267 results on '"Ravassard P"'
Search Results
52. Ngn3 expression during postnatal in vitro beta cell neogenesis induced by the JAK/STAT pathway
- Author
-
Baeyens, L, primary, Bonné, S, additional, German, M S, additional, Ravassard, P, additional, Heimberg, H, additional, and Bouwens, L, additional
- Published
- 2006
- Full Text
- View/download PDF
53. Localization of VGLUT3, the vesicular glutamate transporter type 3, in the rat brain
- Author
-
Herzog, E, primary, Gilchrist, J, additional, Gras, C, additional, Muzerelle, A, additional, Ravassard, P, additional, Giros, B, additional, Gaspar, P, additional, and El Mestikawy, S, additional
- Published
- 2004
- Full Text
- View/download PDF
54. cDNA Libraries from a Low Amount of Cells.
- Author
-
Walker, John M., Bartlett, John M. S., Stirling, David, Ravassard, Philippe, Icard-Liepkalns, Christine, Mallet, Jacques, and Edwards, Jean Baptiste Dumas Milne
- Abstract
Conventional cDNA library construction often requires a minimum available amount of material (typically 1 or 2 μg of polyA+ RNA). For complex organs, such as brain, or certain species, such as humans, as well as subsets of cell types, this condition is often difficult to fulfill. Amplification by polymerase chain reaction (PCR) can be used to circumvent this limitation because it is a powerful method to obtain working quantities of low-abundance DNAs. To effectively apply this method, known sequences need to be attached to the ends of the single-stranded cDNA (ss-cDNA). One at the 5′ end of the ss-cDNA is added during the priming of the synthesis; the other, at the 3′ end, is covalently attached by ligation using the SLIC strategy. With known DNA sequences attached to both ends of the synthetized cDNA, minute quantities can be amplified with sequence-specific primers to provide sufficient material to successfully generate and screen cDNA libraries. The overall scheme is illustrated in Fig. 1. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
55. Méthodologie d’étude de la dégradation des outillages de frappe à froid
- Author
-
Verleene, A., primary, Dubar, L., additional, Dubois, A., additional, Ravassard, P., additional, and Oudin, J., additional
- Published
- 1999
- Full Text
- View/download PDF
56. Three selection criteria for the cold metal forming lubricating oils containing extreme pressure agents
- Author
-
Lazzarotto, L, primary, Dubar, L, additional, Dubois, A, additional, Ravassard, P, additional, and Oudin, J, additional
- Published
- 1998
- Full Text
- View/download PDF
57. A selection methodology for lubricating oils in cold metal forming processes
- Author
-
Lazzarotto, L., primary, Dubar, L., additional, Dubois, A., additional, Ravassard, P., additional, Bricout, J.P., additional, and Oudin, J., additional
- Published
- 1998
- Full Text
- View/download PDF
58. A rare allele of a tetranucleotide polymorphic microsatellite, located in the first intron of the tyrosine hydroxylase gene, is associated with schizophrenia and shows transcription regulatory activity in vitro
- Author
-
Meloni, R., primary, Albanese, V., additional, Ravassard, P., additional, Treilhou, F., additional, and Mallet, J., additional
- Published
- 1998
- Full Text
- View/download PDF
59. Identification of Coulomb's friction coefficient in real contact conditions applied to a wire drawing process
- Author
-
Lazzarotto, L., primary, Dubar, L., additional, Dubois, A., additional, Ravassard, P., additional, and Oudin, J., additional
- Published
- 1997
- Full Text
- View/download PDF
60. Detection of two new polymorphic sites in the human interleukin-1β gene
- Author
-
Laurent, C., primary, Thibaut, F., additional, Ravassard, P., additional, Campion, D., additional, Samolyk, D., additional, Lafargue, C., additional, Petit, M., additional, Martinez, M., additional, and Mallet, J., additional
- Published
- 1997
- Full Text
- View/download PDF
61. Tyrosine hydroxylase regulation in neurotransmission and neuroplasticity
- Author
-
Icard-Liepkalns, C., primary, Berrard, S., additional, Biguet, N.Faucon, additional, Lebourdelles, B., additional, Ravassard, P., additional, Robert, JJ, additional, and Mallet, J, additional
- Published
- 1993
- Full Text
- View/download PDF
62. Specific control of pancreatic endocrine β- and δ-cell mass by class IIa histone deacetylases HDAC4, HDAC5, and HDAC9.
- Author
-
Lenoir O, Flosseau K, Ma FX, Blondeau B, Mai A, Bassel-Duby R, Ravassard P, Olson EN, Haumaitre C, Scharfmann R, Lenoir, Olivia, Flosseau, Kathleen, Ma, Feng Xia, Blondeau, Bertrand, Mai, Antonello, Bassel-Duby, Rhonda, Ravassard, Philippe, Olson, Eric N, Haumaitre, Cécile, and Scharfmann, Raphaël
- Abstract
Objective: Class IIa histone deacetylases (HDACs) belong to a large family of enzymes involved in protein deacetylation and play a role in regulating gene expression and cell differentiation. Previously, we showed that HDAC inhibitors modify the timing and determination of pancreatic cell fate. The aim of this study was to determine the role of class IIa HDACs in pancreas development.Research Design and Methods: We took a genetic approach and analyzed the pancreatic phenotype of mice lacking HDAC4, -5, and -9. We also developed a novel method of lentiviral infection of pancreatic explants and performed gain-of-function experiments.Results: We show that class IIa HDAC4, -5, and -9 have an unexpected restricted expression in the endocrine β- and δ-cells of the pancreas. Analyses of the pancreas of class IIa HDAC mutant mice revealed an increased pool of insulin-producing β-cells in Hdac5(-/-) and Hdac9(-/-) mice and an increased pool of somatostatin-producing δ-cells in Hdac4(-/-) and Hdac5(-/-) mice. Conversely, HDAC4 and HDAC5 overexpression showed a decreased pool of insulin-producing β-cells and somatostatin-producing δ-cells. Finally, treatment of pancreatic explants with the selective class IIa HDAC inhibitor MC1568 enhances expression of Pax4, a key factor required for proper β-and δ-cell differentiation and amplifies endocrine β- and δ-cells.Conclusions: We conclude that HDAC4, -5, and -9 are key regulators to control the pancreatic β/δ-cell lineage. These results highlight the epigenetic mechanisms underlying the regulation of endocrine cell development and suggest new strategies for β-cell differentiation-based therapies. [ABSTRACT FROM AUTHOR]- Published
- 2011
- Full Text
- View/download PDF
63. Paradoxical (REM) sleep deprivation causes a large and rapidly reversible decrease in long-term potentiation, synaptic transmission, glutamate receptor protein levels, and ERK/MAPK activation in the dorsal hippocampus.
- Author
-
Ravassard P, Pachoud B, Comte J, Mejia-Perez C, Scoté-Blachon C, Gay N, Claustrat B, Touret M, Luppi P, and Salin PA
- Published
- 2009
64. The mesenchyme controls the timing of pancreatic beta-cell differentiation.
- Author
-
Duvillié B, Attali M, Bounacer A, Ravassard P, Basmaciogullari A, Scharfmann R, Duvillié, Bertrand, Attali, Myriam, Bounacer, Ali, Ravassard, Philippe, Basmaciogullari, Annie, and Scharfmann, Raphael
- Abstract
The importance of mesenchymal-epithelial interactions in the proliferation of pancreatic progenitor cells is well established. Here, we provide evidence that the mesenchyme also controls the timing of beta-cell differentiation. When rat embryonic pancreatic epithelium was cultured without mesenchyme, we found first rapid induction in epithelial progenitor cells of the transcription factor neurogenin3 (Ngn3), a master gene controlling endocrine cell-fate decisions in progenitor cells; then beta-cell differentiation occurred. In the presence of mesenchyme, Ngn3 induction was delayed, and few beta-cells developed. This effect of the mesenchyme on Ngn3 induction was mediated by cell-cell contacts and required a functional Notch pathway. We then showed that associating Ngn3-expressing epithelial cells with mesenchyme resulted in poor beta-cell development via a mechanism mediated by soluble factors. Thus, in addition to its effect upstream of Ngn3, the mesenchyme regulated beta-cell differentiation downstream of Ngn3. In conclusion, these data indicate that the mesenchyme controls the timing of beta-cell differentiation both upstream and downstream of Ngn3. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
65. Large CTG Repeats Trigger p16-Dependent Premature Senescence in Myotonic Dystrophy Type 1 Muscle Precursor Cells
- Author
-
Bigot, Anne, Klein, Arnaud F., Gasnier, Erwan, Jacquemin, Virginie, Ravassard, Philippe, Butler-Browne, Gillian, Mouly, Vincent, and Furling, Denis
- Abstract
A CTG repeat amplification is responsible for the dominantly inherited neuromuscular disorder, myotonic dystrophy type 1 (DM1), which is characterized by progressive muscle wasting and weakness. The expanded (CTG)n tract not only alters the myogenic differentiation of the DM1 muscle precursor cells but also reduces their proliferative capacity. In this report, we show that these muscle precursor cells containing large CTG expansion sequences have not exhausted their proliferative capacity, but have entered into premature senescence. We demonstrate that an abnormal accumulation of p16 is responsible for this defect because the abolition of p16 activity overcomes early growth arrest and restores an extended proliferative capacity. Our results suggest that the accelerated telomere shortening measured in DM1 cells does not contribute to the aberrant induction of p16. We propose that a cellular stress related to the amplified CTG repeat promotes premature senescence mediated by a p16-dependent pathway in DM1 muscle precursor cells. This mechanism is responsible for the reduced proliferative capacity of the DM1 muscle precursor cells and could participate in both the impaired regeneration and atrophy observed in the DM1 muscles containing large CTG expansions.
- Published
- 2009
- Full Text
- View/download PDF
66. A Human Tyrosine Hydroxylase Isoform Associated with Progressive Supranuclear Palsy Shows Altered Enzymatic Activity*
- Author
-
Bodeau-Péan, Sylvie, Ravassard, Philippe, Neuner-Jehle, Martin, Faucheux, Baptiste, Mallet, Jacques, and Dumas, Sylvie
- 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 Ser31and 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.
- Published
- 1999
- Full Text
- View/download PDF
67. ZNF74,a Gene Deleted in DiGeorge Syndrome, Is Expressed in Human Neural Crest-Derived Tissues and Foregut Endoderm Epithelia
- Author
-
Ravassard, Philippe, Côté, Francine, Grondin, Benoı&̂t, Bazinet, Martine, Mallet, Jacques, and Aubry, Muriel
- Abstract
DiGeorge syndrome (DGS) is a developmental disorder associated with large hemizygous deletions on chromosome 22q11.2. ZNF74zinc finger gene is a candidate from the commonly deleted region. To address the potential involvement of ZNF74in DGS, its human developmental expression pattern has been assessed. In situhybridization on Carnegie Stage 18 embryos revealed that ZNF74expression 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, ZNF74expression 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 ZNF74in structures affected in DGS suggests a role for this putative regulator of gene expression in aspects of the DGS phenotype.
- Published
- 1999
- Full Text
- View/download PDF
68. Human Tyrosine Hydroxylase Isoforms
- Author
-
Alterio, Jeanine, Ravassard, Philippe, Haavik, Jan, Le Caer, Jean-Pierre, Biguet, Nicole Faucon, Waksman, Gilles, and Mallet, Jacques
- 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 coliand 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). Ser40was phosphorylated by PKA, and both Ser19and Ser40were 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.
- Published
- 1998
- Full Text
- View/download PDF
69. Detection of two new polymorphic sites in the human interleukin1β gene
- Author
-
Laurent, C., Thibaut, F., Ravassard, P., Campion, D., Samolyk, D., Lafargue, C., Petit, M., Martinez, M., and Mallet, J.
- Abstract
The pathogenesis of schizophrenia might involve abnormal development of the human brain. Interleukin-1β is a cytokine implicated in the development of the central nervous system and therefore its gene is a candidate gene in schizophrenia. Polymorphisms within the coding sequence and the 3'UTR of the IL1β gene were searched for using PCR-SSCP. Two polymorphisms, 1B-175/1B-173and 1B-1765/1B-1763were found in addition to the previously published TaqI site. Furthermore, a mutant was found in codon 106 (exon 5) of the IL1β gene located next to the published polymorphism at the TaqI site and abolishing this site. This novel mutation encodes an Asp in place of an Asn and was only observed in one patient in our French population. Association studies were conducted with the polymorphisms 1B-175/1B-173and TaqI. There was no allelic or genotypic association between either of the two polymorphisms and schizophrenia. In our population, there is no evidence that the IL1β gene is involved in schizophrenia.
- Published
- 1997
70. Realization and Control of Autonomous Microconveyors with Piezoelectric Actuators
- Author
-
Ravassard, J.C., Abadie, J., Duffaud, J., Ferniot, J.C., Le Moal, P., Chaillet, N., Dembélé, S., and Bourjault, A.
- Abstract
This paper deals with the development of centimeter size robots for tasks of conveying and positioning. Their advantages are their multi-degrees of freedom, their nanometer range of positioning.
- Published
- 1998
- Full Text
- View/download PDF
71. Characterization of the human tryptophan hydroxylase gene promoter. Transcriptional regulation by cAMP requires a new motif distinct from the cAMP-responsive element.
- Author
-
Boularand, S, Darmon, M C, Ravassard, P, and Mallet, J
- Abstract
We isolated and sequenced 2,117 nucleotides of the promoter region of the human tryptophan hydroxylase (TPH) gene. Transient transfection in pinealocyte cultures and PC12 cells was used to investigate the human TPH (hTPH) gene promoter activity and its regulation by the cAMP signaling pathway. A region of 2,117 base pairs upstream of the transcription initiation site of the hTPH gene efficiently directed the transcription of a luciferase reporter gene but not in a cell-specific manner. The hTPH promoter activity was significantly enhanced by a cyclic AMP analog in the two cell types. Deletion analysis showed that the promoter region from -73 to +2 is sufficient to direct cAMP-dependent transcription, although it does not contain a motif exhibiting a significant identity to the cAMP-responsive element (CRE) or AP-2 binding site. Following site-directed mutagenesis of the region between -73 and -51, an inverted CCAAT box motif was identified as essential for cAMP inducibility of the hTPH promoter. This sequence between -73 and -51 alone allowed cAMP enhancement of transcription when fused to a heterologous promoter. Additionally, electrophoretic mobility shift assays showed that a specific protein-DNA complex is formed between an oligonucleotide corresponding to the inverted CCAAT box motif and nuclear proteins from pinealocytes treated or not treated with cAMP. Thus cAMP responsiveness of hTPH gene expression is mediated by a cis-acting element, which shares strong identity with an inverted CCAAT box and which binds to a constitutively produced nuclear factor.
- Published
- 1995
72. Neonatal diabetes mutations disrupt a chromatin pioneering function that activates the human insulin gene
- Author
-
Akerman, Ildem, Maestro, Miguel Angel, De Franco, Elisa, Grau, Vanessa, Flanagan, Sarah, García-Hurtado, Javier, Mittler, Gerhard, Ravassard, Philippe, Piemonti, Lorenzo, Ellard, Sian, Hattersley, Andrew T., and Ferrer, Jorge
- Abstract
Despite the central role of chromosomal context in gene transcription, human noncoding DNA variants are generally studied outside of their genomic location. This limits our understanding of disease-causing regulatory variants. INSpromoter mutations cause recessive neonatal diabetes. We show that all INS promoter point mutations in 60 patients disrupt a CC dinucleotide, whereas none affect other elements important for episomal promoter function. To model CC mutations, we humanized an ∼3.1-kb region of the mouse Ins2gene. This recapitulated developmental chromatin states and cell-specific transcription. A CC mutant allele, however, abrogated active chromatin formation during pancreas development. A search for transcription factors acting through this element revealed that another neonatal diabetes gene product, GLIS3, has a pioneer-like ability to derepress INSchromatin, which is hampered by the CC mutation. Our in vivoanalysis, therefore, connects two human genetic defects in an essential mechanism for developmental activation of the INSgene.
- Published
- 2021
- Full Text
- View/download PDF
73. In vivo imaging of the spatiotemporal activity of the eIF2α-ATF4 signaling pathway: Insights into stress and related disorders
- Author
-
Chaveroux, Cédric, Carraro, Valérie, Canaple, Laurence, Averous, Julien, Maurin, Anne-Catherine, Jousse, Céline, Muranishi, Yuki, Parry, Laurent, Mesclon, Florent, Gatti, Evelina, Mallet, Jacques, Ravassard, Philippe, Pierre, Philippe, Fafournoux, Pierre, and Bruhat, Alain
- Abstract
A transgenic mouse model enables analysis of the involvement of a stress signaling pathway in liver fibrosis.
