seq to establish and describe repertoires of long non-coding RNAs (lncRNAs) and putative DNA regulatory regions specific to this neuronal population. We identified 1,294 lncRNAs constituting the repertoire of DA neurons, among which 939 were novel. Most of them were not found in hindbrain serotonergic (5-HT) neurons, indicating a high degree of cell-specificity. This feature was also observed regarding open chromatin regions, as 39% of the ATAC-seq peaks from the DA repertoire were not detected in the 5-HT neurons. Our work provides for the first time DA-specific catalogues of non-coding elements of the genome that will undoubtedly participate in deepening our knowledge regarding DA neuronal development and dysfunctions.


Asunto(s)
Cromatina/genética , Neuronas Dopaminérgicas/metabolismo , Mesencéfalo/citología , ARN Largo no Codificante/genética , Secuencias Repetitivas de Ácidos Nucleicos/genética , Animales , Secuenciación de Inmunoprecipitación de Cromatina , Femenino , Masculino , Mesencéfalo/embriología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , RNA-Seq , Neuronas Serotoninérgicas/metabolismo , Transcriptoma
4.
Pharmacol Res ; 141: 319-330, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30625359

RESUMEN

Eph/ephrin interactions and their bidirectional signaling are integral part of the complex communication system between ß-cells, essential for glucose homeostasis. Indeed, Eph/ephrin system was shown to be directly involved in the glucose-stimulated insulin secretion (GSIS) process occurring in the pancreatic islets. Here we tested the Eph antagonist UniPR500 as GSIS enhancer. UniPR500 was validated as EphA5-ephrin-A5 inhibitor in vitro and its efficacy as GSIS enhancer was assessed on EndoC-ßH1 cells. The selectivity of UniPR500 was evaluated by testing this compound on a panel of well-known molecular targets responsible for the regulation of glucose homeostasis. Plasmatic levels of UniPR500 were measured by HPLC/MS approach after oral administration. Finally, UniPR500 was tested as hypoglycemic agent in healthy mice, in a non-genetic mouse model of insulin resistance (IR) and in a non-genetic mouse model of type 1 diabetes (T1D). The compound is an orally bioavailable and selective Eph antagonist, able to increase GSIS from EndoC-ßH1 cells. When tested in vivo UniPR500 showed to improve glucose tolerance in healthy and IR mice. As expected by a GSIS enhancer acting on healthy ß-cells, UniPR500 was ineffective when tested on a non-genetic mouse model of type 1 diabetes, where pancreatic function was severely compromised. In conclusion our findings suggest that Eph targeting is a new and valuable pharmacological strategy in the search of new hypoglycemic agents.


Asunto(s)
Efrinas/metabolismo , Glucosa/metabolismo , Hipoglucemiantes/farmacología , Resistencia a la Insulina , Secreción de Insulina/efectos de los fármacos , Mapas de Interacción de Proteínas/efectos de los fármacos , Animales , Línea Celular , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Prueba de Tolerancia a la Glucosa , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL
5.
Sci Rep ; 5: 9322, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25818803

RESUMEN

Islet transplantation has been hampered by loss of function due to poor revascularization. We hypothesize that co-transplantation of islets with human embryonic stem cell-derived mesenchymal stromal cells that conditionally overexpress VEGF (hESC-MSC:VEGF) may augment islet revascularization and reduce the minimal islet mass required to reverse diabetes in mice. HESC-MSCs were transduced by recombinant lentiviruses that allowed conditional (Dox-regulated) overexpression of VEGF. HESC-MSC: VEGF were characterized by tube formation assay. After co-transplantation of hESC-MSC:VEGF with murine islets in collagen-fibrin hydrogel in the omental pouch of diabetic nude mice, we measured blood glucose, body weight, glucose tolerance and serum C-peptide. As control, islets were transplanted alone or with non-transduced hESC-MSCs. Next, we compared functional parameters of 400 islets alone versus 200 islets co-transplanted with hESC-MSC:VEGF. As control, 200 islets were transplanted alone. Metabolic function of islets transplanted with hESC-MSC:VEGF significantly improved, accompanied by superior graft revascularization, compared with control groups. Transplantation of 200 islets with hESC-MSC:VEGF showed superior function over 400 islets alone. We conclude that co-transplantation of islets with VEGF-expressing hESC-MSCs allowed for at least a 50% reduction in minimal islet mass required to reverse diabetes in mice. This approach may contribute to alleviate the need for multiple donor organs per patient.


