Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Cell Transplant ; 29: 963689720971204, 2020.
Article in English | MEDLINE | ID: mdl-33150791

ABSTRACT

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.


Subject(s)
Insulin-Secreting Cells/metabolism , Pancreas/enzymology , Pancreas/metabolism , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Dogs , Female , Insulin/metabolism , Insulinoma/metabolism , Integrases/genetics , Integrases/metabolism , Mice, SCID , Pregnancy , Promoter Regions, Genetic/genetics , RNA, Messenger/metabolism
2.
Cell Transplant ; 28(12): 1641-1651, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31450972

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental , Fetus/metabolism , Gene Expression Regulation , Insulin/biosynthesis , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Organoids , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/therapy , Dogs , Fetus/pathology , Heterografts , Islets of Langerhans/pathology , Mice , Mice, SCID , Organoids/metabolism , Organoids/pathology , Organoids/transplantation
3.
Sci Rep ; 9(1): 1409, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30723217

ABSTRACT

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.


Subject(s)
Chromatin/genetics , Dopaminergic Neurons/metabolism , Mesencephalon/cytology , RNA, Long Noncoding/genetics , Repetitive Sequences, Nucleic Acid/genetics , Animals , Chromatin Immunoprecipitation Sequencing , Female , Male , Mesencephalon/embryology , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA-Seq , Serotonergic Neurons/metabolism , Transcriptome
4.
Pharmacol Res ; 141: 319-330, 2019 03.
Article in English | MEDLINE | ID: mdl-30625359

ABSTRACT

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.


Subject(s)
Ephrins/metabolism , Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin Resistance , Insulin Secretion/drug effects , Protein Interaction Maps/drug effects , Animals , Cell Line , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/metabolism , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice, Inbred C57BL
5.
Sci Rep ; 5: 9322, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25818803

ABSTRACT

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.


Subject(s)
Diabetes Mellitus/therapy , Human Embryonic Stem Cells/transplantation , Islets of Langerhans Transplantation , Mesenchymal Stem Cell Transplantation , Vascular Endothelial Growth Factor A/genetics , Animals , Diabetes Mellitus/metabolism , Diabetes Mellitus/pathology , Gene Expression , Humans , Islets of Langerhans/pathology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Vascular Endothelial Growth Factor A/biosynthesis
6.
Mol Psychiatry ; 20(8): 951-8, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25155879

ABSTRACT

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.


Subject(s)
Nucleus Accumbens/metabolism , Receptors, G-Protein-Coupled/deficiency , Schizophrenia/metabolism , Amphetamine/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Cognition Disorders/metabolism , Discrimination, Psychological/drug effects , Discrimination, Psychological/physiology , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Knockdown Techniques , Gene Silencing , Male , MicroRNAs , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/drug effects , Phencyclidine , Random Allocation , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Social Behavior
7.
Diabetologia ; 56(6): 1327-38, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23462796

ABSTRACT

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.


Subject(s)
Benzamides/pharmacology , Cell Survival/drug effects , Insulin/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/metabolism , Piperazines/pharmacology , Proto-Oncogene Proteins c-abl/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects , Cell Membrane/drug effects , Cells, Cultured , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Imatinib Mesylate , Indoles/pharmacology , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Sunitinib , Time Factors
8.
Diabetologia ; 55(10): 2713-2722, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22760788

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Cell Differentiation/drug effects , Glucose/pharmacology , Insulin-Secreting Cells/cytology , Acetylglucosamine/pharmacology , Animals , Cell Differentiation/physiology , Cells, Cultured , Female , In Vitro Techniques , Models, Animal , Pancreas/cytology , Pancreas/embryology , Pancreas/physiology , Pregnancy , Rats , Rats, Wistar , Xylitol/pharmacology
9.
Oncogene ; 28(27): 2535-44, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19448668

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Repressor Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Carrier Proteins/genetics , Cell Line, Tumor , DNA Damage , Down-Regulation , HCT116 Cells , Humans , Immunoprecipitation , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Molecular Sequence Data , Mutation , NIH 3T3 Cells , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/genetics , RNA Interference , Repressor Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Threonine/metabolism , Two-Hybrid System Techniques
10.
Phys Rev E Stat Nonlin Soft Matter Phys ; 73(5 Pt 2): 056127, 2006 May.
Article in English | MEDLINE | ID: mdl-16803018

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-16514419

ABSTRACT

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.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Janus Kinase 2/metabolism , Nerve Tissue Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Gene Expression Profiling , Gene Silencing , Growth Substances/pharmacology , Hepatocyte Nuclear Factor 6/genetics , Humans , Insulin-Secreting Cells/drug effects , Lipase/metabolism , Male , Models, Animal , Nerve Tissue Proteins/genetics , Niacinamide/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Up-Regulation/drug effects
12.
Diabetologia ; 48(4): 709-19, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15759109

ABSTRACT

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.


Subject(s)
Gene Expression/genetics , Islets of Langerhans/metabolism , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors , Carboxypeptidases A/metabolism , Cell Differentiation/physiology , Cytomegalovirus/genetics , Genetic Vectors/genetics , Glucagon/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Situ Hybridization, Fluorescence , Insulin/genetics , Insulin/metabolism , Lentivirus/genetics , Male , Mice , Mice, SCID , Nerve Tissue Proteins/genetics , Pancreas/cytology , Pancreas/embryology , Promoter Regions, Genetic/genetics , Rats , Rats, Wistar , Stem Cells/cytology , Transfection/methods , Transplantation, Heterologous , Transplants
13.
Neuroscience ; 123(4): 983-1002, 2004.
Article in English | MEDLINE | ID: mdl-14751290

ABSTRACT

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.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Brain/metabolism , Membrane Transport Proteins , Neurons/metabolism , Vesicular Transport Proteins , Amino Acid Transport Systems, Acidic/genetics , Animals , Autoradiography/methods , Brain/cytology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Glutamate Decarboxylase/genetics , Glutamate Decarboxylase/metabolism , Immunohistochemistry/methods , In Situ Hybridization/methods , Male , Rats , Rats, Sprague-Dawley , Vesicular Glutamate Transport Protein 1 , Vesicular Glutamate Transport Protein 2 , Vesicular Glutamate Transport Proteins
14.
J Neurosci ; 21(22): RC181, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698619

ABSTRACT

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.


