56 results on '"Gilbert Bernier"'
Search Results
2. COCO/DAND5 inhibits developmental and pathological ocular angiogenesis
- Author
-
Natalija Popovic, Erika Hooker, Andrea Barabino, Anthony Flamier, Frédéric Provost, Manuel Buscarlet, Gilbert Bernier, and Bruno Larrivée
- Subjects
angiogenesis ,COCO ,energy metabolism ,ocular pathologies ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
Abstract Neovascularization contributes to multiple visual disorders including age‐related macular degeneration (AMD) and retinopathy of prematurity. Current therapies for treating ocular angiogenesis are centered on the inhibition of vascular endothelial growth factor (VEGF). While clinically effective, some AMD patients are refractory or develop resistance to anti‐VEGF therapies and concerns of increased risks of developing geographic atrophy following long‐term treatment have been raised. Identification of alternative pathways to inhibit pathological angiogenesis is thus important. We have identified a novel inhibitor of angiogenesis, COCO, a member of the Cerberus‐related DAN protein family. We demonstrate that COCO inhibits sprouting, migration and cellular proliferation of cultured endothelial cells. Intravitreal injections of COCO inhibited retinal vascularization during development and in models of retinopathy of prematurity. COCO equally abrogated angiogenesis in models of choroidal neovascularization. Mechanistically, COCO inhibited TGFβ and BMP pathways and altered energy metabolism and redox balance of endothelial cells. Together, these data show that COCO is an inhibitor of retinal and choroidal angiogenesis, possibly representing a therapeutic option for the treatment of neovascular ocular diseases.
- Published
- 2021
- Full Text
- View/download PDF
3. Alzheimer’s disease: a tale of two diseases?
- Author
-
Eleonora Nardini, Ryan Hogan, Anthony Flamier, and Gilbert Bernier
- Subjects
aging ,alzheimer’s disease ,bmi1 ,epigenetics ,familial ,late-onset ,sporadic ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Sporadic late-onset Alzheimer’s disease (SLOAD) and familial early-onset Alzheimer’s disease (FEOAD) associated with dominant mutations in APP, PSEN1 and PSEN2, are thought to represent a spectrum of the same disorder based on near identical behavioral and histopathological features. Hence, FEOAD transgenic mouse models have been used in past decades as a surrogate to study SLOAD pathogenic mechanisms and as the gold standard to validate drugs used in clinical trials. Unfortunately, such research has yielded little output in terms of therapeutics targeting the disease’s development and progression. In this short review, we interrogate the widely accepted view of one, dimorphic disease through the prism of the Bmi1+/– mouse model and the distinct chromatin signatures observed between SLOAD and FEOAD brains.
- Published
- 2021
- Full Text
- View/download PDF
4. Deregulation of Neuro-Developmental Genes and Primary Cilium Cytoskeleton Anomalies in iPSC Retinal Sheets from Human Syndromic Ciliopathies
- Author
-
Andrea Barabino, Anthony Flamier, Roy Hanna, Elise Héon, Benjamin S. Freedman, and Gilbert Bernier
- Subjects
Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Summary: Ciliopathies are heterogeneous genetic diseases affecting primary cilium structure and function. Meckel-Gruber (MKS) and Bardet-Biedl (BBS) syndromes are severe ciliopathies characterized by skeletal and neurodevelopment anomalies, including polydactyly, cognitive impairment, and retinal degeneration. We describe the generation and molecular characterization of human induced pluripotent stem cell (iPSC)-derived retinal sheets (RSs) from controls, and MKS (TMEM67) and BBS (BBS10) cases. MKS and BBS RSs displayed significant common alterations in the expression of hundreds of developmental genes and members of the WNT and BMP pathways. Induction of crystallin molecular chaperones was prominent in MKS and BBS RSs suggesting a stress response to misfolded proteins. Unique to MKS photoreceptors was the presence of supernumerary centrioles and cilia, and aggregation of ciliary proteins. Unique to BBS photoreceptors was the accumulation of DNA damage and activation of the mitotic spindle checkpoint. This study reveals how combining cell reprogramming, organogenesis, and next-generation sequencing enables the elucidation of mechanisms involved in human ciliopathies. : In this article, G. Bernier and colleagues report on the production of iPSCs from control, and Meckel-Gruber syndrome (MKS) and Bardet-Biedl syndrome (BBS) cases. These were differentiated into retinal sheets largely composed of photoreceptors. Using molecular and cell biological approaches, they further reveal known and novel disease mechanisms associated with these retinal ciliopathies, including genomic instability in BBS and supernumerary cilia and centrioles in MKS. Keywords: cilia, primary cilium, ciliopathy, basal body, photoreceptor, retinal degeneration, RNA sequencing, organogenesis, development, disease modeling
- Published
- 2020
- Full Text
- View/download PDF
5. Induction of rod versus cone photoreceptor-specific progenitors from retinal precursor cells
- Author
-
Saeed Khalili, Brian G. Ballios, Justin Belair-Hickey, Laura Donaldson, Jeff Liu, Brenda L.K. Coles, Kenneth N. Grisé, Tahani Baakdhah, Gary D. Bader, Valerie A. Wallace, Gilbert Bernier, Molly S. Shoichet, and Derek van der Kooy
- Subjects
Biology (General) ,QH301-705.5 - Abstract
During development, multipotent progenitors undergo temporally-restricted differentiation into post-mitotic retinal cells; however, the mechanisms of progenitor division that occurs during retinogenesis remain controversial. Using clonal analyses (lineage tracing and single cell cultures), we identify rod versus cone lineage-specific progenitors derived from both adult retinal stem cells and embryonic neural retinal precursors. Taurine and retinoic acid are shown to act in an instructive and lineage-restricted manner early in the progenitor lineage hierarchy to produce rod-restricted progenitors from stem cell progeny. We also identify an instructive, but lineage-independent, mechanism for the specification of cone-restricted progenitors through the suppression of multiple differentiation signaling pathways. These data indicate that exogenous signals play critical roles in directing lineage decisions and resulting in fate-restricted rod or cone photoreceptor progenitors in culture. Additional factors may be involved in governing photoreceptor fates in vivo. Keywords: Stem cell, Retina, Rod photoreceptor, Cone photoreceptor, Progenitors
- Published
- 2018
- Full Text
- View/download PDF
6. Modeling Late-Onset Sporadic Alzheimer’s Disease through BMI1 Deficiency
- Author
-
Anthony Flamier, Jida El Hajjar, James Adjaye, Karl J. Fernandes, Mohamed Abdouh, and Gilbert Bernier
- Subjects
Alzheimer’s disease ,dementia ,sporadic ,BMI1 ,polycomb ,amyloid ,Tau ,p53 ,GSK3b ,MAPT ,Biology (General) ,QH301-705.5 - Abstract
Late-onset sporadic Alzheimer’s disease (AD) is the most prevalent form of dementia, but its origin remains poorly understood. The Bmi1/Ring1 protein complex maintains transcriptional repression of developmental genes through histone H2A mono-ubiquitination, and Bmi1 deficiency in mice results in growth retardation, progeria, and neurodegeneration. Here, we demonstrate that BMI1 is silenced in AD brains, but not in those with early-onset familial AD, frontotemporal dementia, or Lewy body dementia. BMI1 expression was also reduced in cortical neurons from AD patient-derived induced pluripotent stem cells but not in neurons overexpressing mutant APP and PSEN1. BMI1 knockout in human post-mitotic neurons resulted in amyloid beta peptide secretion and deposition, p-Tau accumulation, and neurodegeneration. Mechanistically, BMI1 was required to repress microtubule associated protein tau (MAPT) transcription and prevent GSK3beta and p53 stabilization, which otherwise resulted in neurodegeneration. Restoration of BMI1 activity through genetic or pharmaceutical approaches could represent a therapeutic strategy against AD.
- Published
- 2018
- Full Text
- View/download PDF
7. Differentiation of Human Embryonic Stem Cells into Cone Photoreceptors
- Author
-
Anthony Flamier, Andrea Barabino, and Gilbert Bernier
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Photoreceptors are specialized retinal neurons able to respond to light in order to generate visual information. Among photoreceptors, cones are involved in colors discrimination and high-resolution central vision and are selectively depleted in macular degenerations and cone dystrophies. A possible therapeutic solution for these disorders is to replace degenerating cells with functional cones. Here, we describe a simple protocol for the rapid production of large amount of cone photoreceptors from human pluripotent stem cells. The differentiation protocol is based on the “default pathway” of neural induction using the BMP, TGFβ and WNT antagonist COCO.
- Published
- 2016
- Full Text
- View/download PDF
8. Bmi1 is down-regulated in the aging brain and displays antioxidant and protective activities in neurons.
- Author
-
Mohamed Abdouh, Wassim Chatoo, Jida El Hajjar, Jocelyn David, José Ferreira, and Gilbert Bernier
- Subjects
Medicine ,Science - Abstract
Aging increases the risk to develop several neurodegenerative diseases, although the underlying mechanisms are poorly understood. Inactivation of the Polycomb group gene Bmi1 in mice results in growth retardation, cerebellar degeneration, and development of a premature aging-like phenotype. This progeroid phenotype is characterized by formation of lens cataracts, apoptosis of cortical neurons, and increase of reactive oxygen species (ROS) concentrations, owing to p53-mediated repression of antioxidant response (AOR) genes. Herein we report that Bmi1 expression progressively declines in the neurons of aging mouse and human brains. In old brains, p53 accumulates at the promoter of AOR genes, correlating with a repressed chromatin state, down-regulation of AOR genes, and increased oxidative damages to lipids and DNA. Comparative gene expression analysis further revealed that aging brains display an up-regulation of the senescence-associated genes IL-6, p19(Arf) and p16(Ink4a), along with the pro-apoptotic gene Noxa, as seen in Bmi1-null mice. Increasing Bmi1 expression in cortical neurons conferred robust protection against DNA damage-induced cell death or mitochondrial poisoning, and resulted in suppression of ROS through activation of AOR genes. These observations unveil that Bmi1 genetic deficiency recapitulates aspects of physiological brain aging and that Bmi1 over-expression is a potential therapeutic modality against neurodegeneration.
- Published
- 2012
- Full Text
- View/download PDF
9. Alzheimer’s disease: a tale of two diseases?
- Author
-
Gilbert Bernier, Ryan Hogan, Eleonora Nardini, and Anthony Flamier
- Subjects
0301 basic medicine ,Genetically modified mouse ,Review ,Disease ,lcsh:RC346-429 ,03 medical and health sciences ,0302 clinical medicine ,Developmental Neuroscience ,PSEN2 ,PSEN1 ,late-onset ,Medicine ,Epigenetics ,aging ,alzheimer’s disease ,bmi1 ,epigenetics ,familial ,sporadic ,lcsh:Neurology. Diseases of the nervous system ,business.industry ,Alzheimer's disease ,BMI1 ,Chromatin ,Clinical trial ,030104 developmental biology ,business ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Sporadic late-onset Alzheimer’s disease (SLOAD) and familial early-onset Alzheimer’s disease (FEOAD) associated with dominant mutations in APP, PSEN1 and PSEN2, are thought to represent a spectrum of the same disorder based on near identical behavioral and histopathological features. Hence, FEOAD transgenic mouse models have been used in past decades as a surrogate to study SLOAD pathogenic mechanisms and as the gold standard to validate drugs used in clinical trials. Unfortunately, such research has yielded little output in terms of therapeutics targeting the disease’s development and progression. In this short review, we interrogate the widely accepted view of one, dimorphic disease through the prism of the Bmi1+/– mouse model and the distinct chromatin signatures observed between SLOAD and FEOAD brains.