Asunto(s)
Diabetes Mellitus/terapia , Células Madre Embrionarias Humanas/trasplante , Trasplante de Islotes Pancreáticos , Trasplante de Células Madre Mesenquimatosas , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patología , Expresión Génica , Humanos , Islotes Pancreáticos/patología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Factor A de Crecimiento Endotelial Vascular/biosíntesis
6.
Mol Psychiatry ; 20(8): 951-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25155879

RESUMEN

Gpr88, an orphan G-protein-coupled receptor, is highly and almost exclusively expressed in the medium spiny projection neurons of the striatum, and may thus participate in the control of motor functions and cognitive processing that are impaired in neuropsychiatric disorders such as Parkinson's disease or schizophrenia (SZ). This study investigated the relevance of Gpr88 to SZ-associated behavior by knocking down Gpr88 gene expression in the ventral striatum (nucleus accumbens) in a neurodevelopmental rat model of SZ, generated by neonatal treatment with phencyclidine (PCP). In this model, we compared the effects of the local inactivation in the adult animal of the expression of Gpr88 and of Drd2, a gene strongly implicated in the etiology of SZ and coding for the dopamine receptor type 2 (D2). To inactivate specifically Gpr88 and D2 expression, we used the lentiviral vector-mediated microRNA silencing strategy. The neonatal PCP treatment induced in the adult rat hyperlocomotion in response to amphetamine (Amph) and social novelty discrimination (SND) deficits. The inactivation of D2 did not modify the locomotor response to Amph or the cognitive deficits induced by PCP, whereas the silencing of Gpr88 inhibited the Amph-induced hyperlocomotion and reduced the impairment of SND elicited by neonatal exposure to PCP. These observations suggest a role for Gpr88 in the regulation of cognitive and motor functions, and support its relevance to the pathophysiology and treatment of SZ and other disorders involving dysfunction of the accumbens-striatal complex.


Asunto(s)
Núcleo Accumbens/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Esquizofrenia/metabolismo , Anfetamina/farmacología , Animales , Estimulantes del Sistema Nervioso Central/farmacología , Trastornos del Conocimiento/metabolismo , Discriminación en Psicología/efectos de los fármacos , Discriminación en Psicología/fisiología , Modelos Animales de Enfermedad , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Masculino , MicroARNs , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Núcleo Accumbens/efectos de los fármacos , Fenciclidina , Distribución Aleatoria , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Conducta Social
7.
Diabetologia ; 56(6): 1327-38, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23462796

RESUMEN

AIMS/HYPOTHESIS: It is not clear how small tyrosine kinase inhibitors, such as imatinib mesilate, protect against diabetes and beta cell death. The aim of this study was to determine whether imatinib, as compared with the non-cAbl-inhibitor sunitinib, affects pro-survival signalling events in the phosphatidylinositol 3-kinase (PI3K) pathway. METHODS: Human EndoC-ßH1 cells, murine beta TC-6 cells and human pancreatic islets were used for immunoblot analysis of insulin receptor substrate (IRS)-1, Akt and extracellular signal-regulated kinase (ERK) phosphorylation. Phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3] plasma membrane concentrations were assessed in EndoC-ßH1 and MIN6 cells using evanescent wave microscopy. Src homology 2-containing inositol 5'-phosphatase 2 (SHIP2) tyrosine phosphorylation and phosphatase and tensin homologue deleted on chromosome 10 (PTEN) serine phosphorylation, as well as c-Abl co-localisation with SHIP2, were studied in HEK293 and EndoC-ßH1 cells by immunoprecipitation and immunoblot analysis. Gene expression was assessed using RT-PCR. Cell viability was measured using vital staining. RESULTS: Imatinib stimulated ERK(thr202/tyr204) phosphorylation in a c-Abl-dependent manner. Imatinib, but not sunitinib, also stimulated IRS-1(tyr612), Akt(ser473) and Akt(thr308) phosphorylation. This effect was paralleled by oscillatory bursts in plasma membrane PI(3,4,5)P3 levels. Wortmannin induced a decrease in PI(3,4,5)P3 levels, which was slower in imatinib-treated cells than in control cells, indicating an effect on PI(3,4,5)P3-degrading enzymes. In line with this, imatinib decreased the phosphorylation of SHIP2 but not of PTEN. c-Abl co-immunoprecipitated with SHIP2 and its binding to SHIP2 was largely reduced by imatinib but not by sunitinib. Imatinib increased total ß-catenin levels and cell viability, whereas sunitinib exerted negative effects on cell viability. CONCLUSIONS/INTERPRETATION: Imatinib inhibition of c-Abl in beta cells decreases SHIP2 activity, which results in enhanced signalling downstream of PI3 kinase.


Asunto(s)
Benzamidas/farmacología , Supervivencia Celular/efectos de los fármacos , Insulina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Monoéster Fosfórico Hidrolasas/metabolismo , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-abl/metabolismo , Pirimidinas/farmacología , Transducción de Señal/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Células Cultivadas , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Mesilato de Imatinib , Indoles/farmacología , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Fosforilación , Unión Proteica , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Sunitinib , Factores de Tiempo
8.
Diabetologia ; 55(10): 2713-2722, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22760788

RESUMEN

AIMS/HYPOTHESIS: Pancreatic cell development is a tightly controlled process. Although information is available regarding the mesodermal signals that control pancreatic development, little is known about the role of environmental factors such as nutrients, including glucose, on pancreatic development. We previously showed that glucose and its metabolism through the hexosamine biosynthesis pathway (HBP) promote pancreatic endocrine cell differentiation. Here, we analysed the role of the transcription factor carbohydrate-responsive element-binding protein (ChREBP) in this process. This transcription factor is activated by glucose, and has been recently described as a target of the HBP. METHODS: We used an in vitro bioassay in which pancreatic endocrine and exocrine cells develop from rat embryonic pancreas in a way that mimics in vivo pancreatic development. Using this model, gain-of-function and loss-of-function experiments were undertaken. RESULTS: ChREBP was produced in the endocrine lineage during pancreatic development, its abundance increasing with differentiation. When rat embryonic pancreases were cultured in the presence of glucose or xylitol, the production of ChREBP targets was induced. Concomitantly, beta cell differentiation was enhanced. On the other hand, when embryonic pancreases were cultured with inhibitors decreasing ChREBP activity or an adenovirus producing a dominant-negative ChREBP, beta cell differentiation was reduced, indicating that ChREBP activity was necessary for proper beta cell differentiation. Interestingly, adenovirus producing a dominant-negative ChREBP also reduced the positive effect of N-acetylglucosamine, a substrate of the HBP acting on beta cell differentiation. CONCLUSIONS/INTERPRETATION: Our work supports the idea that glucose, through the transcription factor ChREBP, controls beta cell differentiation from pancreatic progenitors.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Diferenciación Celular/efectos de los fármacos , Glucosa/farmacología , Células Secretoras de Insulina/citología , Acetilglucosamina/farmacología , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Femenino , Técnicas In Vitro , Modelos Animales , Páncreas/citología , Páncreas/embriología , Páncreas/fisiología , Embarazo , Ratas , Ratas Wistar , Xilitol/farmacología
9.
Oncogene ; 28(27): 2535-44, 2009 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-19448668

RESUMEN

HIPK2 is a eukaryotic Serine-Threonine kinase that controls cellular proliferation and survival in response to exogenous signals. Here, we show that the human transcription factor ZBTB4 is a new target of HIPK2. The two proteins interact in vitro, colocalize and associate in vivo, and HIPK2 phosphorylates several conserved residues of ZBTB4. Overexpressing HIPK2 causes the degradation of ZBTB4, whereas overexpressing a kinase-deficient mutant of HIPK2 has no effect. The chemical activation of HIPK2 also decreases the amount of ZBTB4 in cells. Conversely, the inhibition of HIPK2 by drugs or by RNA interference causes a large increase in ZBTB4 levels. This negative regulation of ZBTB4 by HIPK2 occurs under normal conditions of cell growth. In addition, the degradation is increased by DNA damage. These findings have two consequences. First, we have recently shown that ZBTB4 inhibits the transcription of p21. Therefore, the activation of p21 by HIPK2 is two-pronged: stimulation of the activator p53, and simultaneous repression of the inhibitor ZBTB4. Second, ZBTB4 is also known to bind methylated DNA and repress methylated sequences. Consequently, our findings raise the possibility that HIPK2 might influence the epigenetic regulation of gene expression at loci that remain to be identified.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Proteínas Portadoras/genética , Línea Celular Tumoral , Daño del ADN , Regulación hacia Abajo , Células HCT116 , Humanos , Inmunoprecipitación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Datos de Secuencia Molecular , Mutación , Células 3T3 NIH , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Treonina/metabolismo , Técnicas del Sistema de Dos Híbridos
10.
Phys Rev E Stat Nonlin Soft Matter Phys ; 73(5 Pt 2): 056127, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16803018