- Published
- 2015
- Full Text
- View/download PDF
74. MiR-132 controls pancreatic beta cell proliferation and survival through Pten/Akt/Foxo3 signaling
- Author
-
Hassan Mziaut, Georg Henniger, Katharina Ganss, Sebastian Hempel, Steffen Wolk, Johanna McChord, Kamal Chowdhury, Philippe Ravassard, Klaus-Peter Knoch, Christian Krautz, Jürgen Weitz, Robert Grützmann, Christian Pilarsky, Michele Solimena, and Stephan Kersting
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: MicroRNAs (miRNAs) play an integral role in maintaining beta cell function and identity. Deciphering their targets and precise role, however, remains challenging. In this study, we aimed to identify miRNAs and their downstream targets involved in the regeneration of islet beta cells following partial pancreatectomy in mice. Methods: RNA from laser capture microdissected (LCM) islets of partially pancreatectomized and sham-operated mice were profiled with microarrays to identify putative miRNAs implicated in beta cell regeneration. Altered expression of the selected miRNAs, including miR-132, was verified by RT-PCR. Potential targets of miR-132 were selected through bioinformatic data mining. Predicted miR-132 targets were validated for their changed RNA, protein expression levels, and signaling upon miR-132 knockdown and/or overexpression in mouse MIN6 and human EndoC-βH1 insulinoma cells. The ability of miR-132 to foster beta cell proliferation in vivo was further assessed in pancreatectomized miR-132−/− and control mice. Results: Partial pancreatectomy significantly increased the number of BrdU+/insulin+ islet cells. Microarray profiling revealed that 14 miRNAs, including miR-132 and -141, were significantly upregulated in the LCM islets of the partially pancreatectomized mice compared to the LCM islets of the control mice. In the same comparison, miR-760 was the only downregulated miRNA. The changed expression of these miRNAs in the islets of the partially pancreatectomized mice was confirmed by RT-PCR only in the case of miR-132 and -141. Based on previous knowledge of its function, we focused our attention on miR-132. Downregulation of miR-132 reduced the proliferation of MIN6 cells while enhancing the levels of pro-apoptotic cleaved caspase-9. The opposite was observed in miR-132 overexpressing MIN6 cells. Microarray profiling, RT-PCR, and immunoblotting of the latter cells demonstrated their downregulated expression of Pten with concomitant increased levels of pro-proliferative factors phospho-Akt and phospho-Creb and inactivation of pro-apoptotic Foxo3a via its phosphorylation. Downregulation of Pten was further confirmed in the LCM islets of pancreatectomized mice compared to the sham-operated mice. Moreover, overexpression of miR-132 correlated with increased proliferation of EndoC-βH1 cells. The regeneration of beta cells following partial pancreatectomy was lower in the miR-132/212−/− mice than the control littermates. Conclusions: This study provides compelling evidence about the critical role of miR-132 for the regeneration of mouse islet beta cells through the downregulation of its target Pten. Hence, the miR-132/Pten/Akt/Foxo3 signaling pathway may represent a suitable target to enhance beta cell mass. Keywords: miR-132, β cell regeneration, Apoptosis, Pten/Akt/Foxo3a, Pancreatectomy
- Published
- 2020
- Full Text
- View/download PDF
75. Production and Characterization of a Conditionally Immortalized Dog Beta-Cell Line from Fetal Canine Pancreas
- Author
-
P Czernichow, K Reynaud, and P Ravassard
- Subjects
Medicine - 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.
- Published
- 2020
- Full Text
- View/download PDF
76. Patient-Derived Midbrain Organoids to Explore the Molecular Basis of Parkinson's Disease
- Author
-
Benjamin Galet, Hélène Cheval, and Philippe Ravassard
- Subjects
Parkinson's disease ,IPS (induced pluripotent stem) cell ,organoid ,midbrain ,dopamine ,genetics ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Induced pluripotent stem cell-derived organoids offer an unprecedented access to complex human tissues that recapitulate features of architecture, composition and function of in vivo organs. In the context of Parkinson's Disease (PD), human midbrain organoids (hMO) are of significant interest, as they generate dopaminergic neurons expressing markers of Substantia Nigra identity, which are the most vulnerable to degeneration. Combined with genome editing approaches, hMO may thus constitute a valuable tool to dissect the genetic makeup of PD by revealing the effects of risk variants on pathological mechanisms in a representative cellular environment. Furthermore, the flexibility of organoid co-culture approaches may also enable the study of neuroinflammatory and neurovascular processes, as well as interactions with other brain regions that are also affected over the course of the disease. We here review existing protocols to generate hMO, how they have been used so far to model PD, address challenges inherent to organoid cultures, and discuss applicable strategies to dissect the molecular pathophysiology of the disease. Taken together, the research suggests that this technology represents a promising alternative to 2D in vitro models, which could significantly improve our understanding of PD and help accelerate therapeutic developments.
- Published
- 2020
- Full Text
- View/download PDF
77. Using Inhibitory DREADDs to Silence LC Neurons in Monkeys
- Author
-
Pauline Perez, Estelle Chavret-Reculon, Philippe Ravassard, and Sebastien Bouret
- Subjects
noradrenaline ,chemogenetics ,neuronal inhibition ,locus coeruleus ,primates ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Understanding the role of the noradrenergic nucleus locus coeruleus (LC) in cognition and behavior is critical: It is involved in several key behavioral functions such as stress and vigilance, as well as in cognitive processes such as attention and decision making. In recent years, the development of viral tools has provided a clear insight into numerous aspects of brain functions in rodents. However, given the specificity of primate brains and the key benefit of monkey research for translational applications, developing viral tools to study the LC in monkeys is essential for understanding its function and exploring potential clinical strategies. Here, we describe a pharmacogenetics approach that allows to selectively and reversibly inactivate LC neurons using Designer Receptors Exclusively Activated by Designer Drugs (DREADD). We show that the expression of the hM4Di DREADD can be restricted to noradrenergic LC neurons and that the amount of LC inhibition can be adjusted by adapting the dose of the specific DREADD activator deschloroclozapine (DCZ). Indeed, even if high doses (>0.3 mg/kg) induce a massive inhibition of LC neurons and a clear decrease in vigilance, smaller doses (
- Published
- 2022
- Full Text
- View/download PDF
78. Production, Characterization, and Function of Pseudoislets from Perinatal Canine Pancreas
- Author
-
P. Czernichow, K. Reynaud, J. Kerr-Conte, E. Furthner, and P. Ravassard
- Subjects
Medicine - 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 β 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.
- Published
- 2019
- Full Text
- View/download PDF
79. The EndoC-βH1 cell line is a valid model of human beta cells and applicable for screenings to identify novel drug target candidates
- Author
-
Violeta Georgieva Tsonkova, Fredrik Wolfhagen Sand, Xenia Asbæk Wolf, Lars Groth Grunnet, Anna Kirstine Ringgaard, Camilla Ingvorsen, Louise Winkel, Mark Kalisz, Kevin Dalgaard, Christine Bruun, Johannes Josef Fels, Charlotte Helgstrand, Sven Hastrup, Fredrik Kryh Öberg, Erik Vernet, Michael Paolo Bastner Sandrini, Allan Christian Shaw, Carsten Jessen, Mads Grønborg, Jacob Hald, Hanni Willenbrock, Dennis Madsen, Rasmus Wernersson, Lena Hansson, Jan Nygaard Jensen, Annette Plesner, Tomas Alanentalo, Maja Borup Kjær Petersen, Anne Grapin-Botton, Christian Honoré, Jonas Ahnfelt-Rønne, Jacob Hecksher-Sørensen, Philippe Ravassard, Ole D. Madsen, Claude Rescan, and Thomas Frogne
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: To characterize the EndoC-βH1 cell line as a model for human beta cells and evaluate its beta cell functionality, focusing on insulin secretion, proliferation, apoptosis and ER stress, with the objective to assess its potential as a screening platform for identification of novel anti-diabetic drug candidates. Methods: EndoC-βH1 was transplanted into mice for validation of in vivo functionality. Insulin secretion was evaluated in cells cultured as monolayer and as pseudoislets, as well as in diabetic mice. Cytokine induced apoptosis, glucolipotoxicity, and ER stress responses were assessed. Beta cell relevant mRNA and protein expression were investigated by qPCR and antibody staining. Hundreds of proteins or peptides were tested for their effect on insulin secretion and proliferation. Results: Transplantation of EndoC-βH1 cells restored normoglycemia in streptozotocin induced diabetic mice. Both in vitro and in vivo, we observed a clear insulin response to glucose, and, in vitro, we found a significant increase in insulin secretion from EndoC-βH1 pseudoislets compared to monolayer cultures for both glucose and incretins.Apoptosis and ER stress were inducible in the cells and caspase 3/7 activity was elevated in response to cytokines, but not affected by the saturated fatty acid palmitate.By screening of various proteins and peptides, we found Bombesin (BB) receptor agonists and Pituitary Adenylate Cyclase-Activating Polypeptides (PACAP) to significantly induce insulin secretion and the proteins SerpinA6, STC1, and APOH to significantly stimulate proliferation.ER stress was readily induced by Tunicamycin and resulted in a reduction of insulin mRNA. Somatostatin (SST) was found to be expressed by 1% of the cells and manipulation of the SST receptors was found to significantly affect insulin secretion. Conclusions: Overall, the EndoC-βH1 cells strongly resemble human islet beta cells in terms of glucose and incretin stimulated insulin secretion capabilities. The cell line has an active cytokine induced caspase 3/7 apoptotic pathway and is responsive to ER stress initiation factors. The cells' ability to proliferate can be further increased by already known compounds as well as by novel peptides and proteins. Based on its robust performance during the functionality assessment assays, the EndoC-βH1 cell line was successfully used as a screening platform for identification of novel anti-diabetic drug candidates. Keywords: EndoC-βH1, Pseudoislets, Glucose stimulated insulin secretion, Somatostatin signaling, Proliferation
- Published
- 2018
- Full Text
- View/download PDF
80. A human beta cell line with drug inducible excision of immortalizing transgenes
- Author
-
Marion Benazra, Marie-José Lecomte, Claire Colace, Andreas Müller, Cécile Machado, Severine Pechberty, Emilie Bricout-Neveu, Maud Grenier-Godard, Michele Solimena, Raphaël Scharfmann, Paul Czernichow, and Philippe Ravassard
- Subjects
Cell engineering ,Human pancreatic beta cell line ,Conditional immortalization ,Tamoxifen inducible CRE ,Human beta cell function ,Internal medicine ,RC31-1245 - Abstract
Objectives: Access to immortalized human pancreatic beta cell lines that are phenotypically close to genuine adult beta cells, represent a major tool to better understand human beta cell physiology and develop new therapeutics for Diabetes. Here we derived a new conditionally immortalized human beta cell line, EndoC-βH3 in which immortalizing transgene can be efficiently removed by simple addition of tamoxifen. Methods: We used lentiviral mediated gene transfer to stably integrate a tamoxifen inducible form of CRE (CRE-ERT2) into the recently developed conditionally immortalized EndoC βH2 line. The resulting EndoC-βH3 line was characterized before and after tamoxifen treatment for cell proliferation, insulin content and insulin secretion. Results: We showed that EndoC-βH3 expressing CRE-ERT2 can be massively amplified in culture. We established an optimized tamoxifen treatment to efficiently excise the immortalizing transgenes resulting in proliferation arrest. In addition, insulin expression raised by 12 fold and insulin content increased by 23 fold reaching 2 μg of insulin per million cells. Such massive increase was accompanied by enhanced insulin secretion upon glucose stimulation. We further observed that tamoxifen treated cells maintained a stable function for 5 weeks in culture. Conclusions: EndoC βH3 cell line represents a powerful tool that allows, using a simple and efficient procedure, the massive production of functional non-proliferative human beta cells. Such cells are close to genuine human beta cells and maintain a stable phenotype for 5 weeks in culture.
- Published
- 2015
- Full Text
- View/download PDF
81. Differentiation of Sendai Virus-Reprogrammed iPSC into β Cells, Compared with Human Pancreatic Islets and Immortalized β Cell Line
- Author
-
Silvia Pellegrini, Fabio Manenti, Raniero Chimienti, Rita Nano, Linda Ottoboni, Francesca Ruffini, Gianvito Martino, Philippe Ravassard, Lorenzo Piemonti, and Valeria Sordi
- Subjects
Medicine - Abstract
Background: New sources of insulin-secreting cells are strongly in demand for treatment of diabetes. Induced pluripotent stem cells (iPSCs) have the potential to generate insulin-producing cells (iβ). However, the gene expression profile and secretory function of iβ still need to be validated in comparison with native β cells. Methods: Two clones of human iPSCs, reprogrammed from adult fibroblasts through integration-free Sendai virus, were differentiated into iβ and compared with donor pancreatic islets and EndoC-βH1, an immortalized human β cell line. Results: Both clones of iPSCs differentiated into insulin + cells with high efficiency (up to 20%). iβ were negative for pluripotency markers (Oct4, Sox2, Ssea4) and positive for Pdx1, Nkx6.1, Chromogranin A, PC1/3, insulin, glucagon and somatostatin. iβ basally secreted C-peptide, glucagon and ghrelin and released insulin in response either to increasing concentration of glucose or a depolarizing stimulus. The comparison revealed that iβ are remarkably similar to donor derived islets in terms of gene and protein expression profile and similar level of heterogeneity. The ability of iβ to respond to glucose instead was more related to that of EndoC-βH1. Discussion: We demonstrated that insulin-producing cells generated from iPSCs recapitulate fundamental gene expression profiles and secretory function of native human β cells.
- Published
- 2018
- Full Text
- View/download PDF
82. A distal intergenic region controls pancreatic endocrine differentiation by acting as a transcriptional enhancer and as a polycomb response element.
- Author
-
Joris van Arensbergen, Sebastien Dussaud, Corinne Pardanaud-Glavieux, Javier García-Hurtado, Claire Sauty, Aline Guerci, Jorge Ferrer, and Philippe Ravassard
- Subjects
Medicine ,Science - Abstract
Lineage-selective expression of developmental genes is dependent on the interplay between activating and repressive mechanisms. Gene activation is dependent on cell-specific transcription factors that recognize transcriptional enhancer sequences. Gene repression often depends on the recruitment of Polycomb group (PcG) proteins, although the sequences that underlie the recruitment of PcG proteins, also known as Polycomb response elements (PREs), remain poorly understood in vertebrates. While distal PREs have been identified in mammals, a role for positive-acting enhancers in PcG-mediated repression has not been described. Here we have used a highly efficient procedure based on lentiviral-mediated transgenesis to carry out in vivo fine-mapping of, cis-regulatory sequences that control lineage-specific activation of Neurog3, a master regulator of pancreatic endocrine differentiation. Our findings reveal an enhancer region that is sufficient to drive correct spacio-temporal expression of Neurog3 and demonstrate that this same region serves as a PRE in alternative lineages where Neurog3 is inactive.
- Published
- 2017
- Full Text
- View/download PDF
83. Characterization of stimulus-secretion coupling in the human pancreatic EndoC-βH1 beta cell line.
- Author
-
Lotta E Andersson, Bérengère Valtat, Annika Bagge, Vladimir V Sharoyko, David G Nicholls, Philippe Ravassard, Raphael Scharfmann, Peter Spégel, and Hindrik Mulder
- Subjects
Medicine ,Science - Abstract
Studies on beta cell metabolism are often conducted in rodent beta cell lines due to the lack of stable human beta cell lines. Recently, a human cell line, EndoC-βH1, was generated. Here we investigate stimulus-secretion coupling in this cell line, and compare it with that in the rat beta cell line, INS-1 832/13, and human islets.Cells were exposed to glucose and pyruvate. Insulin secretion and content (radioimmunoassay), gene expression (Gene Chip array), metabolite levels (GC/MS), respiration (Seahorse XF24 Extracellular Flux Analyzer), glucose utilization (radiometric), lactate release (enzymatic colorimetric), ATP levels (enzymatic bioluminescence) and plasma membrane potential and cytoplasmic Ca2+ responses (microfluorometry) were measured. Metabolite levels, respiration and insulin secretion were examined in human islets.Glucose increased insulin release, glucose utilization, raised ATP production and respiratory rates in both lines, and pyruvate increased insulin secretion and respiration. EndoC-βH1 cells exhibited higher insulin secretion, while plasma membrane depolarization was attenuated, and neither glucose nor pyruvate induced oscillations in intracellular calcium concentration or plasma membrane potential. Metabolite profiling revealed that glycolytic and TCA-cycle intermediate levels increased in response to glucose in both cell lines, but responses were weaker in EndoC-βH1 cells, similar to those observed in human islets. Respiration in EndoC-βH1 cells was more similar to that in human islets than in INS-1 832/13 cells.Functions associated with early stimulus-secretion coupling, with the exception of plasma membrane potential and Ca2+ oscillations, were similar in the two cell lines; insulin secretion, respiration and metabolite responses were similar in EndoC-βH1 cells and human islets. While both cell lines are suitable in vitro models, with the caveat of replicating key findings in isolated islets, EndoC-βH1 cells have the advantage of carrying the human genome, allowing studies of human genetic variants, epigenetics and regulatory RNA molecules.
- Published
- 2015
- Full Text
- View/download PDF
84. Highly efficient in vitro and in vivo delivery of functional RNAs using new versatile MS2-chimeric retrovirus-like particles
- Author
-
Anne Prel, Vincent Caval, Régis Gayon, Philippe Ravassard, Christine Duthoit, Emmanuel Payen, Leila Maouche-Chretien, Alison Creneguy, Tuan Huy Nguyen, Nicolas Martin, Eric Piver, Raphaël Sevrain, Lucille Lamouroux, Philippe Leboulch, Frédéric Deschaseaux, Pascale Bouillé, Luc Sensébé, and Jean-Christophe Pagès
- Subjects
Genetics ,QH426-470 ,Cytology ,QH573-671 - Abstract
RNA delivery is an attractive strategy to achieve transient gene expression in research projects and in cell- or gene-based therapies. Despite significant efforts investigating vector-directed RNA transfer, there is still a requirement for better efficiency of delivery to primary cells and in vivo. Retroviral platforms drive RNA delivery, yet retrovirus RNA-packaging constraints limit gene transfer to two genome-molecules per viral particle. To improve retroviral transfer, we designed a dimerization-independent MS2-driven RNA packaging system using MS2-Coat-retrovirus chimeras. The engineered chimeric particles promoted effective packaging of several types of RNAs and enabled efficient transfer of biologically active RNAs in various cell types, including human CD34+ and iPS cells. Systemic injection of high-titer particles led to gene expression in mouse liver and transferring Cre-recombinase mRNA in muscle permitted widespread editing at the ROSA26 locus. We could further show that the VLPs were able to activate an osteoblast differentiation pathway by delivering RUNX2- or DLX5-mRNA into primary human bone-marrow mesenchymal-stem cells. Thus, the novel chimeric MS2-lentiviral particles are a versatile tool for a wide range of applications including cellular-programming or genome-editing.
- Published
- 2015
- Full Text
- View/download PDF
85. Comparison Between Several Integrase-defective Lentiviral Vectors Reveals Increased Integration of an HIV Vector Bearing a D167H Mutant
- Author
-
Muhammad Qamar Saeed, Noelle Dufour, Cynthia Bartholmae, Urzula Sieranska, Malaika Knopf, Eloïse Thierry, Sylvain Thierry, Olivier Delelis, Nicolas Grandchamp, Héloïse Pilet, Philippe Ravassard, Julie Massonneau, Françoise Pflumio, Christof von Kalle, François Lachapelle, Alexis-Pierre Bemelmans, Manfred Schmidt, and Ché Serguera
- Subjects
CD34+ progenitors ,integrase ,integration sites ,lentiviral vectors ,residual integration ,transduction ,Therapeutics. Pharmacology ,RM1-950 - Abstract
HIV-1 derived vectors are among the most efficient for gene transduction in mammalian tissues. As the parent virus, they carry out vector genome insertion into the host cell chromatin. Consequently, their preferential integration in transcribed genes raises several conceptual and safety issues. To address part of these questions, HIV-derived vectors have been engineered to be nonintegrating. This was mainly achieved by mutating HIV-1 integrase at functional hotspots of the enzyme enabling the development of streamlined nuclear DNA circles functional for transgene expression. Few integrase mutant vectors have been successfully tested so far for gene transfer. They are cleared with time in mitotic cells, but stable within nondividing retina cells or neurons. Here, we compared six HIV vectors carrying different integrases, either wild type or with different mutations (D64V, D167H, Q168A, K186Q+Q214L+Q216L, and RRK262-264AAH) shown to modify integrase enzymatic activity, oligomerization, or interaction with key cellular cofactor of HIV DNA integration as LEDGF/p75 or TNPO3. We show that these mutations differently affect the transduction efficiency as well as rates and patterns of integration of HIV-derived vectors suggesting their different processing in the nucleus. Surprisingly and most interestingly, we report that an integrase carrying the D167H substitution improves vector transduction efficiency and integration in both HEK-293T and primary CD34+ cells.
- Published
- 2014
- Full Text
- View/download PDF
86. Neonatal diabetes mutations disrupt a chromatin pioneering function that activates the human insulin gene
- Author
-
Sian Ellard, Jorge Ferrer, Javier García-Hurtado, Sarah E. Flanagan, Philippe Ravassard, Miguel A. Maestro, Elisa De Franco, Ildem Akerman, Gerhard Mittler, Andrew T. Hattersley, Lorenzo Piemonti, Vanessa Grau, University of Birmingham [Birmingham], Centre for Genomic Regulation [Barcelona] (CRG), Universitat Pompeu Fabra [Barcelona] (UPF)-Centro Nacional de Analisis Genomico [Barcelona] (CNAG), University of Exeter Medical School, University of Exeter, Max Planck Institute of Immunobiology and Epigenetics (MPI-IE), Max-Planck-Gesellschaft, Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), IRCCS Ospedale San Raffaele [Milan, Italy], Gestionnaire, Hal Sorbonne Université, Institut du Cerveau = Paris Brain Institute (ICM), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Akerman, I., Maestro, M. A., De Franco, E., Grau, V., Flanagan, S., Garcia-Hurtado, J., Mittler, G., Ravassard, P., Piemonti, L., Ellard, S., Hattersley, A. T., Ferrer, J., and Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Transcription, Genetic ,[SDV]Life Sciences [q-bio] ,Endocrinology, Diabetes and Metabolism ,medicine.disease_cause ,Sulfonylurea Receptors ,Bioinformatics ,Infant, Newborn, Diseases ,Mice ,0302 clinical medicine ,Endocrinology ,Transcription (biology) ,Insulina ,Human insulin ,Insulin ,Protein Isoforms ,Medicine ,Biology (General) ,Promoter Regions, Genetic ,health care economics and organizations ,Genetics ,Mutation ,GLIS3 ,Diabetis ,Gene Expression Regulation, Developmental ,Chromatin ,3. Good health ,Cell biology ,[SDV] Life Sciences [q-bio] ,DNA-Binding Proteins ,neonatal diabetes ,QH301-705.5 ,Neonatal diabetes ,mouse model ,education ,MEDLINE ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Gene product ,03 medical and health sciences ,Text mining ,Diabetes Mellitus ,Animals ,Humans ,Point Mutation ,Transcription factor ,Pancreas ,Gene ,Alleles ,regulatory element ,HIP ,INS promoter ,business.industry ,Point mutation ,Infants nadons -- Malalties ,Infant, Newborn ,Embryo, Mammalian ,Noncoding DNA ,Repressor Proteins ,030104 developmental biology ,Trans-Activators ,business ,030217 neurology & neurosurgery ,Function (biology) ,Genètica - Abstract
Summary Despite the central role of chromosomal context in gene transcription, human noncoding DNA variants are generally studied outside of their genomic location. This limits our understanding of disease-causing regulatory variants. INS promoter mutations cause recessive neonatal diabetes. We show that all INS promoter point mutations in 60 patients disrupt a CC dinucleotide, whereas none affect other elements important for episomal promoter function. To model CC mutations, we humanized an ∼3.1-kb region of the mouse Ins2 gene. This recapitulated developmental chromatin states and cell-specific transcription. A CC mutant allele, however, abrogated active chromatin formation during pancreas development. A search for transcription factors acting through this element revealed that another neonatal diabetes gene product, GLIS3, has a pioneer-like ability to derepress INS chromatin, which is hampered by the CC mutation. Our in vivo analysis, therefore, connects two human genetic defects in an essential mechanism for developmental activation of the INS gene., Graphical abstract, Highlights • Mutations of a CC dinucleotide in the human INS promoter cause neonatal diabetes • We humanized ∼3.1 kb of mouse Ins2 and created a CC mutant version • Humanized Ins2, but not the CC mutant, recapitulates developmental chromatin activation • GLIS3, also mutated in diabetes, activates INS chromatin and requires an intact CC, Mutations in the CC element of the INS promoter or the transcription factor GLIS3 cause neonatal diabetes. Akerman et al. humanize a 3.1-kb region upstream of the mouse Ins2 gene and show that GLIS3 and the CC element form a pioneering mechanism that activates INS chromatin during pancreas development.
- Published
- 2021
87. The inactivation of Arx in pancreatic α-cells triggers their neogenesis and conversion into functional β-like cells.
- Author
-
Monica Courtney, Elisabet Gjernes, Noémie Druelle, Christophe Ravaud, Andhira Vieira, Nouha Ben-Othman, Anja Pfeifer, Fabio Avolio, Gunter Leuckx, Sandra Lacas-Gervais, Fanny Burel-Vandenbos, Damien Ambrosetti, Jacob Hecksher-Sorensen, Philippe Ravassard, Harry Heimberg, Ahmed Mansouri, and Patrick Collombat
- Subjects
Genetics ,QH426-470 - Abstract
Recently, it was demonstrated that pancreatic new-born glucagon-producing cells can regenerate and convert into insulin-producing β-like cells through the ectopic expression of a single gene, Pax4. Here, combining conditional loss-of-function and lineage tracing approaches, we show that the selective inhibition of the Arx gene in α-cells is sufficient to promote the conversion of adult α-cells into β-like cells at any age. Interestingly, this conversion induces the continuous mobilization of duct-lining precursor cells to adopt an endocrine cell fate, the glucagon(+) cells thereby generated being subsequently converted into β-like cells upon Arx inhibition. Of interest, through the generation and analysis of Arx and Pax4 conditional double-mutants, we provide evidence that Pax4 is dispensable for these regeneration processes, indicating that Arx represents the main trigger of α-cell-mediated β-like cell neogenesis. Importantly, the loss of Arx in α-cells is sufficient to regenerate a functional β-cell mass and thereby reverse diabetes following toxin-induced β-cell depletion. Our data therefore suggest that strategies aiming at inhibiting the expression of Arx, or its molecular targets/co-factors, may pave new avenues for the treatment of diabetes.
- Published
- 2013
- Full Text
- View/download PDF
88. Multisensory control of multimodal behavior: do the legs know what the tongue is doing?
- Author
-
Jesse D Cushman, Daniel B Aharoni, Bernard Willers, Pascal Ravassard, Ashley Kees, Cliff Vuong, Briana Popeney, Katsushi Arisaka, and Mayank R Mehta
- Subjects
Medicine ,Science - Abstract
Understanding of adaptive behavior requires the precisely controlled presentation of multisensory stimuli combined with simultaneous measurement of multiple behavioral modalities. Hence, we developed a virtual reality apparatus that allows for simultaneous measurement of reward checking, a commonly used measure in associative learning paradigms, and navigational behavior, along with precisely controlled presentation of visual, auditory and reward stimuli. Rats performed a virtual spatial navigation task analogous to the Morris maze where only distal visual or auditory cues provided spatial information. Spatial navigation and reward checking maps showed experience-dependent learning and were in register for distal visual cues. However, they showed a dissociation, whereby distal auditory cues failed to support spatial navigation but did support spatially localized reward checking. These findings indicate that rats can navigate in virtual space with only distal visual cues, without significant vestibular or other sensory inputs. Furthermore, they reveal the simultaneous dissociation between two reward-driven behaviors.
- Published
- 2013
- Full Text
- View/download PDF
89. Directed pancreatic acinar differentiation of mouse embryonic stem cells via embryonic signalling molecules and exocrine transcription factors.
- Author
-
Fabien Delaspre, Mohammad Massumi, Marta Salido, Bernat Soria, Philippe Ravassard, Pierre Savatier, and Anouchka Skoudy
- Subjects
Medicine ,Science - Abstract
Pluripotent embryonic stem cells (ESC) are a promising cellular system for generating an unlimited source of tissue for the treatment of chronic diseases and valuable in vitro differentiation models for drug testing. Our aim was to direct differentiation of mouse ESC into pancreatic acinar cells, which play key roles in pancreatitis and pancreatic cancer. To that end, ESC were first differentiated as embryoid bodies and sequentially incubated with activin A, inhibitors of Sonic hedgehog (Shh) and bone morphogenetic protein (BMP) pathways, fibroblast growth factors (FGF) and retinoic acid (RA) in order to achieve a stepwise increase in the expression of mRNA transcripts encoding for endodermal and pancreatic progenitor markers. Subsequent plating in Matrigel® and concomitant modulation of FGF, glucocorticoid, and folllistatin signalling pathways involved in exocrine differentiation resulted in a significant increase of mRNAs encoding secretory enzymes and in the number of cells co-expressing their protein products. Also, pancreatic endocrine marker expression was down-regulated and accompanied by a significant reduction in the number of hormone-expressing cells with a limited presence of hepatic marker expressing-cells. These findings suggest a selective activation of the acinar differentiation program. The newly differentiated cells were able to release α-amylase and this feature was greatly improved by lentiviral-mediated expression of Rbpjl and Ptf1a, two transcription factors involved in the maximal production of digestive enzymes. This study provides a novel method to produce functional pancreatic exocrine cells from ESC.
- Published
- 2013
- Full Text
- View/download PDF
90. Plasticity of adult human pancreatic duct cells by neurogenin3-mediated reprogramming.
- Author
-
Nathalie Swales, Geert A Martens, Stefan Bonné, Yves Heremans, Rehannah Borup, Mark Van de Casteele, Zhidong Ling, Daniel Pipeleers, Philippe Ravassard, Finn Nielsen, Jorge Ferrer, and Harry Heimberg
- Subjects
Medicine ,Science - Abstract
Aims/hypothesisDuct cells isolated from adult human pancreas can be reprogrammed to express islet beta cell genes by adenoviral transduction of the developmental transcription factor neurogenin3 (Ngn3). In this study we aimed to fully characterize the extent of this reprogramming and intended to improve it.MethodsThe extent of the Ngn3-mediated duct-to-endocrine cell reprogramming was measured employing genome wide mRNA profiling. By modulation of the Delta-Notch signaling or addition of pancreatic endocrine transcription factors Myt1, MafA and Pdx1 we intended to improve the reprogramming.ResultsNgn3 stimulates duct cells to express a focused set of genes that are characteristic for islet endocrine cells and/or neural tissues. This neuro-endocrine shift however, is incomplete with less than 10% of full duct-to-endocrine reprogramming achieved. Transduction of exogenous Ngn3 activates endogenous Ngn3 suggesting auto-activation of this gene. Furthermore, pancreatic endocrine reprogramming of human duct cells can be moderately enhanced by inhibition of Delta-Notch signaling as well as by co-expressing the transcription factor Myt1, but not MafA and Pdx1.Conclusions/interpretationThe results provide further insight into the plasticity of adult human duct cells and suggest measurable routes to enhance Ngn3-mediated in vitro reprogramming protocols for regenerative beta cell therapy in diabetes.
- Published
- 2012
- Full Text
- View/download PDF
91. Ectopic PDX-1 expression directly reprograms human keratinocytes along pancreatic insulin-producing cells fate.
- Author
-
Michal Mauda-Havakuk, Naomi Litichever, Ellad Chernichovski, Odelia Nakar, Eyal Winkler, Ram Mazkereth, Arie Orenstein, Eran Bar-Meir, Philippe Ravassard, Irit Meivar-Levy, and Sarah Ferber
- Subjects
Medicine ,Science - Abstract
BackgroundCellular differentiation and lineage commitment have previously been considered irreversible processes. However, recent studies have indicated that differentiated adult cells can be reprogrammed to pluripotency and, in some cases, directly into alternate committed lineages. However, although pluripotent cells can be induced in numerous somatic cell sources, it was thought that inducing alternate committed lineages is primarily only possible in cells of developmentally related tissues. Here, we challenge this view and analyze whether direct adult cell reprogramming to alternate committed lineages can cross the boundaries of distinct developmental germ layers.Methodology/principal findingsWe ectopically expressed non-integrating pancreatic differentiation factors in ectoderm-derived human keratinocytes to determine whether these factors could directly induce endoderm-derived pancreatic lineage and β-cell-like function. We found that PDX-1 and to a lesser extent other pancreatic transcription factors, could rapidly and specifically activate pancreatic lineage and β-cell-like functional characteristics in ectoderm-derived human keratinocytes. Human keratinocytes transdifferentiated along the β cell lineage produced processed and secreted insulin in response to elevated glucose concentrations. Using irreversible lineage tracing for KRT-5 promoter activity, we present supporting evidence that insulin-positive cells induced by ectopic PDX-1 expression are generated in ectoderm derived keratinocytes.Conclusions/significanceThese findings constitute the first demonstration of human ectoderm cells to endoderm derived pancreatic cells transdifferentiation. The study represents a proof of concept which suggests that transcription factors induced reprogramming is wider and more general developmental process than initially considered. These results expanded the arsenal of adult cells that can be used as a cell source for generating functional endocrine pancreatic cells. Directly reprogramming somatic cells into alternate desired tissues has important implications in developing patient-specific, regenerative medicine approaches.
- Published
- 2011
- Full Text
- View/download PDF
92. Insulin-producing cells generated from dedifferentiated human pancreatic beta cells expanded in vitro.
- Author
-
Holger A Russ, Elad Sintov, Leeat Anker-Kitai, Orr Friedman, Ayelet Lenz, Ginat Toren, Chen Farhy, Metsada Pasmanik-Chor, Varda Oron-Karni, Philippe Ravassard, and Shimon Efrat
- Subjects
Medicine ,Science - Abstract
Expansion of beta cells from the limited number of adult human islet donors is an attractive prospect for increasing cell availability for cell therapy of diabetes. However, attempts at expanding human islet cells in tissue culture result in loss of beta-cell phenotype. Using a lineage-tracing approach we provided evidence for massive proliferation of beta-cell-derived (BCD) cells within these cultures. Expansion involves dedifferentiation resembling epithelial-mesenchymal transition (EMT). Epigenetic analyses indicate that key beta-cell genes maintain open chromatin structure in expanded BCD cells, although they are not transcribed. Here we investigated whether BCD cells can be redifferentiated into beta-like cells.Redifferentiation conditions were screened by following activation of an insulin-DsRed2 reporter gene. Redifferentiated cells were characterized for gene expression, insulin content and secretion assays, and presence of secretory vesicles by electron microscopy. BCD cells were induced to redifferentiate by a combination of soluble factors. The redifferentiated cells expressed beta-cell genes, stored insulin in typical secretory vesicles, and released it in response to glucose. The redifferentiation process involved mesenchymal-epithelial transition, as judged by changes in gene expression. Moreover, inhibition of the EMT effector SLUG (SNAI2) using shRNA resulted in stimulation of redifferentiation. Lineage-traced cells also gave rise at a low rate to cells expressing other islet hormones, suggesting transition of BCD cells through an islet progenitor-like stage during redifferentiation.These findings demonstrate for the first time that expanded dedifferentiated beta cells can be induced to redifferentiate in culture. The findings suggest that ex-vivo expansion of adult human islet cells is a promising approach for generation of insulin-producing cells for transplantation, as well as basic research, toxicology studies, and drug screening.
- Published
- 2011
- Full Text
- View/download PDF
93. Quantitative comparison of constitutive promoters in human ES cells.
- Author
-
Karin Norrman, Yvonne Fischer, Blandine Bonnamy, Fredrik Wolfhagen Sand, Philippe Ravassard, and Henrik Semb
- Subjects
Medicine ,Science - Abstract
BACKGROUND: Constitutive promoters that ensure sustained and high level gene expression are basic research tools that have a wide range of applications, including studies of human embryology and drug discovery in human embryonic stem cells (hESCs). Numerous cellular/viral promoters that ensure sustained gene expression in various cell types have been identified but systematic comparison of their activities in hESCs is still lacking. METHODOLOGY/PRINCIPAL FINDINGS: We have quantitatively compared promoter activities of five commonly used constitutive promoters, including the human β-actin promoter (ACTB), cytomegalovirus (CMV), elongation factor-1α, (EF1α), phosphoglycerate kinase (PGK) and ubiquitinC (UbC) in hESCs. Lentiviral gene transfer was used to ensure stable integration of promoter-eGFP constructs into the hESCs genome. Promoter activities were quantitatively compared in long term culture of undifferentiated hESCs and in their differentiated progenies. CONCLUSION/SIGNIFICANCE: The ACTB, EF1α and PGK promoters showed stable activities during long term culture of undifferentiated hESCs. The ACTB promoter was superior by maintaining expression in 75-80% of the cells after 50 days in culture. During embryoid body (EB) differentiation, promoter activities of all five promoters decreased. Although the EF1α promoter was downregulated in approximately 50% of the cells, it was the most stable promoter during differentiation. Gene expression analysis of differentiated eGFP+ and eGFP- cells indicate that promoter activities might be restricted to specific cell lineages, suggesting the need to carefully select optimal promoters for constitutive gene expression in differentiated hESCs.
- Published
- 2010
- Full Text
- View/download PDF
94. Epithelial-mesenchymal transition in cells expanded in vitro from lineage-traced adult human pancreatic beta cells.
- Author
-
Holger A Russ, Philippe Ravassard, Julie Kerr-Conte, Francois Pattou, and Shimon Efrat
- Subjects
Medicine ,Science - Abstract
BackgroundIn-vitro expansion of functional beta cells from adult human islets is an attractive approach for generating an abundant source of cells for beta-cell replacement therapy of diabetes. Using genetic cell-lineage tracing we have recently shown that beta cells cultured from adult human islets undergo rapid dedifferentiation and proliferate for up to 16 population doublings. These cells have raised interest as potential candidates for redifferentiation into functional insulin-producing cells. Previous work has associated dedifferentiation of cultured epithelial cells with epithelial-mesenchymal transition (EMT), and suggested that EMT generates cells with stem cell properties. Here we investigated the occurrence of EMT in these cultures and assessed their stem cell potential.Methodology/principal findingsUsing cell-lineage tracing we provide direct evidence for occurrence of EMT in cells originating from beta cells in cultures of adult human islet cells. These cells express multiple mesenchymal markers, as well as markers associated with mesenchymal stem cells (MSC). However, we do not find evidence for the ability of such cells, nor of cells in these cultures derived from a non-beta-cell origin, to significantly differentiate into mesodermal cell types.Conclusions/significanceThese findings constitute the first demonstration based on genetic lineage-tracing of EMT in cultured adult primary human cells, and show that EMT does not induce multipotency in cells derived from human beta cells.
- Published
- 2009
- Full Text
- View/download PDF
95. A new strategy to generate functional insulin-producing cell lines by somatic gene transfer into pancreatic progenitors.
- Author
-
Philippe Ravassard, Emilie Bricout-Neveu, Yasmine Hazhouz, Severine Pechberty, Jacques Mallet, Paul Czernichow, and Raphael Scharfmann
- Subjects
Medicine ,Science - Abstract
BACKGROUND: There is increasing interest in developing human cell lines to be used to better understand cell biology, but also for drug screening, toxicology analysis and future cell therapy. In the endocrine pancreatic field, functional human beta cell lines are extremely scarce. On the other hand, rodent insulin producing beta cells have been generated during the past years with great success. Many of such cell lines were produced by using transgenic mice expressing SV40T antigen under the control of the insulin promoter, an approach clearly inadequate in human. Our objective was to develop and validate in rodent an alternative transgenic-like approach, applicable to human tissue, by performing somatic gene transfer into pancreatic progenitors that will develop into beta cells. METHODS AND FINDINGS: In this study, rat embryonic pancreases were transduced with recombinant lentiviral vector expressing the SV40T antigen under the control of the insulin promoter. Transduced tissues were next transplanted under the kidney capsule of immuno-incompetent mice allowing insulinoma development from which beta cell lines were established. Gene expression profile, insulin content and glucose dependent secretion, normalization of glycemia upon transplantation into diabetic mice validated the approach to generate beta cell lines. CONCLUSIONS: Somatic gene transfer into pancreatic progenitors represents an alternative strategy to generate functional beta cell lines in rodent. Moreover, this approach can be generalized to derive cells lines from various tissues and most importantly from tissues of human origin.
- Published
- 2009
- Full Text
- View/download PDF
96. Localization of the brainstem GABAergic neurons controlling paradoxical (REM) sleep.
- Author
-
Emilie Sapin, Damien Lapray, Anne Bérod, Romain Goutagny, Lucienne Léger, Pascal Ravassard, Olivier Clément, Lucie Hanriot, Patrice Fort, and Pierre-Hervé Luppi
- Subjects
Medicine ,Science - Abstract
Paradoxical sleep (PS) is a state characterized by cortical activation, rapid eye movements and muscle atonia. Fifty years after its discovery, the neuronal network responsible for the genesis of PS has been only partially identified. We recently proposed that GABAergic neurons would have a pivotal role in that network. To localize these GABAergic neurons, we combined immunohistochemical detection of Fos with non-radioactive in situ hybridization of GAD67 mRNA (GABA synthesis enzyme) in control rats, rats deprived of PS for 72 h and rats allowed to recover after such deprivation. Here we show that GABAergic neurons gating PS (PS-off neurons) are principally located in the ventrolateral periaqueductal gray (vlPAG) and the dorsal part of the deep mesencephalic reticular nucleus immediately ventral to it (dDpMe). Furthermore, iontophoretic application of muscimol for 20 min in this area in head-restrained rats induced a strong and significant increase in PS quantities compared to saline. In addition, we found a large number of GABAergic PS-on neurons in the vlPAG/dDPMe region and the medullary reticular nuclei known to generate muscle atonia during PS. Finally, we showed that PS-on neurons triggering PS localized in the SLD are not GABAergic. Altogether, our results indicate that multiple populations of PS-on GABAergic neurons are distributed in the brainstem while only one population of PS-off GABAergic neurons localized in the vlPAG/dDpMe region exist. From these results, we propose a revised model for PS control in which GABAergic PS-on and PS-off neurons localized in the vlPAG/dDPMe region play leading roles.
- Published
- 2009
- Full Text
- View/download PDF
97. Inhibition of Eph/ephrin interaction with the small molecule UniPR500 improves glucose tolerance in healthy and insulin-resistant mice
- Author
-
Lorenzo Piemonti, Fabio Manenti, Daniele Pala, Paola Chiodelli, Alessio Lodola, Francesca Ferlenghi, Marco Rusnati, Simona Bertoni, Carmine Giorgio, Philippe Ravassard, R. Di Lecce, Elisabetta Barocelli, Matteo Incerti, Massimiliano Tognolini, Simonetta Russo, Anna Maria Cantoni, Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Giorgio, C, Incerti, M, Pala, D, Russo, S, Chiodelli, P, Rusnati, M, Cantoni, A M, Di Lecce, R, Barocelli, E, Bertoni, S, Ravassard, P, Manenti, F, Piemonti, L, Ferlenghi, F, Lodola, A, and Tognolini, M
- Subjects
0301 basic medicine ,Male ,medicine.medical_treatment ,Pharmacology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,In vivo ,Insulin-Secreting Cells ,Insulin Secretion ,medicine ,Glucose homeostasis ,Ephrin ,Animals ,Humans ,Hypoglycemic Agents ,Insulin ,Protein Interaction Maps ,Eph/ephrins ,ComputingMilieux_MISCELLANEOUS ,Chemistry ,Pancreatic islets ,Diabetes ,Erythropoietin-producing hepatocellular (Eph) receptor ,Antagonist ,Glucose Tolerance Test ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Diabetes Mellitus, Type 1 ,Glucose ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,030220 oncology & carcinogenesis ,Insulin Resistance ,Ephrins - Abstract
Eph/ephrin interactions and their bidirectional signaling are integral part of the complex communication system between β-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-βH1 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-βH1 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 β-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.
- Published
- 2019
98. Human pancreatic β cell lncRNAs control cell-specific regulatory networks
- Author
-
Akerman, Ildem, Tu, Zhidong, Beucher, Anthony, Rolando, Delphine M. Y., Sauty-Colace, Claire, Benazra, Marion, Nakic, Nikolina, Yang, Jialiang, Wang, Huan, Pasquali, Lorenzo, Moran, Ignasi, Garcia Hurtado, Javier, Castro, Natalia, Gonzalez Franco, Roser, Stewart, Andrew F., Bonner, Caroline, Piemonti, Lorenzo, Berney, Thierry, Groop, Leif, Kerr-Conte, Julie, Pattou, Francois, Argmann, Carmen, Schadt, Eric, Ravassard, Phillipe, Jorge Ferrer, Universitat Autònoma de Barcelona, Wellcome Trust, Medical Research Council (MRC), National Institutes of Health, Imperial College Healthcare NHS Trust- BRC Funding, Pathology/molecular and cellular medicine, Akerman, I., Tu, Z., Beucher, A., Rolando, D. M. Y., Sauty Colace, C., Benazra, M., Nakic, N., Yang, J., Wang, H., Pasquali, L., Moran, I., Garcia Hurtado, J., Castro, N., Gonzalez Franco, R., Stewart, A. F., Bonner, C., Piemonti, Lorenzo, Berney, T., Groop, L., Kerr Conte, J., Pattou, F., Argmann, C., Schadt, E., Ravassard, P., and Ferrer, J.
- Subjects
0301 basic medicine ,Transcription, Genetic ,Physiology ,Cell ,Gene regulatory network ,LINCRNAS ,0601 Biochemistry and Cell Biology ,TRANSCRIPTIONAL LANDSCAPE ,PLUTO ,transcriptional networks ,0302 clinical medicine ,Transcription (biology) ,Insulin-Secreting Cells ,Gene expression ,Insulin Secretion ,Insulin ,Gene Regulatory Networks ,long noncoding RNAs ,CRISPR interference ,GENE-EXPRESSION ,Genetics ,Regulation of gene expression ,PDX1 ,0303 health sciences ,geography.geographical_feature_category ,ddc:617 ,diabetes ,Islet ,ENDOCRINE PANCREAS ,Chromatin ,Cell biology ,DIFFERENTIATION ,Phenotype ,medicine.anatomical_structure ,1101 Medical Biochemistry and Metabolomics ,Gene Knockdown Techniques ,Multigene Family ,RNA, Long Noncoding ,type 2 diabetes ,Life Sciences & Biomedicine ,lncRNAs ,LINE ,Genomics ,Biology ,LONG NONCODING RNAS ,Article ,Endocrinology & Metabolism ,03 medical and health sciences ,REVEALS ,Journal Article ,medicine ,Humans ,RNA, Messenger ,Molecular Biology ,Transcription factor ,030304 developmental biology ,Homeodomain Proteins ,geography ,Science & Technology ,pancreatic islets ,PDX-1 ,Cell Biology ,EVOLUTION ,030104 developmental biology ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,Trans-Activators ,chromatin ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
Summary Recent studies have uncovered thousands of long non-coding RNAs (lncRNAs) in human pancreatic β cells. β cell lncRNAs are often cell type specific and exhibit dynamic regulation during differentiation or upon changing glucose concentrations. Although these features hint at a role of lncRNAs in β cell gene regulation and diabetes, the function of β cell lncRNAs remains largely unknown. In this study, we investigated the function of β cell-specific lncRNAs and transcription factors using transcript knockdowns and co-expression network analysis. This revealed lncRNAs that function in concert with transcription factors to regulate β cell-specific transcriptional networks. We further demonstrate that the lncRNA PLUTO affects local 3D chromatin structure and transcription of PDX1, encoding a key β cell transcription factor, and that both PLUTO and PDX1 are downregulated in islets from donors with type 2 diabetes or impaired glucose tolerance. These results implicate lncRNAs in the regulation of β cell-specific transcription factor networks., Graphical Abstract, Highlights • A loss-of-function screen reveals functional β cell lncRNAs • Cell-specific lncRNAs and transcription factors regulate common gene networks • The lncRNA PLUTO influences interactions between an enhancer cluster and PDX1 • PLUTO and PDX1 are deregulated in type 2 diabetes and impaired glucose tolerance, Akerman et al. studied the function of human β cell lncRNAs with RNAi, CRISPRi, and co-expression networks. This revealed β cell lncRNAs and transcription factors that control common regulatory networks. One lncRNA, PLUTO, is downregulated in type 2 diabetes and controls PDX1, encoding a key β cell transcription factor.
- Published
- 2016
99. EndoC-βH5 cells are storable and ready-to-use human pancreatic beta cells with physiological insulin secretion.
- Author
-
Blanchi B, Taurand M, Colace C, Thomaidou S, Audeoud C, Fantuzzi F, Sawatani T, Gheibi S, Sabadell-Basallote J, Boot FWJ, Chantier T, Piet A, Cavanihac C, Pilette M, Balguerie A, Olleik H, Carlotti F, Ejarque M, Fex M, Mulder H, Cnop M, Eizirik DL, Jouannot O, Gaffuri AL, Czernichow P, Zaldumbide A, Scharfmann R, and Ravassard P
- Subjects
- Humans, Insulin Secretion, Cell Line, Insulin metabolism, Transcription Factors metabolism, Glucose metabolism, Insulin-Secreting Cells metabolism
- Abstract
Objectives: Readily accessible human pancreatic beta cells that are functionally close to primary adult beta cells are a crucial model to better understand human beta cell physiology and develop new treatments for diabetes. We here report the characterization of EndoC-βH5 cells, the latest in the EndoC-βH cell family., Methods: EndoC-βH5 cells were generated by integrative gene transfer of immortalizing transgenes hTERT and SV40 large T along with Herpes Simplex Virus-1 thymidine kinase into human fetal pancreas. Immortalizing transgenes were removed after amplification using CRE activation and remaining non-excized cells eliminated using ganciclovir. Resulting cells were distributed as ready to use EndoC-βH5 cells. We performed transcriptome, immunological and extensive functional assays., Results: Ready to use EndoC-βH5 cells display highly efficient glucose dependent insulin secretion. A robust 10-fold insulin secretion index was observed and reproduced in four independent laboratories across Europe. EndoC-βH5 cells secrete insulin in a dynamic manner in response to glucose and secretion is further potentiated by GIP and GLP-1 analogs. RNA-seq confirmed abundant expression of beta cell transcription factors and functional markers, including incretin receptors. Cytokines induce a gene expression signature of inflammatory pathways and antigen processing and presentation. Finally, modified HLA-A2 expressing EndoC-βH5 cells elicit specific A2-alloreactive CD8 T cell activation., Conclusions: EndoC-βH5 cells represent a unique storable and ready to use human pancreatic beta cell model with highly robust and reproducible features. Such cells are thus relevant for the study of beta cell function, screening and validation of new drugs, and development of disease models., Competing Interests: Declaration of Competing Interest BB, MT, AP, CC, MP, AB and HO are or were employees at Human Cell Design SA, France, the company that commercializes EndoC-βH1 and EndoC-βh5 cells and associated media. RS, PC and PR are shareholders at HCD., (Copyright © 2023 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
100. C9ORF72 knockdown triggers FTD-like symptoms and cell pathology in mice.
- Author
-
Lopez-Herdoiza MB, Bauché S, Wilmet B, Le Duigou C, Roussel D, Frah M, Béal J, Devely G, Boluda S, Frick P, Bouteiller D, Dussaud S, Guillabert P, Dalle C, Dumont M, Camuzat A, Saracino D, Barbier M, Bruneteau G, Ravassard P, Neumann M, Nicole S, Le Ber I, Brice A, and Latouche M
- Abstract
The GGGGCC intronic repeat expansion within C9ORF72 is the most common genetic cause of ALS and FTD. This mutation results in toxic gain of function through accumulation of expanded RNA foci and aggregation of abnormally translated dipeptide repeat proteins, as well as loss of function due to impaired transcription of C9ORF72 . A number of in vivo and in vitro models of gain and loss of function effects have suggested that both mechanisms synergize to cause the disease. However, the contribution of the loss of function mechanism remains poorly understood. We have generated C9ORF72 knockdown mice to mimic C9-FTD/ALS patients haploinsufficiency and investigate the role of this loss of function in the pathogenesis. We found that decreasing C9ORF72 leads to anomalies of the autophagy/lysosomal pathway, cytoplasmic accumulation of TDP-43 and decreased synaptic density in the cortex. Knockdown mice also developed FTD-like behavioral deficits and mild motor phenotypes at a later stage. These findings show that C9ORF72 partial loss of function contributes to the damaging events leading to C9-FTD/ALS., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Lopez-Herdoiza, Bauché, Wilmet, Le Duigou, Roussel, Frah, Béal, Devely, Boluda, Frick, Bouteiller, Dussaud, Guillabert, Dalle, Dumont, Camuzat, Saracino, Barbier, Bruneteau, Ravassard, Neumann, Nicole, Le Ber, Brice and Latouche.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.