- Published
- 2021
10. Off-target effect of the BMI1 inhibitor PTC596 drives epithelial-mesenchymal transition in glioblastoma multiforme
- Author
-
Rimi Hamam, Roy Hanna, Anthony Flamier, Niraj Patel, Gilbert Bernier, Mohamed Abdouh, Andrea Barabino, and Andy Gao
- Subjects
0301 basic medicine ,Cancer Research ,Brain tumor ,BMI1 Inhibitor PTC596 ,macromolecular substances ,Biology ,lcsh:RC254-282 ,Article ,03 medical and health sciences ,0302 clinical medicine ,Cancer stem cell ,medicine ,Epithelial–mesenchymal transition ,Epigenetics ,Clonogenic assay ,Cancer stem cells ,EZH2 ,medicine.disease ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,3. Good health ,CNS cancer ,030104 developmental biology ,Oncology ,BMI1 ,030220 oncology & carcinogenesis ,Cancer research - Abstract
Glioblastoma multiforme (GBM) is an incurable primary brain tumor containing a sub-population of cancer stem cells (CSCs). Polycomb Repressive Complex (PRC) proteins BMI1 and EZH2 are enriched in CSCs, promoting clonogenic growth and resistance to genotoxic therapies. We report here that when used at appropriate concentrations, pharmaceutical inhibitors of BMI1 could efficiently prevent GBM colony growth and CSC self-renewal in vitro and significantly extend lifespan in terminally ill tumor-bearing mice. Notably, molecular analyses revealed that the commonly used PTC596 molecule targeted both BMI1 and EZH2, possibly providing beneficial therapeutic effects in some contexts. On the other hand, treatment with PTC596 resulted in instant reactivation of EZH2 target genes and induction of a molecular program of epithelial–mesenchymal transition (EMT), possibly explaining the modified phenotype of some PTC596-treated tumors. Treatment with a related but more specific BMI1 inhibitor resulted in tumor regression and maintenance of cell identity. We conclude that inhibition of BMI1 alone is efficient at inducing GBM regression, and that dual inhibition of BMI1 and EZH2 using PTC596 may be also beneficial but only in specific contexts.
- Published
- 2020
- Full Text
- View/download PDF
11. COCO/DAND5 inhibits developmental and pathological ocular angiogenesis
- Author
-
Erika Hooker, Manuel Buscarlet, Anthony Flamier, Andrea Barabino, Natalija Popovic, Frédéric Provost, Bruno Larrivée, and Gilbert Bernier
- Subjects
0301 basic medicine ,Cocos ,Vascular Endothelial Growth Factor A ,Medicine (General) ,Vascular Biology & Angiogenesis ,genetic structures ,Angiogenesis ,ocular pathologies ,QH426-470 ,Article ,Retina ,Neovascularization ,COCO ,03 medical and health sciences ,chemistry.chemical_compound ,angiogenesis ,0302 clinical medicine ,R5-920 ,energy metabolism ,medicine ,Genetics ,Coco ,Humans ,business.industry ,Endothelial Cells ,Retinal ,Retinopathy of prematurity ,Articles ,Macular degeneration ,medicine.disease ,Choroidal Neovascularization ,eye diseases ,Vascular endothelial growth factor ,030104 developmental biology ,Choroidal neovascularization ,chemistry ,Cancer research ,Molecular Medicine ,Intercellular Signaling Peptides and Proteins ,sense organs ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
Neovascularization contributes to multiple visual disorders including age‐related macular degeneration (AMD) and retinopathy of prematurity. Current therapies for treating ocular angiogenesis are centered on the inhibition of vascular endothelial growth factor (VEGF). While clinically effective, some AMD patients are refractory or develop resistance to anti‐VEGF therapies and concerns of increased risks of developing geographic atrophy following long‐term treatment have been raised. Identification of alternative pathways to inhibit pathological angiogenesis is thus important. We have identified a novel inhibitor of angiogenesis, COCO, a member of the Cerberus‐related DAN protein family. We demonstrate that COCO inhibits sprouting, migration and cellular proliferation of cultured endothelial cells. Intravitreal injections of COCO inhibited retinal vascularization during development and in models of retinopathy of prematurity. COCO equally abrogated angiogenesis in models of choroidal neovascularization. Mechanistically, COCO inhibited TGFβ and BMP pathways and altered energy metabolism and redox balance of endothelial cells. Together, these data show that COCO is an inhibitor of retinal and choroidal angiogenesis, possibly representing a therapeutic option for the treatment of neovascular ocular diseases., This study reveals the significant anti‐angiogenic properties of COCO/Dand5 in models of retinal and choroidal neovascularisation. Exogenous COCO inhibited endothelial cell proliferation and migration, limited TGFβ and BMP pathways and altered energy metabolism and cellular redox balance.
- Published
- 2021
12. INK4a/ARF Expression Impairs Neurogenesis in the Brain of Irradiated Mice
- Author
-
Oanh Le, Palacio Lg, Gilbert Bernier, Christian Beauséjour, Gilles R.X. Hickson, and Ines Batinic-Haberle
- Subjects
p53 ,0301 basic medicine ,Senescence ,senescence ,Tumor suppressor gene ,DNA damage ,Neurogenesis ,Subventricular zone ,Apoptosis ,Mice, Transgenic ,Radiation Dosage ,Biochemistry ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Neural Stem Cells ,Biomimetics ,Radiation, Ionizing ,Genetics ,medicine ,Animals ,neoplasms ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,INK4a/ARF ,irradiation ,biology ,Microglia ,Superoxide Dismutase ,Brain ,Cell Biology ,Molecular Imaging ,Doublecortin ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,biology.protein ,Tumor Suppressor Protein p53 ,Biomarkers ,Bromodeoxyuridine ,Developmental Biology - Abstract
Summary Brain neurogenesis is severely impaired following exposure to ionizing radiation (IR). We and others have shown that the expression of the tumor suppressor gene p16INK4a is increased in tissues exposed to IR and thus hypothesized that its expression could limit neurogenesis in the irradiated brain. Here, we found that exposure to IR leads to persistent DNA damage and the expression of p16INK4a in the hippocampus and subventricular zone regions. This was accompanied by a decline in neurogenesis, as determined by doublecortin expression and bromodeoxyuridine incorporation, an effect partially restored in Ink4a/arf-null mice. Increased neurogenesis in the absence of INK4a/ARF expression was independent of apoptosis and activation of the microglia. Moreover, treatment of irradiated mice with a superoxide dismutase mimetic or clearance of p16INK4a-expressing cells using mouse genetics failed to increase neurogenesis. In conclusion, our results suggest that IR-induced p16INK4a expression is a mechanism that limits neurogenesis., Highlights • INK4a/ARF expression is induced in neural progenitor cells following exposure to IR • Absence of INK4a/ARF expression favors neurogenesis in the irradiated brain • Genetic ablation of p16INK4a-expressing cells does not restore neurogenesis • IR-induced activation of the microglia is dependent on p53 but not INK4a/ARF expression, Palacio et al. find that ionizing radiation induces the senescence marker INK4a/ARF in neural progenitor cells and demonstrate that such expression as a mechanism leading to loss of neurogenesis. Their results suggest that treatments looking to preserve stem cell regenerative potential, by limiting INK4a/ARF expression, may help prevent radiation-related loss of neurogenesis.
- Published
- 2018
- Full Text
- View/download PDF
13. G-quadruplexes originating from evolutionary conserved L1 elements interfere with neuronal gene expression in Alzheimer's disease
- Author
-
Gilbert Bernier, Roy Hanna, Andrea Barabino, and Anthony Flamier
- Subjects
0301 basic medicine ,Male ,Pluripotent Stem Cells ,Heterochromatin ,Science ,Neurogenesis ,RNA Splicing ,General Physics and Astronomy ,Fluorescent Antibody Technique ,Stem-cell differentiation ,Biology ,Chromatin structure ,General Biochemistry, Genetics and Molecular Biology ,Article ,Euchromatin ,Histones ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Tandem repeat ,Transcription (biology) ,Alzheimer Disease ,Gene expression ,Animals ,Humans ,Cells, Cultured ,Mice, Knockout ,Neurons ,Polycomb Repressive Complex 1 ,Multidisciplinary ,Gene Expression Profiling ,General Chemistry ,Phenotype ,Cell biology ,Chromatin ,Gene expression profiling ,G-Quadruplexes ,Mice, Inbred C57BL ,030104 developmental biology ,Gene Ontology ,Long Interspersed Nucleotide Elements ,Gene Expression Regulation ,Gene Knockdown Techniques ,RNA splicing ,Chromatin Immunoprecipitation Sequencing ,Female ,030217 neurology & neurosurgery - Abstract
DNA sequences containing consecutive guanines organized in 4-interspaced tandem repeats can form stable single-stranded secondary structures, called G-quadruplexes (G4). Herein, we report that the Polycomb group protein BMI1 is enriched at heterochromatin regions containing putative G4 DNA sequences, and that G4 structures accumulate in cells with reduced BMI1 expression and/or relaxed chromatin, including sporadic Alzheimer’s disease (AD) neurons. In AD neurons, G4 structures preferentially accumulate in lamina-associated domains, and this is rescued by re-establishing chromatin compaction. ChIP-seq analyses reveal that G4 peaks correspond to evolutionary conserved Long Interspersed Element-1 (L1) sequences predicted to be transcriptionally active. Hence, G4 structures co-localize with RNAPII, and inhibition of transcription can reverse the G4 phenotype without affecting chromatin’s state, thus uncoupling both components. Intragenic G4 structures affecting splicing events are furthermore associated with reduced neuronal gene expression in AD. Active L1 sequences are thus at the origin of most G4 structures observed in human neurons., G-quadruplexes (G4) can be formed from DNA sequences containing consecutive guanines organized in 4-interspaced tandem repeats. Here the authors reveal the association between G4 structures present at specific loci in Alzheimer’s disease neurons and reduced gene expression and perturbed alternative splicing.
- Published
- 2019
14. Heterochromatic genome instability and neurodegeneration sharing similarities with Alzheimer’s disease in old Bmi1+/− mice
- Author
-
Gilbert Bernier, Patrick Nkanza, Yiu Chung Tse, Tak Pan Wong, Wassim Chatoo, Mohamed Abdouh, Nicolas Tétreault, Roy Hanna, and Jida El Hajjar
- Subjects
Male ,0301 basic medicine ,Genome instability ,Amyloid ,Heterochromatin ,DNA damage ,Transgene ,Long-Term Potentiation ,lcsh:Medicine ,Mice, Transgenic ,tau Proteins ,Kaplan-Meier Estimate ,macromolecular substances ,Biology ,Genomic Instability ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,Proto-Oncogene Proteins ,medicine ,Animals ,Humans ,Gene silencing ,Maze Learning ,lcsh:Science ,Spatial Memory ,Neurons ,Polycomb Repressive Complex 1 ,Amyloid beta-Peptides ,Multidisciplinary ,lcsh:R ,Neurodegeneration ,Brain ,medicine.disease ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Female ,lcsh:Q ,030217 neurology & neurosurgery - Abstract
Sporadic Alzheimer’s disease (AD) is the most common cause of dementia. However, representative experimental models of AD have remained difficult to produce because of the disease’s uncertain origin. The Polycomb group protein BMI1 regulates chromatin compaction and gene silencing. BMI1 expression is abundant in adult brain neurons but down-regulated in AD brains. We show here that mice lacking one allele of Bmi1 (Bmi1+/−) develop normally but present with age cognitive deficits and neurodegeneration sharing similarities with AD. Bmi1+/− mice also transgenic for the amyloid beta precursor protein died prematurely and present aggravated disease. Loss of heterochromatin and DNA damage response (DDR) at repetitive DNA sequences were predominant in Bmi1+/− mouse neurons and inhibition of the DDR mitigated the amyloid and Tau phenotype. Heterochromatin anomalies and DDR at repetitive DNA sequences were also found in AD brains. Aging Bmi1+/− mice may thus represent an interesting model to identify and study novel pathogenic mechanisms related to AD.
- Published
- 2019
- Full Text
- View/download PDF
15. Deregulation of neuro‐developmental genes and primary cilium cytoskeleton anomalies in iPSc‐derived retinal sheets from human syndromic ciliopathies
- Author
-
Roy Hanna, Gilbert Bernier, Benjamin S. Freedman, Elise Héon, Andrea Barabino, and Anthony Flamier
- Subjects
Developmental genes ,chemistry.chemical_compound ,chemistry ,Cilium ,Genetics ,Retinal ,Biology ,Cytoskeleton ,Molecular Biology ,Biochemistry ,Ciliopathies ,Biotechnology ,Cell biology - Published
- 2020
- Full Text
- View/download PDF
16. Differentiation of human embryonic stem cells into cone photoreceptors through simultaneous inhibition of BMP, TGFβ and Wnt signaling
- Author
-
Shufeng Zhou, Shashi Wadhwa, Anthony Flamier, Mohamed Abdouh, Nicolas Tétreault, Andrea Barabino, and Gilbert Bernier
- Subjects
genetic structures ,Cellular differentiation ,Blotting, Western ,Population ,Biology ,Bone morphogenetic protein ,Retinal Cone Photoreceptor Cells ,Retina ,Cell Line ,Mice ,Transforming Growth Factor beta ,Animals ,Humans ,education ,Molecular Biology ,Embryonic Stem Cells ,In Situ Hybridization ,Analysis of Variance ,education.field_of_study ,Reverse Transcriptase Polymerase Chain Reaction ,Wnt signaling pathway ,Cell Differentiation ,Anatomy ,Flow Cytometry ,Immunohistochemistry ,Embryonic stem cell ,Cell biology ,Wnt Proteins ,Transplantation ,Bone Morphogenetic Proteins ,Cerberus gene family ,Intercellular Signaling Peptides and Proteins ,sense organs ,Signal Transduction ,Developmental Biology - Abstract
Cone photoreceptors are required for color discrimination and high-resolution central vision and are lost in macular degenerations, cone and cone/rod dystrophies. Cone transplantation could represent a therapeutic solution. However, an abundant source of human cones remains difficult to obtain. Work performed in model organisms suggests that anterior neural cell fate is induced ‘by default' if BMP, TGFβ and Wnt activities are blocked, and that photoreceptor genesis operates through an S-cone default pathway. We report here that Coco (Dand5), a member of the Cerberus gene family, is expressed in the developing and adult mouse retina. Upon exposure to recombinant COCO, human embryonic stem cells (hESCs) differentiated into S-cone photoreceptors, developed an inner segment-like protrusion, and could degrade cGMP when exposed to light. Addition of thyroid hormone resulted in a transition from a unique S-cone population toward a mixed M/S-cone population. When cultured at confluence for a prolonged period of time, COCO-exposed hESCs spontaneously developed into a cellular sheet composed of polarized cone photoreceptors. COCO showed dose-dependent and synergistic activity with IGF1 at blocking BMP/TGFβ/Wnt signaling, while its cone-inducing activity was blocked in a dose-dependent manner by exposure to BMP, TGFβ or Wnt-related proteins. Our work thus provides a unique platform to produce human cones for developmental, biochemical and therapeutic studies and supports the hypothesis that photoreceptor differentiation operates through an S-cone default pathway during human retinal development.
- Published
- 2015
- Full Text
- View/download PDF
17. Photoreceptor Cell Replacement Therapy from Stem Cells
- Author
-
Gilbert Bernier
- Subjects
Retinal degeneration ,genetic structures ,Cellular differentiation ,Biology ,medicine.disease ,eye diseases ,Photoreceptor cell ,Cell therapy ,Transplantation ,medicine.anatomical_structure ,Retinitis pigmentosa ,medicine ,sense organs ,Stem cell ,Induced pluripotent stem cell ,Neuroscience - Abstract
Macular degenerations, retinitis pigmentosa, and retinal dystrophies affect millions of people worldwide. In most cases, loss of visual function results from the death of photoreceptors, the specialized cells involved in photo-transduction. An innovative and efficient therapeutic solution for retinal degenerative diseases may be photoreceptor cell transplantation. Yet, the human eye contains about one hundred million photoreceptors, and cell replacement therapy would require at least a fraction of this, raising the issue of where to find an abundant source of healthy human photoreceptors to treat patients. Human pluripotent stem cells can be expanded quasi-indefinitely and differentiate into all cell types of the human body. Methods to direct the differentiation of human pluripotent stem cells into retinal cells and photoreceptors have been developed based on developmental biology principles. Here, we review the history and evolution of these methods, looking at two-dimensional and three-dimensional cell culture systems. We also analyze the current outcomes of photoreceptor cell transplantation therapy and explore the upcoming challenges for its clinical translation.
- Published
- 2018
- Full Text
- View/download PDF
18. Modeling Late-Onset Sporadic Alzheimer's Disease through BMI1 Deficiency
- Author
-
Karl J.L. Fernandes, Gilbert Bernier, Jida El Hajjar, Anthony Flamier, James Adjaye, and Mohamed Abdouh
- Subjects
0301 basic medicine ,p53 ,Amyloid ,Amyloid beta ,Induced Pluripotent Stem Cells ,tau Proteins ,macromolecular substances ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Alzheimer Disease ,medicine ,PSEN1 ,MAPT ,Dementia ,Humans ,Age of Onset ,Phosphorylation ,lcsh:QH301-705.5 ,Neurons ,Polycomb Repressive Complex 1 ,Progeria ,Glycogen Synthase Kinase 3 beta ,biology ,Lewy body ,Neurodegeneration ,Brain ,Peptide secretion ,medicine.disease ,BMI1 ,030104 developmental biology ,lcsh:Biology (General) ,sporadic ,biology.protein ,Cancer research ,GSK3b ,Tau ,Tumor Suppressor Protein p53 ,Alzheimer’s disease ,polycomb ,Frontotemporal dementia - Abstract
Late-onset sporadic Alzheimer's disease (AD) is the most prevalent form of dementia, but its origin remains poorly understood. The Bmi1/Ring1 protein complex maintains transcriptional repression of developmental genes through histone H2A mono-ubiquitination, and Bmi1 deficiency in mice results in growth retardation, progeria, and neurodegeneration. Here, we demonstrate that BMI1 is silenced in AD brains, but not in those with early-onset familial AD, frontotemporal dementia, or Lewy body dementia. BMI1 expression was also reduced in cortical neurons from AD patient-derived induced pluripotent stem cells but not in neurons overexpressing mutant APP and PSEN1. BMI1 knockout in human post-mitotic neurons resulted in amyloid beta peptide secretion and deposition, p-Tau accumulation, and neurodegeneration. Mechanistically, BMI1 was required to repress microtubule associated protein tau (MAPT) transcription and prevent GSK3beta and p53 stabilization, which otherwise resulted in neurodegeneration. Restoration of BMI1 activity through genetic or pharmaceutical approaches could represent a therapeutic strategy against AD.
- Published
- 2017
19. The type II transmembrane serine protease matriptase cleaves the amyloid precursor protein and reduces its processing to β-amyloid peptide
- Author
-
Christine Lavoie, François Béliveau, Denis Gris, Antoine Désilets, Anthony Flamier, Shaimaa Mahmoud, Gilbert Bernier, Erwan Lanchec, and Richard Leduc
- Subjects
0301 basic medicine ,Proteases ,Recombinant Fusion Proteins ,Prefrontal Cortex ,Nerve Tissue Proteins ,Biochemistry ,ST14 ,Gene Expression Regulation, Enzymologic ,Cell Line ,Substrate Specificity ,03 medical and health sciences ,Amyloid beta-Protein Precursor ,Young Adult ,mental disorders ,Amyloid precursor protein ,Cadaver ,Humans ,Matriptase ,RNA, Messenger ,Molecular Biology ,Aged ,Serine protease ,Neurons ,Amyloid beta-Peptides ,biology ,Chemistry ,Serine Endopeptidases ,P3 peptide ,Age Factors ,Brain ,Computational Biology ,Molecular Bases of Disease ,Cell Biology ,Peptide Fragments ,Biochemistry of Alzheimer's disease ,Cell biology ,030104 developmental biology ,Organ Specificity ,Mutation ,Proteolysis ,biology.protein ,Mutagenesis, Site-Directed ,Amyloid precursor protein secretase - Abstract
Recent studies have reported that many proteases, besides the canonical α-, β-, and γ-secretases, cleave the amyloid precursor protein (APP) and modulate β-amyloid (Aβ) peptide production. Moreover, specific APP isoforms contain Kunitz protease-inhibitory domains, which regulate the proteolytic activity of serine proteases. This prompted us to investigate the role of matriptase, a member of the type II transmembrane serine protease family, in APP processing. Using quantitative RT-PCR, we detected matriptase mRNA in several regions of the human brain with an enrichment in neurons. RNA sequencing data of human dorsolateral prefrontal cortex revealed relatively high levels of matriptase RNA in young individuals, whereas lower levels were detected in older individuals. We further demonstrate that matriptase and APP directly interact with each other and that matriptase cleaves APP at a specific arginine residue (Arg-102) both in vitro and in cells. Site-directed (Arg-to-Ala) mutagenesis of this cleavage site abolished matriptase-mediated APP processing. Moreover, we observed that a soluble, shed matriptase form cleaves endogenous APP in SH-SY5Y cells and that this cleavage significantly reduces APP processing to Aβ40. In summary, this study identifies matriptase as an APP-cleaving enzyme, an activity that could have important consequences for the abundance of Aβ and in Alzheimer's disease pathology.
- Published
- 2017
20. Brain Cancer Stem Cells: The Emerging Role of Chromatin in Glioma Biology
- Author
-
Gilbert Bernier
- Subjects
Cancer stem cell ,Glioma ,medicine ,Biology ,Stem cell ,medicine.disease ,Brain cancer ,Chromatin ,Cell biology - Published
- 2017
- Full Text
- View/download PDF
21. Brain Cancer Stem Cells: Current Status on Glioblastoma Multiforme
- Author
-
Mohamed Abdouh, Sabrina Facchino, and Gilbert Bernier
- Subjects
Cancer Research ,cancer stem cell ,medicine.medical_treatment ,Brain tumor ,Review ,Bioinformatics ,urologic and male genital diseases ,lcsh:RC254-282 ,glioblastoma multiforme ,astrocyte ,Cancer stem cell ,Radioresistance ,Glioma ,glioma ,Medicine ,CD133 ,polycomb, BMI1 ,astrocytoma ,business.industry ,urogenital system ,prominin ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Neural stem cell ,nervous system diseases ,Transplantation ,Radiation therapy ,radioresistance ,Oncology ,Cancer research ,Stem cell ,business - Abstract
Glioblastoma multiforme (GBM), an aggressive brain tumor of astrocytic/neural stem cell origin, represents one of the most incurable cancers. GBM tumors are highly heterogeneous. However, most tumors contain a subpopulation of cells that display neural stem cell characteristics in vitro and that can generate a new brain tumor upon transplantation in mice. Hence, previously identified molecular pathways regulating neural stem cell biology were found to represent the cornerstone of GBM stem cell self-renewal mechanism. GBM tumors are also notorious for their resistance to radiation therapy. Notably, GBM “cancer stem cells” were also found to be responsible for this radioresistance. Herein, we will analyze the data supporting or not the cancer stem cell model in GBM, overview the current knowledge regarding GBM stem cell self-renewal and radioresistance molecular mechanisms, and discuss the potential therapeutic application of these findings.
- Published
- 2011
22. Bmi1 Distinguishes Immature Retinal Progenitor/Stem Cells from the Main Progenitor Cell Population and Is Required for Normal Retinal Development
- Author
-
Mohamed Abdouh, Gilbert Bernier, Robert-Hugues Duparc, and Wassim Chatoo
- Subjects
Population ,macromolecular substances ,Biology ,Stem cell marker ,Retina ,Mice ,SOX2 ,Proto-Oncogene Proteins ,Neurosphere ,medicine ,Animals ,Progenitor cell ,education ,Polycomb Repressive Complex 1 ,education.field_of_study ,Reverse Transcriptase Polymerase Chain Reaction ,Stem Cells ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,Flow Cytometry ,Embryonic stem cell ,Cell biology ,Repressor Proteins ,medicine.anatomical_structure ,Immunology ,Molecular Medicine ,sense organs ,Stem cell ,Developmental Biology - Abstract
The developing mammalian retina is generated by the proliferation and differentiation of multipotent retinal progenitor cells (RPCs) giving rise to neuronal and glial lineages. Whether an immature progenitor/stem cell subpopulation is present in the developing mammalian retina remains undefined. Deficiency in the polycomb group gene Bmi1 results in reduced proliferation and postnatal depletion of neural and hematopoietic stem cells. Here, we show that Bmi1 is required for the self-renewal of most immature RPCs and for postnatal retinal development. In the embryo, Bmi1 is highly enriched in a rare stage-specific embryonic antigen-1-positive RPC subpopulation expressing the stem cell markers Sox2, Lhx2, and Musashi. Gain-of-function experiments revealed that Bmi1 overexpression could convert RPCs having limited proliferation capacity into RPCs showing extensive proliferation and multiple differentiation capacities over time. At all developmental stages analyzed using the neurosphere assay, Bmi1 deficiency resulted in reduced proliferation and self-renewal of most immature RPCs. Reduced RPCs proliferation was also observed in the peripheral retina of Bmi1−/− fetus and newborn mice. The biological impact of these developmental anomalies was revealed by the reduced retinal diameter of Bmi1-deficient pups. P19Arf and p16Ink4a were upregulated in vivo and in vitro and coinactivation of p53, which lies downstream of p19Arf, partially restored Bmi1-deficient RPCs self-renewal phenotype. Bmi1 thus distinguishes immature RPCs from the main RPC population and is required for normal retinal development.
- Published
- 2010
- Full Text
- View/download PDF
23. AB023. Characterization of a novel anti-angiogenic protein for the treatment of exudative age-related macular degeneration
- Author
-
Natalija Popovic, Bruno Larrivée, Anthony Flamier, Erika Hooker, Gilbert Bernier, and Andrea Barabino
- Subjects
Ophthalmology ,Pathology ,medicine.medical_specialty ,business.industry ,Anti angiogenic ,medicine ,Exudative age-related macular degeneration ,business - Published
- 2018
- Full Text
- View/download PDF
24. Retinal development anomalies and cone photoreceptors degeneration upon Bmi1 deficiency
- Author
-
Gilbert Bernier, Shufeng Zhou, Marc Hébert, Andrea Barabino, Roy Hanna, Joëlle Lavoie, Vicky Plamondon, Mohamed Abdouh, Noboru Motoyama, Anthony Flamier, and Wassim Chatoo
- Subjects
0301 basic medicine ,Retina ,Cell type ,genetic structures ,Heterochromatin ,Necroptosis ,Intrinsically photosensitive retinal ganglion cells ,Retinal ,macromolecular substances ,Biology ,Bioinformatics ,Embryonic stem cell ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,medicine ,sense organs ,Molecular Biology ,Developmental Biology ,Visual phototransduction - Abstract
Retinal development occurs through the sequential but overlapping generation of six types of neuronal cells and one glial cell type. Of these, rod and cone photoreceptors represent the functional unit of light detection and phototransduction and are frequently affected in retinal degenerative diseases. During mouse development, the Polycomb group protein Bmi1 is expressed in immature retinal progenitors and differentiated retinal neurons, including cones. We show here that Bmi1 is required to prevent post natal degeneration of cone photoreceptors and bipolar neurons and that inactivation of Chk2 or p53 could improve but not overcome cone degeneration in Bmi1(-/-) mice. The retinal phenotype of Bmi1(-/-) mice was also characterized by loss of heterochromatin, activation of tandem repeats, oxidative stress and Rip3-associated necroptosis. In the human retina, BMI1 was preferentially expressed in cones at heterochromatic foci. BMI1 inactivation in human embryonic stem cells was compatible with retinal induction but impaired cone terminal differentiation. Despite this developmental arrest, BMI1-deficient cones recapitulated several anomalies observed in Bmi1(-/-) photoreceptors, such as loss of heterochromatin, activation of tandem repeats and induction of p53, revealing partly conserved biological functions between mouse and man.
- Published
- 2016
- Full Text
- View/download PDF
25. The Polycomb Repressive Complex 1 Protein BMI1 Is Required for Constitutive Heterochromatin Formation and Silencing in Mammalian Somatic Cells
- Author
-
Jida El Hajjar, Gilbert Bernier, Roy Hanna, Mohamed Abdouh, and Anthony Flamier
- Subjects
0301 basic medicine ,Euchromatin ,Heterochromatin ,Nuclear Envelope ,macromolecular substances ,Biochemistry ,Histones ,03 medical and health sciences ,Mice ,Non-histone protein ,Neural Stem Cells ,Proto-Oncogene Proteins ,Constitutive heterochromatin ,Animals ,Humans ,Gene Silencing ,Molecular Biology ,Pericentric heterochromatin ,Repetitive Sequences, Nucleic Acid ,Genetics ,Cerebral Cortex ,Mammals ,Neurons ,Polycomb Repressive Complex 1 ,biology ,BRCA1 Protein ,Ubiquitin ,EZH2 ,fungi ,Cell Biology ,030104 developmental biology ,Histone ,Gene Knockdown Techniques ,biology.protein ,Heterochromatin protein 1 - Abstract
The polycomb repressive complex 1 (PRC1), containing the core BMI1 and RING1A/B proteins, mono-ubiquitinylates histone H2A (H2A(ub)) and is associated with silenced developmental genes at facultative heterochromatin. It is, however, assumed that the PRC1 is excluded from constitutive heterochromatin in somatic cells based on work performed on mouse embryonic stem cells and oocytes. We show here that BMI1 is required for constitutive heterochromatin formation and silencing in human and mouse somatic cells. BMI1 was highly enriched at intergenic and pericentric heterochromatin, co-immunoprecipitated with the architectural heterochromatin proteins HP1, DEK1, and ATRx, and was required for their localization. In contrast, BRCA1 localization was BMI1-independent and partially redundant with that of BMI1 for H2A(ub) deposition, constitutive heterochromatin formation, and silencing. These observations suggest a dynamic and developmentally regulated model of PRC1 occupancy at constitutive heterochromatin, and where BMI1 function in somatic cells is to stabilize the repetitive genome.
- Published
- 2015
26. Loss of Bmi1 causes anomalies in retinal development and degeneration of cone photoreceptors
- Author
-
Andrea, Barabino, Vicky, Plamondon, Mohamed, Abdouh, Wassim, Chatoo, Anthony, Flamier, Roy, Hanna, Shufeng, Zhou, Noboru, Motoyama, Marc, Hébert, Joëlle, Lavoie, and Gilbert, Bernier
- Subjects
Mice, Knockout ,Polycomb Repressive Complex 1 ,Retina ,Cell Line ,Mice, Inbred C57BL ,Checkpoint Kinase 2 ,Mice ,Necrosis ,Oxidative Stress ,Retinal Rod Photoreceptor Cells ,Heterochromatin ,Proto-Oncogene Proteins ,Receptor-Interacting Protein Serine-Threonine Kinases ,Retinal Cone Photoreceptor Cells ,Animals ,Humans ,Tumor Suppressor Protein p53 ,Embryonic Stem Cells - Abstract
Retinal development occurs through the sequential but overlapping generation of six types of neuronal cells and one glial cell type. Of these, rod and cone photoreceptors represent the functional unit of light detection and phototransduction and are frequently affected in retinal degenerative diseases. During mouse development, the Polycomb group protein Bmi1 is expressed in immature retinal progenitors and differentiated retinal neurons, including cones. We show here that Bmi1 is required to prevent post natal degeneration of cone photoreceptors and bipolar neurons and that inactivation of Chk2 or p53 could improve but not overcome cone degeneration in Bmi1(-/-) mice. The retinal phenotype of Bmi1(-/-) mice was also characterized by loss of heterochromatin, activation of tandem repeats, oxidative stress and Rip3-associated necroptosis. In the human retina, BMI1 was preferentially expressed in cones at heterochromatic foci. BMI1 inactivation in human embryonic stem cells was compatible with retinal induction but impaired cone terminal differentiation. Despite this developmental arrest, BMI1-deficient cones recapitulated several anomalies observed in Bmi1(-/-) photoreceptors, such as loss of heterochromatin, activation of tandem repeats and induction of p53, revealing partly conserved biological functions between mouse and man.
- Published
- 2015
27. In vivo reactivation of a quiescent cell population located in the ocular ciliary body of adult mammals
- Author
-
Gilbert Bernier and Mohamed Abdouh
- Subjects
PAX6 Transcription Factor ,Cell division ,Iris ,Nerve Tissue Proteins ,Biology ,Retina ,Injections ,Nestin ,Cellular and Molecular Neuroscience ,Intermediate Filament Proteins ,In Situ Nick-End Labeling ,medicine ,Animals ,Paired Box Transcription Factors ,Cyclin D1 ,Rats, Wistar ,Progenitor cell ,Eye Proteins ,Growth Substances ,Homeodomain Proteins ,Neurons ,Cell growth ,Stem Cells ,Cell Cycle ,Ciliary Body ,Neurogenesis ,Cell Differentiation ,Epithelial Cells ,Immunohistochemistry ,Embryonic stem cell ,Sensory Systems ,Rats ,Cell biology ,Repressor Proteins ,Ophthalmology ,Ki-67 Antigen ,medicine.anatomical_structure ,Stem cell ,Biomarkers ,Cell Division ,Transcription Factors ,Adult stem cell - Abstract
Rare quiescent cells with stem cell characteristics have been isolated from the ocular ciliary body (CB) of adult mammals. In vitro, adult retinal stem cells were reported to generate sphere colonies containing multipotent retinal progenitor cells. Whether proliferation of this stem cell population can be stimulated in vivo in order to generate new retinal cells is an important issue. Herein we report on the in vivo reactivation of a quiescent cell population present in the CB upon growth factors (GF) stimulation. GF stimulation resulted in the re-acquisition of embryonic characteristics (Nestin) and expression of the cell cycle entry markers CyclinD1 and Ki67 by a subset of CB epithelial cells. This inductive effect was not observed in the neural retina. GF-activated CB epithelial cells co-express the retinal progenitor homeodomain transcription factors Pax6 and Chx10. Serial GF injections led to do novo proliferation of clusters of cells in the CB, in a dose-dependent manner, as revealed by bromodeoxyuridine (BrdU) incorporation. Analysis of cells' BrdU content within individual clusters suggests a mode of cell division that is predominantly asymmetric. Cell proliferation was not induced by CB or retinal damage, as indicated by the absence of TUNEL-labeled cells. Newly produced cells did not migrate into the retina nor did they differentiate into retinal neurons. This study demonstrates that proliferation of a quiescent cell population with retinal stem/progenitor cell characteristics can be reactivated in vivo upon GF injections and suggests that, in adult mammals, the CB is a non-permissive environment for cell migration and neurogenesis.
- Published
- 2006
- Full Text
- View/download PDF
28. Genetic alterations at the Bpag1 locus in dt mice and their impact on transcript expression
- Author
-
Madeline Pool, Rashmi Kothary, Kevin G. Young, Gilbert Bernier, and Céline Larivière
- Subjects
Genetics ,Dystonin ,Point mutation ,Mutant ,Dystonia Musculorum Deformans ,Brain ,Chromosome Mapping ,Mice, Transgenic ,Nerve Tissue Proteins ,Locus (genetics) ,Biology ,Molecular biology ,Cytoskeletal Proteins ,Mice ,Exon ,Mutation ,Animals ,Coding region ,Allele ,Carrier Proteins ,Muscle, Skeletal ,Gene ,Alleles - Abstract
The dystonin/Bpag1 gene encodes several tissue-specific alternatively spliced transcripts that encode cytoskeletal binding proteins. These various isoforms are necessary for maintaining the structural integrity of epithelial, neural, and muscle tissues. Mutations in the dystonin/Bpag1 gene cause dystonia musculorum (dt), a hereditary neuropathy of the mouse characterized by the progressive degeneration of sensory neurons. Several dt mutant alleles exist, most of which have arisen through spontaneous mutations. In this article we demonstrate that the dt locus encodes 107 exons spanning 400 kb. The high frequency of occurrence of spontaneous dt mutants may therefore be a result of the large size of the gene. Analysis of genomic DNA from several dt spontaneous mutant alleles, dt 24J , dt 27J , dt Alb , and dt Frk , shows a deletion of the central portion of the gene in dt Alb but no large rearrangements or deletions in the other alleles. These other alleles likely have small deletions or rearrangements, or point mutations. To determine the impact of the known and unknown mutations on transcript levels, RT-PCR was performed to detect various coding regions of the dystonin/Bpag1 transcripts in brain and muscle from multiple dt alleles: dt Tg4 , dt Alb , dt 24J , dt 27J , and dt Frk . With the exception of dt Frk , reduced transcript levels were observed for all alleles tested. Such alterations likely result in reduced or absent dystonin/Bpag1 protein levels. Thus, distinct genetic defects lead to a common outcome of reduced transcript expression causing the same phenotype in multiple dt alleles.
- Published
- 2005
- Full Text
- View/download PDF
29. Isolation and characterization of a downstream target ofPax6in the mammalian retinal primordium
- Author
-
Peter Gruss, Gilbert Bernier, Bernhard G. Herrmann, Lorenz Neidhardt, and Wolfgang Vukovich
- Subjects
DNA, Complementary ,PAX6 Transcription Factor ,Molecular Sequence Data ,Biology ,Retina ,Mice ,Animals ,Humans ,Paired Box Transcription Factors ,Amino Acid Sequence ,Eye Proteins ,Molecular Biology ,Transcription factor ,In Situ Hybridization ,Homeodomain Proteins ,Eye morphogenesis ,Base Sequence ,Sequence Homology, Amino Acid ,Neuroectoderm ,Calcium-Binding Proteins ,Genes, Homeobox ,Gene Expression Regulation, Developmental ,Optic vesicle ,Molecular biology ,eye diseases ,Repressor Proteins ,Multigene Family ,Homeobox ,Ectopic expression ,sense organs ,PAX6 ,Signal transduction ,Signal Transduction ,Developmental Biology - Abstract
The transcription factor Pax6 is required for eye morphogenesis in humans, mice and insects, and can induce ectopic eye formation in vertebrate and invertebrate organisms. Although the role of Pax6 has intensively been studied, only a limited number of genes have been identified that depend on Pax6 activity for their expression in the mammalian visual system. Using a large-scale in situ hybridization screen approach, we have identified a novel gene expressed in the mouse optic vesicle. This gene, Necab , encodes a putative cytoplasmic Ca 2+ -binding protein and coincides with Pax6 expression pattern in the neural ectoderm of the optic vesicle and in the forebrain pretectum. Remarkably, Necab expression is absent in both structures in Pax6 mutant embryos. By contrast, the optic vesicle-expressed homeobox genes Rx , Six3 , Otx2 and Lhx2 do not exhibit an altered expression pattern. Using gain-of-function experiments, we show that Pax6 can induce ectopic expression of Necab , suggesting that Necab is a direct or indirect transcriptional target of Pax6 . In addition, we have found that Necab misexpression can induce ectopic expression of the homeobox gene Chx10 , a transcription factor implicated in retina development. Taken together, our results provide evidence that Necab is genetically downstream of Pax6 and that it is a part of a signal transduction pathway in retina development.
- Published
- 2001
- Full Text
- View/download PDF
30. Acf7 (MACF) is an actin and microtubule linker protein whose expression predominates in neural, muscle, and lung development
- Author
-
Rashmi Kothary, Jessica Alföldi, Gilbert Bernier, Yves De Repentigny, Madeline Pool, and Michael Kilcup
- Subjects
Plakin ,Neural fold ,Microtubule ,biology.protein ,Actin-binding protein ,Plectin ,Biology ,Cytoskeleton ,Dystonin ,Molecular biology ,Actin ,Developmental Biology ,Cell biology - Abstract
Several proteins belonging to the plakin family of cytoskeletal linker proteins have recently been identified, including dystonin/Bpag1 and plectin. These proteins are unique in their abilities to form bridges between different cytoskeletal elements through specialized modular domains. We have previously reported the cloning and partial characterization of Acf7, a novel member of the plakin family. More recently, the full-length cDNA for mouse Acf7 has been reported. Acf7 has a hybrid composition, with extended homology to dystonin/Bpag1 and plectin in the N-terminal half, and to dystrophin in the central and C-terminal half. Recent studies have demonstrated that Acf7 has functional actin and microtubule binding domains. Here, we describe the developmental expression profile for mouse Acf7. RNA in situ hybridization experiments revealed Acf7 transcripts in the dermomyotome and neural fold of day 8.5 mouse embryos. Later in development, Acf7 expression was predominant in neural and muscle tissues and was strongly up-regulated just before birth in type II alveolar cells of the lung. Altogether, our results suggest that Acf7 functions as a versatile cytoskeletal linker protein and plays an important role in neural, muscle, and lung development.
- Published
- 2000
- Full Text
- View/download PDF
31. Six6 (Optx2) is a novel murine Six3-related homeobox gene that demarcates the presumptive pituitary/hypothalamic axis and the ventral optic stalk
- Author
-
Gilbert Bernier, Dominique Jean, and Peter Gruss
- Subjects
Embryology ,genetic structures ,PAX6 Transcription Factor ,Molecular Sequence Data ,Hypothalamus ,Nerve Tissue Proteins ,Biology ,Retina ,Mice ,medicine ,Animals ,Paired Box Transcription Factors ,Optic stalk ,Primordium ,Amino Acid Sequence ,Cloning, Molecular ,Eye Proteins ,Phylogeny ,Visual Cortex ,Homeodomain Proteins ,Base Sequence ,Sequence Homology, Amino Acid ,Anatomy ,eye diseases ,Rathke's pouch ,Mice, Mutant Strains ,DNA-Binding Proteins ,Repressor Proteins ,medicine.anatomical_structure ,Optic Chiasm ,Pituitary Gland ,Forebrain ,Mutation ,Optic chiasma ,Trans-Activators ,Homeobox ,sense organs ,PAX6 ,Developmental Biology - Abstract
We report on the isolation of a murine homeobox-containing gene, Six6 (Optx2), that shows extended identity in its coding region with Six3, the only member of the mammalian Six gene family known to be expressed in the optic primordium. Phylogenetic analysis demonstrates that Six6 and Six3 belong to a separate group of homeobox-genes that are closely related to the recently identified Drosophila optix. Earliest Six6 expression was detected in the floor of the diencephalic portion of the primitive forebrain, a region predicted to give rise to the neurohypophysis and to the hypothalamus. Later on, Six6 mRNA was found in the primordial tissues giving rise to the mature pituitary: the Rathke’s pouch and the infundibular recess. In the optic primordium, Six6 demarcates the presumptive ventral optic stalk and the ventral portion of the future neural retina. In the developing eye, Six6 expression was detected in the neural retina, the optic chiasma and optic stalk, but not in the lens. When compared to Six6, Six3 expression pattern was highly similar, but with a generally broader transcripts distribution in the brain and in the visual system. We finally show that Six6 does not require Pax6 for its expression in the optic primordium, suggesting that Six6 acts on a parallel and/or independent pathway with Pax6 in the genetic cascade governing early development of the eye. q 1999 Elsevier Science Ireland Ltd. All rights reserved.
- Published
- 1999
- Full Text
- View/download PDF
32. ASPP1/2 regulate p53-dependent death of retinal ganglion cells through PUMA and Fas/CD95 activation in vivo
- Author
-
Ariel M. Wilson, Barbara Morquette, Gilbert Bernier, E. Feinstein, Adriana Di Polo, Mohamed Abdouh, Nicolas Unsain, and Philip A. Barker
- Subjects
Retinal Ganglion Cells ,Transcriptional Activation ,Down-Regulation ,Retinal ganglion ,Neuroprotection ,Rats, Sprague-Dawley ,Mice ,Downregulation and upregulation ,Puma ,Gene expression ,Animals ,fas Receptor ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Mice, Knockout ,Gene knockdown ,biology ,Cell Death ,General Neuroscience ,Tumor Suppressor Proteins ,Articles ,biology.organism_classification ,Fas receptor ,Axons ,Cell biology ,Rats ,Female ,sense organs ,Tumor Suppressor Protein p53 ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
The transcription factor p53 mediates neuronal death in a variety of stress-related and neurodegenerative conditions. The proapoptotic activity of p53 is tightly regulated by the apoptosis-stimulating proteins of p53 (ASPP) family members: ASPP1 and ASPP2. However, whether ASPP1/2 play a role in the regulation of p53-dependent neuronal death in the CNS is currently unknown. To address this, we asked whether ASPP1/2 contribute to the death of retinal ganglion cells (RGCs) usingin vivomodels of acute optic nerve damage in mice and rats. Here, we show that p53 is activated in RGCs soon after injury and that axotomy-induced RGC death is attenuated in p53 heterozygote and null mice. We demonstrate that ASPP1/2 proteins are abundantly expressed by injured RGCs, and that short interfering (si)RNA-based ASPP1 or ASPP2 knockdown promotes robust RGC survival. Comparative gene expression analysis revealed that siASPP-mediated downregulation of p53-upregulated-modulator-of-apoptosis (PUMA), Fas/CD95, and Noxa depends on p53 transcriptional activity. Furthermore, siRNA against PUMA or Fas/CD95 confers neuroprotection, demonstrating a functional role for these p53 targets in RGC death. Our study demonstrates a novel role for ASPP1 and ASPP2 in the death of RGCs and provides evidence that blockade of the ASPP–p53 pathway is beneficial for central neuron survival after axonal injury.
- Published
- 2013
33. Glioblastoma Multiforme: Role of Polycomb Group Proteins
- Author
-
Sabrina Facchino, Mohamed Abdouh, and Gilbert Bernier
- Subjects
education.field_of_study ,Population ,Brain tumor ,macromolecular substances ,Tumor initiation ,Biology ,Bioinformatics ,medicine.disease ,Stem cell marker ,Cancer stem cell ,BMI1 ,Glioma ,medicine ,Cancer research ,Stem cell ,education - Abstract
Glioblastoma multiforme (GBM) is the most common and lethal primary brain tumor found in adult. Even with all the advances made in the field of cancer therapy for the last decade, the prognosis has not significantly changed, with a median survival of less than 1 year. Therefore, a better understanding of GBM biology is needed. Unraveling the basic mechanisms responsible for the initiation and progression of this tumor may open new fields for the development of efficient therapeutic strategies. Proteins of the polycomb (PcG) group family generally operate as transcription repressors and BMI1 is one of the best-characterized member. The implication of BMI1 in normal and cancerous stem cells survival, self-renewal and maintenance has been widely investigated. In most GBM, a relatively rare cell population, characterized as the tumor initiating cell population, expresses the stem cell marker CD133. In experimental systems, CD133-positive GBM cells are responsible for tumor initiation, maintenance, progression and resistance to chemo/radiotherapy. CD133-positive GBM “stem cells” thus possibly represent a valuable and specific cellular target to eradicate the tumor. Here, we report on the implication of BMI1 in GBM stem cell self-renewal and how BMI1 is indispensable for GBM tumor establishment and progression in a xenograft mouse model.
- Published
- 2011
- Full Text
- View/download PDF
34. p53 pro-oxidant activity in the central nervous system: implication in aging and neurodegenerative diseases
- Author
-
Mohamed Abdouh, Gilbert Bernier, and Wassim Chatoo
- Subjects
Senescence ,Aging ,Cell cycle checkpoint ,Physiology ,DNA repair ,DNA damage ,Clinical Biochemistry ,Central nervous system ,Cellular homeostasis ,Context (language use) ,Apoptosis ,Biology ,medicine.disease_cause ,Biochemistry ,Mice ,medicine ,Animals ,Humans ,Molecular Biology ,General Environmental Science ,Neurons ,Brain ,Neurodegenerative Diseases ,Cell Biology ,Cell biology ,medicine.anatomical_structure ,General Earth and Planetary Sciences ,Tumor Suppressor Protein p53 ,Oxidation-Reduction ,Oxidative stress - Abstract
Recent advances in delineating the biological functions of p53 had shed the light on its key role in the multifacets of cellular homeostasis. After its activation, via DNA damage, oxidative stress, or aberrant expression of oncogenes, p53 transduces its classical effect through several mechanisms comprising activation of the DNA repair machinery, cell cycle arrest, and initiation of apoptosis or senescence. In the mammalian brain, p53 plays critical functions in normal development, tumor suppression, neurodegenerative diseases, and aging. Herein, we focus on the constitutive pro-oxidant activity of p53 in neurons and discuss the potential implication of this finding in the context of neurodegenerative diseases and normal brain aging.
- Published
- 2010
35. BMI1 confers radioresistance to normal and cancerous neural stem cells through recruitment of the DNA damage response machinery
- Author
-
Sabrina Facchino, Mohamed Abdouh, Wassim Chatoo, and Gilbert Bernier
- Subjects
DNA damage ,Green Fluorescent Proteins ,Polycomb-Group Proteins ,macromolecular substances ,Protein Serine-Threonine Kinases ,Transfection ,Radiation Tolerance ,Fetus ,Histone H1 ,Antigens, CD ,Tandem Mass Spectrometry ,Radioresistance ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Humans ,Immunoprecipitation ,DNA Breaks, Double-Stranded ,AC133 Antigen ,Phosphorylation ,Embryonic Stem Cells ,Glycoproteins ,Polycomb Repressive Complex 1 ,biology ,Brain Neoplasms ,General Neuroscience ,Nuclear Proteins ,Articles ,Flow Cytometry ,Chromatin ,Non-homologous end joining ,DNA-Binding Proteins ,Repressor Proteins ,Checkpoint Kinase 2 ,Histone ,Cell Transformation, Neoplastic ,BMI1 ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Comet Assay ,Stem cell ,Glioblastoma ,Peptides ,Reactive Oxygen Species ,Chromatography, Liquid ,DNA Damage - Abstract
Glioblastoma multiforme (GBM) is an aggressive brain tumor that is resistant to all known therapies. Within these tumors, a CD133-positive cancer-initiating neural stem cell (NSC) population was shown to be resistant to gamma radiation through preferential activation of the DNA double-strand break (DSB) response machinery, including the ataxia-telangiectasia-mutated (ATM) kinase. The polycomb group protein BMI1 is enriched in CD133-positive GBM cells and required for their self-renewal in anINK4A/ARF-independent manner through transcriptional repression of alternate tumor suppressor pathways. We report here that BMI1 copurifies with DNA DSB response and nonhomologous end joining (NHEJ) repair proteins in GBM cells. BMI1 was enriched at the chromatin after irradiation and colocalized and copurified with ATM and the histone γH2AX. BMI1 also preferentially copurified with NHEJ proteins DNA-PK, PARP-1, hnRNP U, and histone H1 in CD133-positive GBM cells. BMI1 deficiency in GBM cells severely impaired DNA DSB response, resulting in increased sensitivity to radiation. In turn, BMI1 overexpression in normal NSCs enhanced ATM recruitment to the chromatin, the rate of γH2AX foci resolution, and resistance to radiation. BMI1 thus displays a previously uncharacterized function in controlling DNA DSB response and repair. Pharmacological inhibition of BMI1 combined with radiation therapy may provide an effective mean to target GBM stem cells.
- Published
- 2010
36. BMI1 sustains human glioblastoma multiforme stem cell renewal
- Author
-
Gilbert Bernier, Sabrina Facchino, Wassim Chatoo, José Ferreira, Mohamed Abdouh, and Vijayabalan Balasingam
- Subjects
Chromatin Immunoprecipitation ,Cellular differentiation ,Apoptosis ,macromolecular substances ,Mice, SCID ,Biology ,Mice ,Antigens, CD ,Mice, Inbred NOD ,Cell Line, Tumor ,Proto-Oncogene Proteins ,Gene silencing ,Animals ,Humans ,Enhancer of Zeste Homolog 2 Protein ,AC133 Antigen ,Gene Silencing ,RNA, Small Interfering ,Cells, Cultured ,Tumor Stem Cell Assay ,Glycoproteins ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Polycomb Repressive Complex 1 ,Analysis of Variance ,Oncogene ,Brain Neoplasms ,General Neuroscience ,Polycomb Repressive Complex 2 ,Nuclear Proteins ,Cell Differentiation ,Articles ,Flow Cytometry ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,Disease Models, Animal ,Cell Transformation, Neoplastic ,BMI1 ,Cell Transdifferentiation ,Cancer research ,biology.protein ,Neoplastic Stem Cells ,Stem cell ,PRC2 ,Glioblastoma ,Peptides ,Transcription Factors - Abstract
Glioblastoma multiforme (GBM) is one of the most common and aggressive types of brain tumors. In GBM, a subpopulation of CD133-positive cancer initiating cells displays stem cell characteristics. The Polycomb group (PcG) and oncogene BMI1 is part of the Polycomb repressive complex 1 (PRC1) that regulates gene expression by modifying chromatin organization. Here we show that BMI1 is expressed in human GBM tumors and highly enriched in CD133-positive cells. Stable BMI1 knockdown using short hairpin RNA-expressing lentiviruses resulted in inhibition of clonogenic potential in vitro and of brain tumor formation in vivo. Cell biology studies support the notion that BMI1 prevents CD133-positive cell apoptosis and/or differentiation into neurons and astrocytes, depending on the cellular context. Gene expression analyses suggest that BMI1 represses alternate tumor suppressor pathways that attempt to compensate for INK4A/ARF/P53 deletion and PI(3)K/AKT hyperactivity. Inhibition of EZH2, the main component of the PRC2, also impaired GBM tumor growth. Our results reveal that PcG proteins are involved in GBM tumor growth and required to sustain cancer initiating stem cell renewal.
- Published
- 2009
37. The Polycomb group gene Bmi1 regulates antioxidant defenses in neurons by repressing p53 pro-oxidant activity
- Author
-
Mohamed Abdouh, Wassim Chatoo, Jocelyn David, Gilbert Bernier, José Ferreira, Francis Rodier, and Marie-Pier Champagne
- Subjects
Antioxidant ,PAX6 Transcription Factor ,medicine.medical_treatment ,Apoptosis ,Lipid peroxidation ,Histones ,chemistry.chemical_compound ,Mice ,Aging brain ,Paired Box Transcription Factors ,Cells, Cultured ,Regulation of gene expression ,chemistry.chemical_classification ,Cerebral Cortex ,Mice, Knockout ,Neurons ,Polycomb Repressive Complex 1 ,General Neuroscience ,Age Factors ,Nuclear Proteins ,Cell biology ,Microtubule-Associated Proteins ,Cell Survival ,Neurotoxins ,macromolecular substances ,Biology ,Article ,Downregulation and upregulation ,Proto-Oncogene Proteins ,Glial Fibrillary Acidic Protein ,medicine ,Animals ,Eye Proteins ,Cell Proliferation ,Homeodomain Proteins ,Reactive oxygen species ,Analysis of Variance ,Hydrogen Peroxide ,Peroxiredoxins ,Pro-oxidant ,Embryo, Mammalian ,beta-Galactosidase ,Molecular biology ,Mice, Inbred C57BL ,Repressor Proteins ,chemistry ,Gene Expression Regulation ,BMI1 ,Phosphopyruvate Hydratase ,Lipid Peroxidation ,Tumor Suppressor Protein p53 ,Reactive Oxygen Species - Abstract
Aging may be determined by a genetic program and/or by the accumulation rate of molecular damages. Reactive oxygen species (ROS) generated by the mitochondrial metabolism have been postulated to be the central source of molecular damages and imbalance between levels of intracellular ROS and antioxidant defenses is a characteristic of the aging brain. How aging modifies free radicals concentrations and increases the risk to develop most neurodegenerative diseases is poorly understood, however. Here we show that the Polycomb group and oncogene Bmi1 is required in neurons to suppress apoptosis and the induction of a premature aging-like program characterized by reduced antioxidant defenses. Before weaning,Bmi1−/−mice display a progeroid-like ocular and brain phenotype, whileBmi1+/−mice, although apparently normal, have reduced lifespan. Bmi1 deficiency in neurons results in increased p19Arf/p53 levels, abnormally high ROS concentrations, and hypersensitivity to neurotoxic agents. MostBmi1functions on neurons' oxidative metabolism are genetically linked to repression ofp53pro-oxidant activity, which also operates in physiological conditions. InBmi1−/−neurons, p53 and corepressors accumulate at antioxidant gene promoters, correlating with a repressed chromatin state and antioxidant gene downregulation. These findings provide a molecular mechanism explaining howBmi1regulates free radical concentrations and reveal the biological impact ofBmi1deficiency on neuronal survival and aging.
- Published
- 2009
38. Pax6 controls the proliferation rate of neuroepithelial progenitors from the mouse optic vesicle
- Author
-
Jocelyn David, Gilbert Bernier, Mohamed Abdouh, Nicolas Tétreault, Mireille Lépine, and Robert-Hugues Duparc
- Subjects
PAX6 Transcription Factor ,Neuroepithelial Cells ,Cell Separation ,Eye ,Mice ,0302 clinical medicine ,Paired Box Transcription Factors ,Cells, Cultured ,Cyclin-Dependent Kinase Inhibitor Proteins ,0303 health sciences ,education.field_of_study ,Stem Cells ,Neurogenesis ,Gene Expression Regulation, Developmental ,Optic vesicle ,Cyclin-Dependent Kinases ,Neuroepithelial cell ,medicine.anatomical_structure ,Population ,Biology ,Cell cycle ,Retina ,Homeobox transcription factor ,03 medical and health sciences ,Ectoderm ,medicine ,Animals ,Progenitor cell ,education ,Eye Proteins ,Molecular Biology ,030304 developmental biology ,Progenitor ,Cell Proliferation ,Homeodomain Proteins ,Lentivirus ,Cell Biology ,Molecular biology ,eye diseases ,Pax6 ,Neuroepithelial progenitors ,Repressor Proteins ,Neurosphere assay ,Mutation ,PAX6 ,sense organs ,030217 neurology & neurosurgery ,Biomarkers ,Developmental Biology - Abstract
In vertebrates, a limited number of homeobox-containing transcription factors are expressed in the optic vesicle primordium and are required and sufficient for eye formation. At present, little is known about the distinct functions of these factors in optic vesicle growth and on the nature of the main neuroepithelial (NE) progenitor population present in this organ. We have characterized a multipotent cell population present in the mouse optic vesicle that shows extensive proliferation potential and which expresses NE progenitor and retinal markers in vitro. In Pax6 mutant embryos, which form an optic vesicle, we found that the number of resident NE progenitors was greater than normal. In vitro, Pax6-null NE progenitors overproliferate and display reduced p16(Ink4a), p19(Arf), p27(kip1), p57(kip2), and p21(cip1) expression. Pax6 overexpression repressed cellular proliferation and secondary colonies formation, supporting the hypothesis that Pax6 acts cell-autonomously on NE progenitors cell cycle. Notably, these in vitro data correlated with aberrant numbers of mitosis observed in the optic vesicle of early stage Pax6 mutants, with Pax6 association with the chromatin upstream of p27(kip1) promoter region, and with reduced expression levels of p27(kip1), p57(kip2), and p21(cip1) in the primitive forebrain of Pax6 mutants. Taken together, our results suggest that, prior to retinal progenitor cell identity and neurogenesis, Pax6 is required to regulate the proliferation rate of NE progenitors present in the mouse optic vesicle.
- Published
- 2006
39. Identification of genes expressed in retinal progenitor/stem cell colonies isolated from the ocular ciliary body of adult mice
- Author
-
Julie Lord-Grignon, Mohamed Abdouh, and Gilbert Bernier
- Subjects
PAX6 Transcription Factor ,Mice, Transgenic ,Nerve Tissue Proteins ,In situ hybridization ,Biology ,Retina ,chemistry.chemical_compound ,Mice ,Ciliary body ,Genetics ,medicine ,Animals ,Paired Box Transcription Factors ,Eye Proteins ,Molecular Biology ,Homeodomain Proteins ,Microphthalmia-Associated Transcription Factor ,Eye morphogenesis ,cDNA library ,Stem Cells ,Ciliary Body ,HMGA2 Protein ,Gene Expression Regulation, Developmental ,Membrane Proteins ,Retinal ,Embryo, Mammalian ,Molecular biology ,Embryonic stem cell ,Mice, Inbred C57BL ,Repressor Proteins ,medicine.anatomical_structure ,chemistry ,Trans-Activators ,PAX6 ,Stem cell ,Developmental Biology - Abstract
Rare pigmented cells showing retinal stem cell characteristics have been identified in the ocular ciliary body (CB) of adult mammals. In vitro, these cells were reported to clonally proliferate and generate pigmented sphere colonies (PSC) containing multipotent retinal progenitor-like cells. Because these cells may have important clinical applications and because their embryonic origin is unclear, we have analyzed their local environment and gene expression profile. We found that transcription factors Pax6, Six3, and Rx, all involved in early eye morphogenesis, were expressed in the CB of adult mice. By sequencing a PSC cDNA library, we found that PSC expressed at high levels transcripts involved in the control of redox metabolism and cellular proliferation. PSC also expressed the retinal transcription factor Six6, which expression was not detected in the CB epithelium. By in situ hybridization screen, we found that Palmdelphin (Palm), Hmga2, and a novel transcript were expressed in the central nervous system of early embryos. Palm expression delineated the pigmented epithelium of the future CB and the developing myotome. Hmga2 was expressed in the ventricular zone of the telencephalon, the developing retinal ciliary margin and lens. Several genes expressed in PSC were also expressed in the nasal anlagen. Taken together, our study reveals that PSC isolated from the ocular CB express genes involved in the control of embryonic development, retinal identity, redox metabolism, and cellular proliferation.
- Published
- 2006
40. Characterization of new transcripts enriched in the mouse retina and identification of candidate retinal disease genes
- Author
-
Alan J. Mears, Julie Lord-Grignon, Nicolas Tétreault, Gilbert Bernier, and Anand Swaroop
- Subjects
genetic structures ,Sequence Homology ,Gene mutation ,Biology ,Retina ,chemistry.chemical_compound ,Mice ,Retinal Diseases ,Retinitis pigmentosa ,medicine ,Animals ,RNA, Messenger ,Eye Proteins ,Pigment Epithelium of Eye ,In Situ Hybridization ,Genetics ,Expressed Sequence Tags ,Mice, Knockout ,Differential display ,Expressed sequence tag ,Retinal pigment epithelium ,Computational Biology ,Retinal ,medicine.disease ,eye diseases ,DNA-Binding Proteins ,medicine.anatomical_structure ,Basic-Leucine Zipper Transcription Factors ,chemistry ,Mutation ,Data Display ,sense organs ,Visual phototransduction ,Photoreceptor Cells, Vertebrate - Abstract
Purpose Most retinal disease genes are preferentially expressed in photoreceptors, the light-sensitive cells involved in phototransduction. In addition, some of the genes linked to retinal diseases are essential for normal retinal development. The goal of this study was to identify new transcripts enriched in photoreceptors involved in retinal development or diseases. Methods To isolate uncharacterized retinal transcripts, the bioinformatic method Digital Differential Display (DDD) was used. RNA in situ hybridization was used to characterize gene-expression patterns. Results Twenty-seven mouse ESTs highly represented in retinal libraries were identified. Eight ESTs were predominantly expressed in photoreceptors and/or in the retinal pigment epithelium (RPE), whereas transcripts for other ESTs were detected more ubiquitously in the retinal cells or abundantly in ganglion cells and/or the inner nuclear layer. Mapping of the corresponding human orthologues of the photoreceptor/RPE-enriched genes revealed that two of them are candidate disease genes for retinitis pigmentosa, loci RP22 and RP28. Both of these are predominantly expressed in rod photoreceptors. The candidate RP22 gene codes for a putative transmembrane protein showing homology to Cln8 (ceroid lipofuscinosis, neuronal 8), in which gene mutations are associated with photoreceptors degeneration in mice. Also identified were two genes expressed in photoreceptors that are candidate disease genes for recessive Bardet-Biedl syndrome type 3 (BBS3) and recessive ataxia with RP (AXPC1). Conclusions This study demonstrates how bioinformatic analysis can be used to identify novel tissue-specific genes relevant to development and diseases.
- Published
- 2004
41. Pathological and genetic analysis of the degenerating muscle (dmu) mouse: a new allele of Scn8a
- Author
-
Rashmi Kothary, Sonia Girard, Silvia M. Vidal, Madeline Pool, Gilbert Bernier, Patrice D. Côté, and Yves De Repentigny
- Subjects
Candidate gene ,Mutant ,Locus (genetics) ,Nerve Tissue Proteins ,Biology ,Sodium Channels ,Chromosome 15 ,Mice ,Genetic linkage ,Genetics ,medicine ,Animals ,Humans ,Allele ,Muscle, Skeletal ,Molecular Biology ,Genetics (clinical) ,Alleles ,Crosses, Genetic ,Mice, Inbred C3H ,Chromosomes, Human, Pair 12 ,Myocardium ,Homozygote ,Cardiac muscle ,Chromosome Mapping ,General Medicine ,Neuromuscular Diseases ,Molecular biology ,Muscle atrophy ,Mice, Mutant Strains ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,NAV1.6 Voltage-Gated Sodium Channel ,Mutation ,medicine.symptom - Abstract
Here, we describe a novel spontaneous autosomal recessive mutation in the mouse that is characterized by skeletal and cardiac muscle degeneration. We have named this mutant degenerating muscle (dmu). At birth, mutant mice are indistinguishable from their normal littermates. Thereafter, the disease progresses rapidly and a phenotype is first observed at approximately 11 days after birth; the dmu mice are weak and have great difficulty in moving. The principal cause of the lack of mobility is muscle atrophy and wasting in the hindquarters. Affected mice die at or around the time of weaning of unknown causes. Histopathological observations and ultrastructural analysis revealed muscle degeneration in both skeletal and cardiac muscle, but no abnormalities in sciatic nerves. Using linkage analysis, we have mapped the dmu locus to the distal portion of mouse chromosome 15 in a region syntenic to human chromosome 12q13. Interestingly, scapuloperoneal muscular dystrophy (SPMD) in humans has been linked to this region. SPMD patients with associated cardiomyopathy have also been described in the past. Initial analysis of candidate genes on mouse chromosome 15 reveal that although intact transcripts for Scn8a, the gene encoding the sodium channel 8a subunit, are present in dmu mice, their levels are dramatically reduced. Furthermore, genetic complementation crosses between dmu and med (mutation in Scn8a) mice revealed that they are allelic. Our results suggest that at least a portion of the dmu phenotype is caused by a down-regulation of Scn8a, making dmu a new allele of Scn8a.
- Published
- 2001
42. Dystonin is an essential component of the Schwann cell cytoskeleton at the time of myelination
- Author
-
Martine Mathieu, S. David, Rashmi Kothary, Y. De Repentigny, and Gilbert Bernier
- Subjects
Heterozygote ,Dystonin ,Schwann cell ,Nerve Tissue Proteins ,Matrix (biology) ,Biology ,medicine.disease_cause ,Mice ,medicine ,Animals ,Neurons, Afferent ,Peripheral Nerves ,RNA, Messenger ,Cytoskeleton ,Molecular Biology ,Myelin Sheath ,Basement membrane ,Mutation ,Homozygote ,RNA ,Heterozygote advantage ,Mice, Mutant Strains ,Cell biology ,Cytoskeletal Proteins ,Dystonia ,medicine.anatomical_structure ,nervous system ,Animals, Newborn ,Immunology ,Schwann Cells ,Carrier Proteins ,Spinal Nerve Roots ,Myelin P0 Protein ,Developmental Biology ,Demyelinating Diseases - Abstract
A central role for the Schwann cell cytoskeleton in the process of peripheral nerve myelination has long been suggested. However, there is no genetic or biological evidence as yet to support this assumption. Here we show that dystonia musculorum (dt) mice, which carry mutations in dystonin, a cytoskeletal crosslinker protein, have hypo/amyelinated peripheral nerves. In neonatal dt mice, Schwann cells were arrested at the promyelinating stage and had multiple myelinating lips. Nerve graft experiments and primary cultures of Schwann cells demonstrated that the myelination abnormality in dt mice was autonomous to Schwann cells. In culture, dt Schwann cells showed abnormal polarization and matrix attachment, and had a disorganized cytoskeleton. Finally, we show that the dt mutation was semi-dominant, heterozygous animals presenting hypo- and hyper-myelinated peripheral nerves. Altogether, our results suggest that dt Schwann cells are deficient for basement membrane interaction and demonstrate that dystonin is an essential component of the Schwann cell cytoskeleton at the time of myelination.
- Published
- 1998
43. Cloning and characterization of mouse ACF7, a novel member of the dystonin subfamily of actin binding proteins
- Author
-
Rashmi Kothary, Silvia M. Vidal, Gilbert Bernier, De Repentigny Y, and Martine Mathieu
- Subjects
Gene isoform ,Male ,Subfamily ,DNA, Complementary ,Dystonin ,Molecular Sequence Data ,Nerve Tissue Proteins ,Biology ,Mice ,Gene expression ,Genetics ,Animals ,Actin-binding protein ,Amino Acid Sequence ,RNA, Messenger ,Cloning, Molecular ,Actin ,Mice, Inbred BALB C ,Base Sequence ,Microfilament Proteins ,Chromosome Mapping ,Cell biology ,Cytoskeletal Proteins ,MACF1 ,biology.protein ,Carrier Proteins ,Binding domain - Abstract
We have recently cloned the gene responsible for the mouse neurological disorder dystonia musculorum. The predicted product of this gene, dystonin (Dst), is a neural isoform of bullous pemphigoid antigen 1 (Bpag1) with an N-terminal actin binding domain. Here we report on the cloning and characterization of mouse ACF7. Sequence analysis revealed extended homology of mACF7 with both the actin binding domain (ABD) and the Bpag1 portions of dystonin. Moreover, mACF7 and Dst display similar isoform diversity and encode similar sized transcripts in the nervous system. Phylogenetic analysis of mACF7 and dystonin ABD sequences suggests a recent evolutionary origin and that these proteins form a separate novel subfamily within the β-spectrin superfamily of actin binding proteins. Given the implication of several actin binding proteins in genetic disorders, it is important to know the pattern of mACF7 expression. mACF7 transcripts are detected principally in lung, brain, spinal cord, skeletal and cardiac muscle, and skin. Intriguingly, mACF7 expression in lung is strongly induced just before birth and is restricted to type II alveolar cells. To determine whether spontaneous mutants that may be defective in mACF7 exist, we have mapped the mACF7 gene to mouse chromosome 4.
- Published
- 1996
44. Dystonin expression in the developing nervous system predominates in the neurons that degenerate in dystonia musculorum mutant mice
- Author
-
Yves De Repentigny, Gratien Dalpé, Rashmi Kothary, Arthur Brown, Martine Mathieu, and Gilbert Bernier
- Subjects
Cerebellum ,Dystonin ,Sensory system ,Mice, Inbred Strains ,Nerve Tissue Proteins ,Biology ,Motor Activity ,Axonal Transport ,Cellular and Molecular Neuroscience ,Embryonic and Fetal Development ,Mice ,Mice, Neurologic Mutants ,Sensory ataxia ,Motor system ,medicine ,Animals ,Neurons, Afferent ,Axon ,Molecular Biology ,In Situ Hybridization ,Dystonia ,Neurons ,Brain ,Gene Expression Regulation, Developmental ,Cell Biology ,medicine.disease ,Blotting, Northern ,Embryo, Mammalian ,Axons ,Cytoskeletal Proteins ,medicine.anatomical_structure ,nervous system ,Spinal Cord ,Nerve Degeneration ,Axoplasmic transport ,medicine.symptom ,Carrier Proteins ,Neuroscience - Abstract
Dystonia musculorum (dt) is an inherited neurodegenerative disorder in mice. The dt gene product, dystonin, contains the bullous pemphigoid antigen 1 coding region at its C-terminus and an actin binding domain at its N-terminus. We demonstrate that dystonin expression throughout mouse development predominates in neurons of the cranial and spinal sensory ganglia. These structures are the most severely affected in dystonic mice which could explain their severe sensory ataxia. Since we show expression in sensory neurons with small and large axoplasmic volumes, but degeneration is restricted primarily to the latter type, we suggest that caliber and size of the axon is an important factor in the disease process. Dystonin is also expressed in the extrapyramidal motor system and in the cerebellum. Functional defects in these cell types could account for the dystonic symptoms of dt mice not explained by simple sensory denervation. We also detect dystonin expression in motor neurons most of which are unaffected by the degenerative process in dt mice.
- Published
- 1995
45. Dystonin transcripts are altered and their levels are reduced in the mouse neurological mutant dt24J
- Author
-
Gilbert Bernier, Y. De Repentigny, Martine Mathieu, Gratien Dalpé, Arthur Brown, and Rashmi Kothary
- Subjects
Genetics ,Dystonia ,Nervous system ,Candidate gene ,Dystonin ,Mutant ,Sensory system ,Mice, Transgenic ,Nerve Tissue Proteins ,Cell Biology ,Biology ,medicine.disease ,Biochemistry ,Protein Structure, Tertiary ,Cytoskeletal Proteins ,Mice ,Mice, Neurologic Mutants ,medicine.anatomical_structure ,medicine ,Animals ,RNA, Messenger ,Carrier Proteins ,Molecular Biology - Abstract
Dystonia musculorum is a hereditary mouse neurodegenerative disorder that primarily affects the sensory arm of the nervous system. We have recently cloned and identified a candidate gene for this disorder and designated it dystonin. The sequence of dystonin predicts a rod-shaped cytoskeletal-associated protein with an actin-binding domain at the N-terminal end and a hemidesmosomal protein sequence (bpag1) at the C-terminal end. Here we show that abnormal dystonin transcripts are present in neural tissues of a spontaneous dystonia musculorum mutant, dt24J. We further show that dystonin transcript levels are reduced 2- to 3-fold in dt24Jmice.Key words: mouse, dystonia musculorum, neurodegeneration, cytoskeleton, bpag1.
- Published
- 1995
46. The mouse dystonia musculorum gene is a neural isoform of bullous pemphigoid antigen 1
- Author
-
Martine Mathieu, Janet Rossant, Gilbert Bernier, Arthur Brown, and Rashmi Kothary
- Subjects
Gene isoform ,DNA, Complementary ,Transcription, Genetic ,Dystonin ,Transgene ,Mutant ,Molecular Sequence Data ,Dystonia Musculorum Deformans ,Gene Expression ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Autoantigens ,Mice ,Species Specificity ,Pemphigoid, Bullous ,Genetics ,medicine ,Animals ,Humans ,Amino Acid Sequence ,In Situ Hybridization ,Plakin ,Base Sequence ,Sequence Homology, Amino Acid ,Neurodegeneration ,Non-Fibrillar Collagens ,medicine.disease ,Molecular biology ,Cytoskeletal Proteins ,Mutation ,Bullous pemphigoid ,Collagen ,Carrier Proteins - Abstract
Dystonia musculorum (dt) is a hereditary neurodegenerative disease in mice that leads to a sensory ataxia. We describe cloning of a candidate dt gene, dystonin, that is predominantly expressed in the dorsal root ganglia and other sites of neurodegeneration in dt mice. Dystonin encodes an N-terminal actin binding domain and a C-terminal portion comprised of the hemidesmosomal protein, bullous pemphigoid antigen 1 (bpag1). dt and bpag1 are part of the same transcription unit which is partially deleted in a transgenic strain of mice, Tg4, that harbours an insertional mutation at the dt locus, and in mice that carry a spontaneous dt mutation, dtAlb. We also demonstrate abnormal dystonin transcripts in a second dt mutant, dt24J. We conclude that mutations in the dystonin gene are the primary genetic lesion in dt mice.
- Published
- 1995
47. p53 Pro-Oxidant Activity in the Central Nervous System: Implication in Aging and Neurodegenerative Diseases.
- Author
-
Wassim Chatoo, Mohamed Abdouh, and Gilbert Bernier
- Published
- 2011
- Full Text
- View/download PDF
48. The LIM homeobox transcription factor Lhx2 is required to specify the retina field and synergistically cooperates with Pax6 for Six6 trans-activation
- Author
-
Marie-Pier Champagne, Gilbert Bernier, and Nicolas Tétreault
- Subjects
PAX6 Transcription Factor ,Six6 ,Eye ,Mice ,0302 clinical medicine ,Paired Box Transcription Factors ,Cells, Cultured ,In Situ Hybridization ,Mice, Knockout ,0303 health sciences ,Stem cell ,Stem Cells ,Gene Expression Regulation, Developmental ,Lhx2 ,Optic vesicle ,Trans-activation ,Chromatin ,Cell biology ,medicine.anatomical_structure ,embryonic structures ,Neural plate ,Transcription ,LIM-Homeodomain Proteins ,Molecular Sequence Data ,Biology ,Retina ,03 medical and health sciences ,Homeobox gene ,medicine ,Animals ,Eye Proteins ,Transcription factor ,Molecular Biology ,030304 developmental biology ,Homeodomain Proteins ,Cell Biology ,eye diseases ,Pax6 ,Repressor Proteins ,Eye development ,Cancer research ,Trans-Activators ,Homeobox ,PAX6 ,sense organs ,030217 neurology & neurosurgery ,Transcription Factors ,Developmental Biology - Abstract
In mammals, a limited set of homeobox-containing transcription factors are expressed in the presumptive eye field and required to initiate eye development. How these factors interact together at the genetic and molecular level to coordinate this developmental process is poorly understood. We found that the Lhx2 and Pax6 transcription factors operate in a concerted manner during retinal development to promote transcriptional activation of the Six6 homeobox-gene in primitive and mature retinal progenitors. Lhx2 demarcates the presumptive retina field at the neural plate stage and Lhx2 inactivation delays initiation of Rx, Six3 and Pax6 expression in this domain. The later expressed Six6 is properly activated in the pituitary/hypothalamic axis of Lhx2−/− embryos, but expression fails to be initiated in the optic vesicle. Lhx2 and Pax6 associate with the chromatin at several regions of Six6 in vivo and cooperate for trans-activation of Six6 regulatory elements in vitro. In retinal progenitor/stem cells, both Lhx2 and Pax6 are genetically required for proper Six6 expression and forced co-expression of Lhx2 and Pax6 can synergistically trans-activate the Six6 locus. Our work reveals how two master regulators of eye development coordinate their action to sequentially promote tissue-specific transcriptional initiation and full activation of a retinal determinant gene.
- Full Text
- View/download PDF
49. Pax6 is required for delta-catenin/neurojugin expression during retinal, cerebellar and cortical development in mice
- Author
-
Djamila Boutemmine, Robert-Hugues Duparc, Gilbert Bernier, Marie-Pier Champagne, and Nicolas Tétreault
- Subjects
Delta Catenin ,endocrine system ,Neurite ,PAX6 Transcription Factor ,Neocortex ,Biology ,Microarray ,Development ,Retina ,Delta-catenin ,Adherens junction ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cerebellum ,Animals ,Humans ,Paired Box Transcription Factors ,Optic vesicle ,Cytoskeleton ,Eye Proteins ,Transcription factor ,Molecular Biology ,030304 developmental biology ,Cerebral Cortex ,Homeodomain Proteins ,Mice, Knockout ,0303 health sciences ,Neuroectoderm ,Catenins ,Cell Biology ,Vinculin ,Phosphoproteins ,Molecular biology ,eye diseases ,Cell biology ,Pax6 ,Repressor Proteins ,biology.protein ,NIH 3T3 Cells ,PAX6 ,sense organs ,Delta catenin ,Cell Adhesion Molecules ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
The transcription factor Pax6 regulates multiple aspects of central nervous system (CNS) development. At the cellular level, the Pax6 mutation was reported to affect homophilic and heterophilic cellular adhesion, neuron polarity and neurite outgrowth. These abnormalities were observed in multiple regions of Pax6-mutant CNS, suggesting a common function for Pax6 in regulating cytoskeletal dynamics. However, target genes mediating Pax6 function in cytoskeletal dynamics remain largely unknown. Using DNA microarrays, we identified delta-catenin (delta-catenin /neurojugin) as a potential direct target of Pax6 in the CNS. delta-catenin encodes a large cytoskeletal protein that localizes at adherens junction in the CNS and that can modulate neurite outgrowth and N-cadherin turnover. delta-catenin was found to be co-expressed with Pax6 in several regions of the developing CNS. In Pax6 mutant embryos, delta-catenin expression was severely reduced in the optic vesicle neural ectoderm, in the ventricular zone of the neocortex and in the external granule layer of the cerebellum. We identified a Pax6 binding site in delta-catenin promoter that is conserved between mice and humans and which is effectively bound by Pax6 in vitro. Our results suggest that Pax6 regulates delta-catenin expression during CNS development in mice.
- Full Text
- View/download PDF
50. Spatial specification of mammalian eye territories by reciprocal transcriptional repression of Pax2 and Pax6
- Author
-
Nicole Andrejewski, Gilbert Bernier, Birgitta Kammandel, Peter Gruss, Martin Wagner, Martin K. Schwarz, and Francesco Cecconi
- Subjects
animal structures ,PAX6 Transcription Factor ,Transcription, Genetic ,genetic structures ,Mammalian eye ,Mice, Transgenic ,Optic cup (anatomical) ,Biology ,Eye ,Mice ,medicine ,Animals ,Paired Box Transcription Factors ,Optic stalk ,Eye Proteins ,Enhancer ,Molecular Biology ,Body Patterning ,Homeodomain Proteins ,Genetics ,Retina ,Models, Genetic ,PAX2 Transcription Factor ,Pax genes ,eye diseases ,Cell biology ,DNA-Binding Proteins ,Repressor Proteins ,Enhancer Elements, Genetic ,medicine.anatomical_structure ,embryonic structures ,Ectopic expression ,sense organs ,PAX6 ,Transcription Factors ,Developmental Biology - Abstract
We have studied the molecular basis of the Pax2 and Pax6 function in the establishment of visual system territories. Loss-of-function mutants have revealed crucial roles for Pax2 in the generation of the optic stalk and for Pax6 in the development of the optic cup. Ectopic expression of Pax6 in the optic stalk under control of Pax2 promoter elements resulted in a shift of the optic cup/optic stalk boundary indicated by the presence of retinal pigmented cells on the optic stalk. By studying mouse embryos at early developmental stages we detected an expansion of Pax2 expression domain in the Pax6−/− mutant and of Pax6 expression domain in the Pax2−/− embryo. These results suggest that the position of the optic cup/optic stalk boundary depends on Pax2 and Pax6 expression, hinting at a possible molecular interaction. Using gel shift experiments, we confirmed the presence of Pax2- and Pax6-binding sites on the retina enhancer of the Pax6 gene and on the Pax2 upstream control region, respectively. Co-transfection experiments revealed a reciprocal inhibition of Pax2 promoter/enhancer activity by Pax6 protein and vice versa. Based on our findings, we propose a model for Pax gene regulation that establishes the proper spatial regionalization of the mammalian visual system.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.