RESUMEN

In psychiatric and neurological diseases, sleep is often perturbed. Moreover, recent works on humans and animals tend to show that sleep plays a strong role in memory processes. Reciprocally, sleep dynamics following a learning task is modified [Hubert, Nature (London) 02663, 1 (2004), Peigneux, Neuron 44, 535 (2004)]. However, sleep analysis in humans and animals is often limited to the total sleep and wake duration quantification. These two parameters are not fully able to characterize the sleep dynamics. In mammals sleep presents a complex organization with an alternation of slow wave sleep (SWS) and paradoxical sleep (PS) episodes. Moreover, it has been shown recently that these sleep episodes are frequently interrupted by micro-arousal (without awakening). We present here a detailed analysis of the basal sleep properties emerging from the mechanisms underlying the vigilance states alternation in an animal model. These properties present a self-organized critical system signature and reveal the existence of two W, two SWS, and a PS structure exhibiting a criticality as met in sand piles. We propose a theoretical model of the sleep dynamics based on several interacting neuronal populations. This new model of sleep dynamics presents the same properties as experimentally observed, and explains the variability of the collected data. This experimental and theoretical study suggests that sleep dynamics shares several common features with critical systems.

11.
Cell Death Differ ; 13(11): 1892-9, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16514419

RESUMEN

The basic helix-loop-helix protein Neurogenin3 specifies precursor cells of the endocrine pancreas during embryonic development, and is thought to be absent postnatally. We have studied Ngn3 expression during in vitro generation of beta-cells from adult rat exocrine pancreas tissue treated with epidermal growth factor and leukaemia inhibitory factor. This treatment induced a transient expression of both Ngn3 and its upstream activator hepatocyte nuclear factor 6. Inhibition of EGF and LIF signalling by pharmacological antagonists of the JAK2/STAT3 pathway, or knockdown of Ngn3 by RNA interference prevented the generation of new insulin-positive cells. This study demonstrates that in vitro growth factor stimulation can induce recapitulation of an embryonic endocrine differentiation pathway in adult dedifferentiated exocrine cells. This could prove to be important for understanding the mechanism of beta-cell regeneration and for therapeutic ex vivo neogenesis of beta cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Janus Quinasa 2/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Cultivadas , Perfilación de la Expresión Génica , Silenciador del Gen , Sustancias de Crecimiento/farmacología , Factor Nuclear 6 del Hepatocito/genética , Humanos , Células Secretoras de Insulina/efectos de los fármacos , Lipasa/metabolismo , Masculino , Modelos Animales , Proteínas del Tejido Nervioso/genética , Niacinamida/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
12.
Diabetologia ; 48(4): 709-19, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15759109

RESUMEN

AIMS/HYPOTHESIS: Gene transfer into pancreatic beta cells, which produce and secrete insulin, is a promising strategy to protect such cells against autoimmune destruction and also to generate beta cells in mass, thereby providing a novel therapeutic approach to treat diabetic patients. Until recently, exogenous DNA has been directly transferred into mature beta cells with various levels of success. We investigated whether exogenous DNA could be stably transferred into pancreatic stem/progenitor cells, which would subsequently differentiate into mature beta cells expressing the transgene. METHODS: We designed transplantation and tissue culture procedures to obtain ex vivo models of pancreatic development. We next constructed recombinant lentiviruses expressing enhanced green fluorescent protein (eGFP) under the control of either the rat insulin promoter or a ubiquitous promoter, and performed viral infection of rat embryonic pancreatic tissue. RESULTS: Embryonic pancreas infected with recombinant lentiviruses resulted in endocrine cell differentiation and restricted cell type expression of the transgene according to the specificity of the promoter used in the viral construct. We next demonstrated that the efficiency of infection could be further improved upon infection of embryonic pancreatic epithelia, followed by their in vitro culture, using conditions that favour endocrine cell differentiation. Under these conditions, endocrine stem/progenitor cells expressing neurogenin 3 are efficiently transduced by recombinant lentiviral vectors. Moreover, when eGFP was placed under the control of the insulin promoter, 70.4% of the developed beta cells were eGFP-expressing cells. All of the eGFP-positive cells were insulin-producing cells. CONCLUSIONS/INTERPRETATION: We have demonstrated that mature rat pancreatic beta cells can be stably modified by infecting pancreatic stem/progenitor cells that undergo endocrine differentiation.


Asunto(s)
Expresión Génica/genética , Islotes Pancreáticos/metabolismo , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Carboxipeptidasas A/metabolismo , Diferenciación Celular/fisiología , Citomegalovirus/genética , Vectores Genéticos/genética , Glucagón/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hibridación Fluorescente in Situ , Insulina/genética , Insulina/metabolismo , Lentivirus/genética , Masculino , Ratones , Ratones SCID , Proteínas del Tejido Nervioso/genética , Páncreas/citología , Páncreas/embriología , Regiones Promotoras Genéticas/genética , Ratas , Ratas Wistar , Células Madre/citología , Transfección/métodos , Trasplante Heterólogo , Trasplantes
13.
Neuroscience ; 123(4): 983-1002, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14751290

RESUMEN

We have recently identified a third subtype of glutamate vesicular transporter (VGLUT) named VGLUT3. In the present study, we provide a detailed account of the regional and cellular distributions of VGLUT3 in the rat brain, using specific nucleotide probes and antisera. The distribution of VGLUT3 protein was compared with that of the other vesicular transporters (VGLUT1 and VGLUT2). All the areas expressing VGLUT3 also contain high levels of VGLUT1 and -2 proteins, but, at a finer level of analysis, the distribution of the three subtypes differs. Unlike VGLUT1 and -2, VGLUT3 expression is limited to discrete cell populations. Neurons containing VGLUT3 transcript are essentially observed in the caudate-putamen, the olfactory tubercle, the nucleus accumbens, the hippocampus, the interpeduncular nucleus and the dorsal and medial raphe nuclei. More scattered populations of VGLUT3 expressing neurons are found in the cerebral cortex. The distribution of the VGLUT3 protein, as determined with specific antisera, overlaps with that of the transcript in the caudate-putamen, olfactory tubercles, hippocampus, cortex, interpeduncular nucleus, and raphe nuclei, suggesting that VGLUT3 is essentially present in local projection neurons in these regions. Microscopic examination reveals staining of terminals and perikarya. Furthermore, co-localization studies indicate that VGLUT3 is present in GABAergic interneurons in the hippocampus, as well as in the interpeduncular nucleus. However, other regions, such as the substantia nigra (pars compacta), the ventral tegmental area, and the parabigeminal nucleus, receive a dense VGLUT3 terminal labeling although they do not contain VGLUT3 expressing neurons. In these regions, VGLUT3 immunoreactivity may be present in terminals of long projecting neurons. This subclass of glutamatergic afferents differs from other "classical" excitatory terminals that express VGLUT1 or VGLUT2. The distribution of VGLUT3 in the rat brain suggests an unsuspected function of vesicular glutamate transport in subsets of interneurons and in neuromodulatory neurons.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Encéfalo/metabolismo , Proteínas de Transporte de Membrana , Neuronas/metabolismo , Proteínas de Transporte Vesicular , Sistemas de Transporte de Aminoácidos Acídicos/genética , Animales , Autorradiografía/métodos , Encéfalo/citología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Glutamato Descarboxilasa/genética , Glutamato Descarboxilasa/metabolismo , Inmunohistoquímica/métodos , Hibridación in Situ/métodos , Masculino , Ratas , Ratas Sprague-Dawley , Proteína 1 de Transporte Vesicular de Glutamato , Proteína 2 de Transporte Vesicular de Glutamato , Proteínas de Transporte Vesicular de Glutamato
14.
J Neurosci ; 21(22): RC181, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11698619

RESUMEN

Before their exocytotic release during stimulation of nerve terminals, nonpeptide neurotransmitters are loaded into synaptic vesicles by specific transporters. Recently, a protein initially identified as brain-specific Na(+)-dependent inorganic phosphate transporter I (BNPI) has been shown to represent a vesicular glutamate transporter (VGLUT1). In this study, we investigated whether a highly homologous "differentiation-associated Na(+)-dependent inorganic phosphate transporter" (DNPI) is involved in glutamatergic transmission. Vesicles isolated from BON cells expressing recombinant DNPI accumulated l-glutamate with bioenergetical and pharmacological characteristics identical to those displayed by VGLUT1 and by brain synaptic vesicles. Moreover, DNPI localized to synaptic vesicles, at synapses exhibiting classical excitatory features. DNPI thus represents a novel vesicular glutamate transporter (VGLUT2). The distributions of each VGLUT transcript in brain were highly complementary, with only a partial regional and cellular overlap. At the protein level, we could only detect either VGLUT1- or VGLUT2-expressing presynaptic boutons. The existence of two VGLUTs thus defines distinct subsets of glutamatergic neurons.


Asunto(s)
Sistemas de Transporte de Aminoácidos , Proteínas Portadoras/metabolismo , Ácido Glutámico/metabolismo , Proteínas de Transporte de Membrana , Neuronas/metabolismo , Proteínas de Transporte de Fosfato/metabolismo , Proteínas de Transporte Vesicular , Animales , Transporte Biológico , Encéfalo/citología , Encéfalo/metabolismo , Proteínas Portadoras/genética , Diferenciación Celular , Línea Celular , Hibridación in Situ , Neuronas/clasificación , Especificidad de Órganos , Terminales Presinápticos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Vesículas Sinápticas/metabolismo , Transfección , Proteína 1 de Transporte Vesicular de Glutamato , Proteína 2 de Transporte Vesicular de Glutamato , Proteínas del Transporte Vesicular de Aminoácidos Inhibidores
15.
J Neurosci Res ; 65(2): 91-9, 2001 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-11438978

RESUMEN

Reserpine treatment leads to a trans-synaptic increase of the tyrosine hydroxylase (TH) gene transcription rate, mRNA and protein levels in catecholaminergic tissues including the adrenal medulla (AM) and the superior cervical ganglia (SCG). The TPA-responsive element plays an important role in the trans-synaptically-induced transcription of the TH gene in the AM, whereas it does not appear to be involved in the SCG (Trocmé et al. [1997] J. Neurosci. Res. 48:489-498). In this study, we show that another regulatory sequence of the TH proximal promoter, the cAMP-responsive element (CRE), binds different factors in the AM and in the SCG. To elucidate the dynamics of promoter regulation a complete time course analysis was conducted. Reserpine treatment enhances, between 1 hr and 8 hr after the injection, the expression and the binding of the repressor ICER in the AM, whereas in the SCG it enhances the binding of CREM factors. These results suggest that the mechanisms mediating trans-synaptic induction of the TH gene are different in the AM and SCG. The interplay between positive and negative transcription factors and their kinetics of action are responsive of the long-term regulation of the TH gene.


Asunto(s)
Médula Suprarrenal/enzimología , Proteínas de Unión al ADN/metabolismo , Ganglios Simpáticos/enzimología , Regulación Enzimológica de la Expresión Génica/fisiología , Neuronas/enzimología , Proteínas Represoras , Sinapsis/enzimología , Tirosina 3-Monooxigenasa/genética , Médula Suprarrenal/citología , Médula Suprarrenal/efectos de los fármacos , Inhibidores de Captación Adrenérgica/farmacología , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Modulador del Elemento de Respuesta al AMP Cíclico , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Ganglios Simpáticos/citología , Ganglios Simpáticos/efectos de los fármacos , Inmunohistoquímica , Masculino , Neuronas/citología , Neuronas/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , Isoformas de Proteínas/efectos de los fármacos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Reserpina/farmacología , Sinapsis/efectos de los fármacos , Sinapsis/ultraestructura , Transcripción Genética/efectos de los fármacos , Transcripción Genética/fisiología , Tirosina 3-Monooxigenasa/metabolismo
16.
Proc Natl Acad Sci U S A ; 98(13): 7206-11, 2001 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-11390972

RESUMEN

In eukaryotic cells, lysosomes represent a major site for macromolecule degradation. Hydrolysis products are eventually exported from this acidic organelle into the cytosol through specific transporters. Impairment of this process at either the hydrolysis or the efflux step is responsible of several lysosomal storage diseases. However, most lysosomal transporters, although biochemically characterized, remain unknown at the molecular level. In this study, we report the molecular and functional characterization of a lysosomal amino acid transporter (LYAAT-1), remotely related to a family of H+-coupled plasma membrane and synaptic vesicle amino acid transporters. LYAAT-1 is expressed in most rat tissues, with highest levels in the brain where it is present in neurons. Upon overexpression in COS-7 cells, the recombinant protein mediates the accumulation of neutral amino acids, such as gamma-aminobutyric acid, l-alanine, and l-proline, through an H+/amino acid symport. Confocal microscopy on brain sections revealed that this transporter colocalizes with cathepsin D, an established lysosomal marker. LYAAT-1 thus appears as a lysosomal transporter that actively exports neutral amino acids from lysosomes by chemiosmotic coupling to the H+-ATPase of these organelles. Homology searching in eukaryotic genomes suggests that LYAAT-1 defines a subgroup of lysosomal transporters in the amino acid/auxin permease family.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Sistemas de Transporte de Aminoácidos , Aminoácidos Neutros/metabolismo , Encéfalo/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Lisosomas/metabolismo , Alanina/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans , Proteínas Portadoras/química , Línea Celular , Corteza Cerebral/metabolismo , Clonación Molecular , Drosophila melanogaster , Evolución Molecular , Hipocampo/metabolismo , Cinética , Masculino , Datos de Secuencia Molecular , Filogenia , Prolina/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Simportadores , Transfección , Ácido gamma-Aminobutírico/metabolismo
17.
Genomics ; 62(1): 82-5, 1999 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-10585771

RESUMEN

DiGeorge syndrome (DGS) is a developmental disorder associated with large hemizygous deletions on chromosome 22q11.2. ZNF74 zinc finger gene is a candidate from the commonly deleted region. To address the potential involvement of ZNF74 in DGS, its human developmental expression pattern has been assessed. In situ hybridization on Carnegie Stage 18 embryos revealed that ZNF74 expression is limited to specific neural crest-derived tissues and neuroepithelium of the spinal cord as well as to foregut endoderm epithelia (esophagus and respiratory tract). Interestingly, ZNF74 expression was detected in the wall of the pulmonary artery and aorta and in the aortic valve, which are populated by neural crest-derived cells. This finding is significant, considering that DGS is believed to result from defective neural crest contributions and that outflow tract and aorticopulmonary septation defects are typical features of the DGS phenotype. Thus, the restricted expression of ZNF74 in structures affected in DGS suggests a role for this putative regulator of gene expression in aspects of the DGS phenotype.


Asunto(s)
Cromosomas Humanos Par 22/genética , Síndrome de DiGeorge/genética , Endodermo/metabolismo , Proteínas Fetales/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/metabolismo , Proteínas de Unión al ARN/genética , Aorta/embriología , Aorta/metabolismo , Válvula Aórtica/embriología , Válvula Aórtica/metabolismo , Linaje de la Célula , Síndrome de DiGeorge/patología , Desarrollo Embrionario y Fetal/genética , Células Epiteliales/metabolismo , Proteínas Fetales/biosíntesis , Genes , Humanos , Factores de Transcripción de Tipo Kruppel , Datos de Secuencia Molecular , Especificidad de Órganos , Fenotipo , Arteria Pulmonar/embriología , Arteria Pulmonar/metabolismo , Proteínas de Unión al ARN/biosíntesis , Dedos de Zinc/genética
18.
J Biol Chem ; 274(6): 3469-75, 1999 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-9920892

RESUMEN

A novel human tyrosine hydroxylase (HTH) messenger RNA subgroup generated by alternative splicing and characterized by the absence of the third exon was recently identified. The corresponding putative protein lacks 74 amino acids including Ser31 and Ser40, two major phosphorylation sites implicated in the regulation of HTH activity. These mRNA species are detected in adrenal medulla and are overexpressed in patients suffering from progressive supranuclear palsy, a neurodegenerative disease mostly affecting catecholaminergic neurons of the basal ganglia. In the present work, an HTH protein isoform lacking exon 3 was identified in human adrenal medulla. For this purpose, an antibody was raised against the HTH exon 3. The effect of the removal of exon 3 on the enzymatic activity of HTH was studied in vitro by comparing a purified recombinant fusion protein without exon 3 (glutathione S-transferase (GST)-HTHDelta3) to the equivalent protein containing exon 3 (GST-HTH3). In initial velocity conditions, GST-HTHDelta3 has 30% of the maximal velocity of GST-HTH3. Moreover, the skipping of exon 3 results in the absence of activation of GST-HTH by heparin and increases by 10-fold the retroinhibition constant for dopamine, demonstrating the involvement of exon 3 in the regulation of HTH enzymatic activity. The identification of a variably expressed HTH isoform that lacks an exon implicated in activity regulation supports the view that HTH alternative splicing contributes to the functional diversity within the catecholaminergic system and may be implicated in some neurological diseases.


Asunto(s)
Isoenzimas/metabolismo , Parálisis Supranuclear Progresiva/enzimología , Tirosina 3-Monooxigenasa/metabolismo , Glándulas Suprarrenales/enzimología , Secuencia de Bases , Cartilla de ADN , Dopamina/fisiología , Exones , Regulación de la Expresión Génica/fisiología , Glutatión Transferasa/genética , Heparina/fisiología , Humanos , Isoenzimas/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tirosina 3-Monooxigenasa/genética
19.
J Biol Chem ; 273(17): 10196-201, 1998 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-9553069

RESUMEN

Human tyrosine hydroxylase exists as four isoforms (hTH1-4), generated by alternative splicing of pre-mRNA, with tissue-specific distribution. Unphosphorylated hTH3 and hTH1 were produced in large amounts in Escherichia coli and purified to homogeneity. The phosphorylation sites were determined after labeling with [32P]phosphate in the presence of cAMP-dependent protein kinase (PKA) and calmodulin-dependent protein kinase II (CaM-PKII). Ser40 was phosphorylated by PKA, and both Ser19 and Ser40 were phosphorylated by CaM-PKII. The enzyme kinetics of hTH3 were determined in the presence of various concentrations of the natural co-substrate (6R)-tetrahydrobiopterin and compared with those of recombinant hTH1 (similar to rat TH). We show that, under initial velocity conditions, excess (6R)-tetrahydrobiopterin inhibits hTH3 and hTH1. The TH catalytic constants (kcat) were determined for each of the two isoenzymes: hTH3 is about five times more active than hTH1. Phosphorylation by CaM-PKII did not affect the kinetic parameters of hTH3. The classical activation of TH by PKA phosphorylation, demonstrated for hTH1, was not observed with hTH3. Furthermore, hTH3 escapes activity regulation by phosphorylation and is always more active than phosphorylated hTH1. The properties of the hTH3 enzyme may be relevant to diseases affecting dopaminergic cells.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Isoenzimas/antagonistas & inhibidores , Pterinas/farmacología , Tirosina 3-Monooxigenasa/antagonistas & inhibidores , Humanos , Isoenzimas/aislamiento & purificación , Isoenzimas/metabolismo , Fosforilación , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Tirosina 3-Monooxigenasa/aislamiento & purificación , Tirosina 3-Monooxigenasa/metabolismo
20.
Hum Mol Genet ; 7(3): 423-8, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9466999

RESUMEN

The polymorphic HUMTH01 microsatellite, located in the first intron of the tyrosine hydroxylase gene is characterized by a tetranucleotide core motif. The 10 repeat allele of this microsatellite exhibits two sequence variants: an imperfect repeat and a perfect repeat. Here we present evidence that this tetrarepeat is endowed with regulatory properties. Constructions were made linking the 10 repetition alleles to the luciferase reporter gene under the control of a thymidine kinase minimal promoter. In transient transfection experiments in HeLa, PC12 and SK-NSH cell lines these repeated sequences increased the basal transcription up to 9-fold. This effect was independent of the sequence orientation, a feature characteristic of an enhancer element. In electrophoretic mobility shift assays these tetrameric repeated sequences form specific complexes with HeLa cell nuclear extracts. Competition experiments with heterologous sequences suggest that proteins of the Fos-Jun family may be involved in the formation of these complexes, although other unidentified transacting factors bind to these sequences. These results thus implicate the HUMTH01 microsatellite in the regulation of tyrosine hydroxylase gene expression. Tetrarepeated sequences of this type may constitute a new class of regulatory elements.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Repeticiones de Microsatélite , Polimorfismo Genético , Transcripción Genética , Tirosina 3-Monooxigenasa/biosíntesis , Tirosina 3-Monooxigenasa/genética , Alelos , Animales , Núcleo Celular/metabolismo , Células HeLa , Humanos , Intrones , Luciferasas/biosíntesis , Células PC12 , Ratas , Proteínas Recombinantes de Fusión/biosíntesis , Secuencias Reguladoras de Ácidos Nucleicos , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...