Subject(s)
Amino Acid Transport Systems , Carrier Proteins/metabolism , Glutamic Acid/metabolism , Membrane Transport Proteins , Neurons/metabolism , Phosphate Transport Proteins/metabolism , Vesicular Transport Proteins , Animals , Biological Transport , Brain/cytology , Brain/metabolism , Carrier Proteins/genetics , Cell Differentiation , Cell Line , In Situ Hybridization , Neurons/classification , Organ Specificity , Presynaptic Terminals/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Synaptic Vesicles/metabolism , Transfection , Vesicular Glutamate Transport Protein 1 , Vesicular Glutamate Transport Protein 2 , Vesicular Inhibitory Amino Acid Transport Proteins
15.
J Neurosci Res ; 65(2): 91-9, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11438978

ABSTRACT

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.


Subject(s)
Adrenal Medulla/enzymology , DNA-Binding Proteins/metabolism , Ganglia, Sympathetic/enzymology , Gene Expression Regulation, Enzymologic/physiology , Neurons/enzymology , Repressor Proteins , Synapses/enzymology , Tyrosine 3-Monooxygenase/genetics , Adrenal Medulla/cytology , Adrenal Medulla/drug effects , Adrenergic Uptake Inhibitors/pharmacology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Cyclic AMP Response Element Modulator , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/genetics , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Immunohistochemistry , Male , Neurons/cytology , Neurons/drug effects , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/physiology , Protein Isoforms/drug effects , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology , Synapses/drug effects , Synapses/ultrastructure , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Tyrosine 3-Monooxygenase/metabolism
16.
Proc Natl Acad Sci U S A ; 98(13): 7206-11, 2001 Jun 19.
Article in English | MEDLINE | ID: mdl-11390972

ABSTRACT

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.


Subject(s)
Amino Acid Transport Systems, Neutral , Amino Acid Transport Systems , Amino Acids, Neutral/metabolism , Brain/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Lysosomes/metabolism , Alanine/metabolism , Amino Acid Sequence , Animals , Caenorhabditis elegans , Carrier Proteins/chemistry , Cell Line , Cerebral Cortex/metabolism , Cloning, Molecular , Drosophila melanogaster , Evolution, Molecular , Hippocampus/metabolism , Kinetics , Male , Molecular Sequence Data , Phylogeny , Proline/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Sequence Alignment , Sequence Homology, Amino Acid , Symporters , Transfection , gamma-Aminobutyric Acid/metabolism
17.
Genomics ; 62(1): 82-5, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10585771

ABSTRACT

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.


Subject(s)
Chromosomes, Human, Pair 22/genetics , DiGeorge Syndrome/genetics , Endoderm/metabolism , Fetal Proteins/genetics , Gene Deletion , Gene Expression Regulation, Developmental , Neural Crest/metabolism , RNA-Binding Proteins/genetics , Aorta/embryology , Aorta/metabolism , Aortic Valve/embryology , Aortic Valve/metabolism , Cell Lineage , DiGeorge Syndrome/pathology , Embryonic and Fetal Development/genetics , Epithelial Cells/metabolism , Fetal Proteins/biosynthesis , Genes , Humans , Kruppel-Like Transcription Factors , Molecular Sequence Data , Organ Specificity , Phenotype , Pulmonary Artery/embryology , Pulmonary Artery/metabolism , RNA-Binding Proteins/biosynthesis , Zinc Fingers/genetics
18.
J Biol Chem ; 274(6): 3469-75, 1999 Feb 05.
Article in English | MEDLINE | ID: mdl-9920892

ABSTRACT

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.


Subject(s)
Isoenzymes/metabolism , Supranuclear Palsy, Progressive/enzymology , Tyrosine 3-Monooxygenase/metabolism , Adrenal Glands/enzymology , Base Sequence , DNA Primers , Dopamine/physiology , Exons , Gene Expression Regulation/physiology , Glutathione Transferase/genetics , Heparin/physiology , Humans , Isoenzymes/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Tyrosine 3-Monooxygenase/genetics
19.
J Biol Chem ; 273(17): 10196-201, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9553069

ABSTRACT

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.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Isoenzymes/antagonists & inhibitors , Pterins/pharmacology , Tyrosine 3-Monooxygenase/antagonists & inhibitors , Humans , Isoenzymes/isolation & purification , Isoenzymes/metabolism , Phosphorylation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Substrate Specificity , Tyrosine 3-Monooxygenase/isolation & purification , Tyrosine 3-Monooxygenase/metabolism
20.
Hum Mol Genet ; 7(3): 423-8, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9466999

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Enzymologic , Microsatellite Repeats , Polymorphism, Genetic , Transcription, Genetic , Tyrosine 3-Monooxygenase/biosynthesis , Tyrosine 3-Monooxygenase/genetics , Alleles , Animals , Cell Nucleus/metabolism , HeLa Cells , Humans , Introns , Luciferases/biosynthesis , PC12 Cells , Rats , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Nucleic Acid , Transfection , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL