Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Am J Med Genet A ; : e63642, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38711237

ABSTRACT

The autosomal dominant Okur-Chung neurodevelopmental syndrome (OCNDS: OMIM #617062) is a rare neurodevelopmental disorder first described in 2016. Features include developmental delay (DD), intellectual disability (ID), behavioral problems, hypotonia, language deficits, congenital heart abnormalities, and non-specific dysmorphic facial features. OCNDS is caused by heterozygous pathogenic variants in CSNK2A1 (OMIM *115440; NM_177559.3). To date, 160 patients have been diagnosed worldwide. The number will likely increase due to the growing use of exome sequencing (ES) and genome sequencing (GS). Here, we describe a novel OCNDS patient carrying a CSNK2A1 variant (NM_177559.3:c.140G>A; NP_808227.1:p.Arg47Gln). Phenotypically, he presented with DD, ID, generalized hypotonia, speech delay, short stature, microcephaly, and dysmorphic features such as low-set ears, hypertelorism, thin upper lip, and a round face. The patient showed several signs not yet described that may extend the phenotypic spectrum of OCNDS. These include prenatal bilateral clubfeet, exotropia, and peg lateral incisors. However, unlike the majority of descriptions, he did not present sleep disturbance, seizures or gait difficulties. A literature review shows phenotypic heterogeneity for OCNDS, whether these patients have the same variant or not. This case report is an opportunity to refine the phenotype of this syndrome and raise the question of the genotype-phenotype correlation.

2.
Mol Neurobiol ; 56(2): 892-906, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29804229

ABSTRACT

Gestational methyl donor (especially B9 and B12 vitamins) deficiency is involved in birth defects and brain development retardation. The underlying molecular mechanisms that are dysregulated still remain poorly understood, in particular in the cerebellum. As evidenced from previous data, females are more affected than males. In this study, we therefore took advantage of a validated rat nutritional model and performed a microarray analysis on female progeny cerebellum, in order to identify which genes and molecular pathways were disrupted in response to methyl donor deficiency. We found that cerebellum development is altered in female pups, with a decrease of the granular cell layer thickness at postnatal day 21. Furthermore, we investigated the involvement of the Wnt signaling pathway, a major molecular pathway involved in neuronal development and later on in synaptic assembly and neurotransmission processes. We found that Wnt canonical pathway was disrupted following early methyl donor deficiency and that neuronal targets were selectively enriched in the downregulated genes. These results could explain the structural brain defects previously observed and highlighted new genes and a new molecular pathway affected by nutritional methyl donor deprivation.


Subject(s)
Brain/metabolism , Neurogenesis/physiology , Neurons/cytology , Wnt Signaling Pathway/physiology , Animals , Cells, Cultured , Female , Rats, Wistar , Sex Factors
3.
Brain Behav Immun ; 63: 137-147, 2017 Jul.
Article in English | MEDLINE | ID: mdl-27702682

ABSTRACT

Several lines of evidence indicate that early-life inflammation may predispose to mental illness, including depression, in later-life. We investigated the impact of perinatal exposure to polluted eels on neonatal, postnatal, and adult brain inflammation, and on the resignation behavior of male and female adult offspring mice. The effects of maternal standard diet (laboratory food) were compared to the same diet enriched with low, intermediate, or highly polluted eels. Brain inflammatory markers including cytokines were assessed in offspring mice on the day of birth (i.e., on the postnatal day-PND 1), upon weaning (PND 21) and at adulthood (PND 100). Plasma myeloperoxidase and corticosterone levels were evaluated at PND 100. Immobility behavior of offspring was assessed in adulthood (i.e., at PNDs 95-100), using the tail suspension and forced swimming tests. Chronic brain inflammation was found in male and female offspring mice compared to controls, as assessed at PNDs 1, 21, and 100. The level of myeloperoxidase was found to be significantly higher in both adult males and females vs. control offspring. However, high corticosterone levels were only found in male offspring mice that were perinatally exposed to eels, suggesting a gender-selective dysregulation of the adult hypothalamic-pituitaryadrenal (HPA) axis. Gender-specific differences were also detected in adulthood in regard to offspring resignation behavior. Thus, compared to controls, males, but not females, whose mothers were fed eels during pregnancy and lactation exhibited a depressive-like behavior in adult age in both behavioral models of depression. Depressive symptoms were more pronounced in male mice perinatally exposed to either intermediate or highly polluted eels than those exposed to only lowly polluted eels. Our results indicate that early-life inflammatory insult is a plausible causative factor that induces the depressive phenotype exhibited by male adult offspring mice, most likely through a gender-specific HPA axis enhanced activation.


Subject(s)
Depressive Disorder/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Behavior, Animal/physiology , Biomarkers/blood , Brain , Corticosterone/blood , Depression/genetics , Depression/immunology , Depressive Disorder/genetics , Female , Hippocampus/drug effects , Hypothalamo-Hypophyseal System/drug effects , Inflammation , Mice , Pituitary-Adrenal System/drug effects , Pregnancy , Sex Factors
4.
Toxicol Lett ; 245: 7-14, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26724586

ABSTRACT

The developing central nervous system is particularly vulnerable to environmental contaminants such as non-dioxin-like polychlorinated biphenyls (NDL-PCBs). This study investigated the potential oxidative effects in mice pups exposed via lactation to the sum of the six indicator NDL-PCBs (∑6 NDL-PCBs) at 0, 1, 10 and 100 ng/kg per 14 days, constituting levels below the guidance values fixed by French food safety agencies for humans at 10 ng/kg body weight per day. For this purpose, the oxidative status was assessed by flow cytometry via dichloro-dihydro-fluorescein diacetate in the cerebellum of juvenile male offspring mice during brain growth spurt [postnatal day (PND) 14]. No significant differences were found in the levels of reactive oxygen species in the cerebellar neurons or glial cells (astrocytes, oligodendrocytes and microglia) of lactationally exposed male mice at PND 14 (p>0.05). Concordantly, oxidative-stress related gene expression was measured by qPCR for catalase, copper zinc superoxide dismutase 1, glyoxalase 1, glutathione peroxidase 1, and glutathione reductase 1, in the cerebellum at PND 14 appeared unaffected, as also verified at the protein level by immunoblots. Moreover, transcriptomic data from our previous work have not shown differences in the mRNA expressions of genes belonging to GO terms involved in oxidative stress in neurons of male mice exposed to ∑6 NDL-PCBs compared to controls; except for glyoxalase 1 which was downregulated in neurons isolated from exposed group compared to controls. Our findings suggest that lactational exposure to NDL-PCBs at environmental relevant concentrations may not cause significant oxidative effect on juvenile cerebellum.


Subject(s)
Cerebellum/drug effects , Lactation , Oxidative Stress/drug effects , Polychlorinated Biphenyls/toxicity , Animals , Antioxidants/metabolism , Brain/growth & development , Cerebellum/cytology , Cerebellum/growth & development , Female , Food Safety , Male , Mice , Neuroglia/metabolism , Neurons/metabolism , Polychlorinated Biphenyls/pharmacokinetics , Reactive Oxygen Species/metabolism , Transcriptome
5.
Toxicology ; 328: 57-65, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25510870

ABSTRACT

Previously, we evaluated the effects of lactational exposure to a representative mixture of the six indicator non-dioxin-like polychlorinated biphenyls (∑6 NDL-PCBs) at low levels on the neurobiological changes and developmental/behavioral performances in mice. In this study, we analyzed the global gene expression profile in cerebellar neurons isolated from male mice presenting the most significant induction of anxiety-like behavior in our previous study (10 ng/kg ∑6 NDL-PCBs). Our results revealed changes in the expression of 16658 genes in the neurons of the exposed mice. Among these, 693 upregulated [fold change (FC)>2; p<0.05] and 665 downregulated (FC<2; p<0.05) genes were statistically linked to gene ontology terms (GO). Overexpressed genes belonged to GO terms involved with the cell cycle, DNA replication, cell cycle checkpoint, response to DNA damage stimulus, regulation of RNA biosynthetic processes, and microtubule cytoskeleton organization. Downregulated genes belonged to terms involved with the transmission of nerve impulses, projection neurons, synapse hands, cell junctions, and regulation of RNA biosynthetic processes. Using qPCR, we quantified gene expression related to DNA damage and validated the transcriptomic study, as a significant overexpression of Atm-Atr Bard1, Brca2, Fancd2, Figf, Mycn, p53 and Rad51 was observed between groups (p<0.001). Finally, using immunoblots we determined the expression level of six selected proteins. We found that changes in the protein expression of Atm Brca1, p53, Kcnma1, Npy4r and Scn1a was significant between exposed and control groups (p<0.05), indicating that the expression pattern of these proteins agreed with the expression pattern of their genes by qPCR, further validating our transcriptomic findings. In conclusion, our study showed that early life exposure of male mice to a low level of ∑6 NDL-PCBs induced p53-dependent responses to cellular stress and a decrease in the expression of proteins involved in the generation, conduction, and transmission of electrical signals in neurons.


Subject(s)
Cerebellum/drug effects , DNA Damage , Lactation , Maternal Exposure/adverse effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Polychlorinated Biphenyls/toxicity , Age Factors , Animals , Blotting, Western , Cerebellum/growth & development , Cerebellum/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/drug effects , Gene Regulatory Networks/drug effects , Genetic Markers , Male , Mice , Nerve Tissue Proteins/genetics , Neural Conduction/drug effects , Neural Conduction/genetics , Neurons/metabolism , Oligonucleotide Array Sequence Analysis , Pregnancy , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Sex Factors , Synaptic Transmission/drug effects , Synaptic Transmission/genetics
6.
Leuk Lymphoma ; 54(3): 587-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22860893

ABSTRACT

Interleukin (IL)-24 has death-promoting effects on various proliferating cells including B-cells from chronic lymphocytic leukemia (CLL) and germinal center B-cells, but its molecular mechanisms are poorly understood. Using a B-cell differentiation model and mRNA profiling, we found that recombinant (r)IL-24 stimulated genes of the mitochondrial apoptotic pathway (Bax, Bid, Casp8, COX6C, COX7B) after 36 h, whereas the transcription of genes involved in DNA replication and metabolism was inhibited within 6 h. Unexpectedly, insulin-like growth factor 1 (IGF1), a hormone known to promote cell growth, was stimulated by IL-24. Activated B-cells express receptor for IGF1, to which they become sensitized and undergo apoptosis, a mechanism similar in this respect to IL-24-induced cell death. Furthermore, inhibition of the IGF1 pathway reversed the effects of IL-24. IL-24-mediated apoptosis was also antagonized by pifithrin-alpha, an inhibitor of p53 transactivation. Altogether, these results disclose sequential molecular signals generated by IL-24 in activated B-cells.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Cell Cycle Checkpoints/drug effects , Interleukins/pharmacology , Mitochondria/drug effects , Signal Transduction/drug effects , Apoptosis/genetics , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Blotting, Western , Caspase 8/genetics , Caspase 8/metabolism , Cell Cycle Checkpoints/genetics , Cells, Cultured , Child , Child, Preschool , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Humans , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Interleukins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mitochondria/genetics , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Time Factors , Transcriptome/drug effects , Transcriptome/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
7.
Eur J Hum Genet ; 20(12): 1216-23, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22739344

ABSTRACT

The Forkhead box G1 (FOXG1) gene has been implicated in severe Rett-like phenotypes. It encodes the Forkhead box protein G1, a winged-helix transcriptional repressor critical for forebrain development. Recently, the core FOXG1 syndrome was defined as postnatal microcephaly, severe mental retardation, absent language, dyskinesia, and dysgenesis of the corpus callosum. We present seven additional patients with a severe Rett-like neurodevelopment disorder associated with de novo FOXG1 point mutations (two cases) or 14q12 deletions (five cases). We expand the mutational spectrum in patients with FOXG1-related encephalopathies and precise the core FOXG1 syndrome phenotype. Dysgenesis of the corpus callosum and dyskinesia are not always present in FOXG1-mutated patients. We believe that the FOXG1 gene should be considered in severely mentally retarded patients (no speech-language) with severe acquired microcephaly (-4 to-6 SD) and few clinical features suggestive of Rett syndrome. Interestingly enough, three 14q12 deletions that do not include the FOXG1 gene are associated with phenotypes very reminiscent to that of FOXG1-mutation-positive patients. We physically mapped a putative long-range FOXG1-regulatory element in a 0.43 Mb DNA segment encompassing the PRKD1 locus. In fibroblast cells, a cis-acting regulatory sequence located more than 0.6 Mb away from FOXG1 acts as a silencer at the transcriptional level. These data are important for clinicians and for molecular biologists involved in the management of patients with severe encephalopathies compatible with a FOXG1-related phenotype.


Subject(s)
Chromosomes, Human, Pair 14/genetics , Forkhead Transcription Factors/genetics , Intellectual Disability/genetics , Microcephaly/genetics , Nerve Tissue Proteins/genetics , Silencer Elements, Transcriptional/genetics , Agenesis of Corpus Callosum/diagnosis , Agenesis of Corpus Callosum/genetics , Cell Line , Child , Child, Preschool , DNA Copy Number Variations , Dyskinesias/diagnosis , Dyskinesias/genetics , Female , Forkhead Transcription Factors/metabolism , Gene Deletion , Humans , Intellectual Disability/diagnosis , Male , Microcephaly/diagnosis , Nerve Tissue Proteins/metabolism , Phenotype , Physical Chromosome Mapping , Point Mutation , Protein Kinase C/genetics , Rett Syndrome/diagnosis , Rett Syndrome/genetics , Syndrome , Transcription, Genetic
8.
BMC Cell Biol ; 12: 12, 2011 Mar 30.
Article in English | MEDLINE | ID: mdl-21450070

ABSTRACT

BACKGROUND: In the bone marrow, hematopietic and mesenchymal stem cells form a unique niche in which the oxygen tension is low. Hypoxia may have a role in maintaining stem cell fate, self renewal and multipotency. However, whereas most studies addressed the effect of transient in vitro exposure of MSC to hypoxia, permanent culture under hypoxia should reflect the better physiological conditions. RESULTS: Morphologic studies, differentiation and transcriptional profiling experiments were performed on MSC cultured in normoxia (21% O2) versus hypoxia (5% O2) for up to passage 2. Cells at passage 0 and at passage 2 were compared, and those at passage 0 in hypoxia generated fewer and smaller colonies than in normoxia. In parallel, MSC displayed (>4 fold) inhibition of genes involved in DNA metabolism, cell cycle progression and chromosome cohesion whereas transcripts involved in adhesion and metabolism (CD93, ESAM, VWF, PLVAP, ANGPT2, LEP, TCF1) were stimulated. Compared to normoxic cells, hypoxic cells were morphologically undifferentiated and contained less mitochondrias. After this lag phase, cells at passage 2 in hypoxia outgrew the cells cultured in normoxia and displayed an enhanced expression of genes (4-60 fold) involved in extracellular matrix assembly (SMOC2), neural and muscle development (NOG, GPR56, SNTG2, LAMA) and epithelial development (DMKN). This group described herein for the first time was assigned by the Gene Ontology program to "plasticity". CONCLUSION: The duration of hypoxemia is a critical parameter in the differentiation capacity of MSC. Even in growth promoting conditions, hypoxia enhanced a genetic program that maintained the cells undifferentiated and multipotent. This condition may better reflect the in vivo gene signature of MSC, with potential implications in regenerative medicine.


Subject(s)
Cell Differentiation , Cell Hypoxia , Gene Expression , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Cell Count , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Flow Cytometry , Gene Expression Profiling , Gene Expression Regulation , Humans , Mesenchymal Stem Cells/metabolism , Microscopy, Electron , Multipotent Stem Cells/metabolism , Stem Cell Research
9.
Am J Med Genet A ; 149A(11): 2493-500, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19842196

ABSTRACT

Monosomy 1p36 is the most frequent terminal deletion known in Humans. Typical craniofacial features, developmental delay/mental retardation, seizures and sensorineural defects characterize 1p36 deletion syndrome. Aicardi syndrome (AIS) is a rare genetic disorder characterized by chorioretinal lacunae, corpus callosum agenesis and infantile spasms responsible for mental retardation. By screening DNA from diagnosed AIS patients with oligonucleotide array-based comparative genomic hybridization (aCGH), we report a 1p36 monosomy in this study. There were no other deletions or duplications. Regarding clinical criteria, the patient did not have the typical facial appearance commonly described for 1p36 monosomy patients. We showed that this 1p36 monosomy corresponded to combined interstitial and terminal de novo deletions of the chromosome 1 leading to an 11.73 Mb deletion confirmed with qPCR. By microsatellite markers and FISH analyses, we have concluded that this deletion occurred on maternal chromosome 1 during oogenesis. We did find some clinical features shared by the 1p36 monosomy and AIS: infantile spasms, corpus callosum dysgenesis, ophthalmological abnormalities, and skeletal malformations. To date, no relationship between these two phenotypes has been established. We conclude that the monosomy 1p36 should be considered in the differential diagnosis of AIS.


Subject(s)
Abnormalities, Multiple/genetics , Chromosomes, Human, Pair 1/genetics , Monosomy/genetics , Adult , Child , Chromosome Deletion , Female , Gene Rearrangement/genetics , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Parents , Phenotype , Polymerase Chain Reaction , Syndrome
10.
PLoS One ; 4(9): e7077, 2009 Sep 22.
Article in English | MEDLINE | ID: mdl-19771162

ABSTRACT

BACKGROUND: The human PAF (hPAF) complex is part of the RNA polymerase II transcription apparatus and regulates multiple steps in gene expression. Further, the yeast homolog of hPaf1 has a role in regulating the expression of a subset of genes involved in the cell-cycle. We therefore investigated the role of hPaf1 during progression of the cell-cycle. METHODOLOGY/FINDINGS: Herein, we report that the expression of hPaf1, a subunit of the hPAF complex, increases with cell-cycle progression and is regulated in a cell-cycle dependant manner. hPaf1 specifically regulates a subclass of genes directly implicated in cell-cycle progression during G1/S, S/G2, and G2/M. In prophase, hPaf1 aligns in filament-like structures, whereas in metaphase it is present within the pole forming a crown-like structure, surrounding the centrosomes. Moreover, hPaf1 is degraded during the metaphase to anaphase transition. In the nucleus, hPaf1 regulates the expression of cyclins A1, A2, D1, E1, B1, and Cdk1. In addition, expression of hPaf1 delays DNA replication but favors the G2/M transition, in part through microtubule assembly and mitotic spindle formation. CONCLUSION/SIGNIFICANCE: Our results identify hPaf1 and the hPAF complex as key regulators of cell-cycle progression. Mutation or loss of stoichiometry of at least one of the members may potentially lead to cancer development.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Anaphase , Cell Cycle , Cell Line, Tumor , Cell Nucleus/metabolism , Centrosome/metabolism , Disease Progression , Humans , Metaphase , Microtubules/metabolism , Models, Biological , Spindle Apparatus , Transcription Factors , Transcription, Genetic
11.
Eur J Med Genet ; 50(5): 386-91, 2007.
Article in English | MEDLINE | ID: mdl-17625997

ABSTRACT

Aicardi syndrome (AIC) is an uncommon neurodevelopmental disorder affecting almost exclusively females. Chief features include infantile spasms, corpus callosal agenesis, and chorioretinal abnormalities. AIC is a sporadic disorder and hypothesized to be caused by heterozygous mutations in an X-linked gene but up to now without any defined candidate region on the X chromosome. Array based comparative genomic hybridisation (array-CGH) has become the method of choice for the detection of microdeletions and microduplications at high resolution. In this study, for the first time, 18 AIC patients were analyzed with a full coverage X chromosomal BAC arrays at a theoretical resolution of 82 kb. Copy number changes were validated by real-time quantitation (qPCR). No disease associated aberrations were identified. For such conditions as AIC, in which there are no familial cases, additional patients should be studied in order to identify rare cases with submicroscopic abnormalities, and to pursue a positional candidate gene approach.


Subject(s)
Abnormalities, Multiple/genetics , Agenesis of Corpus Callosum , Choroid/abnormalities , Chromosomes, Human, X/genetics , Genetic Diseases, X-Linked/genetics , Retina/abnormalities , Spasms, Infantile/genetics , Adolescent , Adult , Base Sequence , Child , Contractile Proteins/genetics , DNA Primers/genetics , Developmental Disabilities/genetics , Female , Filamins , Gene Dosage , Humans , Infant , Microfilament Proteins/genetics , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Syndrome
12.
Biochim Biophys Acta ; 1731(1): 23-31, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16153720

ABSTRACT

The gec1/GABARAPL1 (GABA(A)-receptor-associated protein like-1) gene has been identified as an early estrogen-regulated gene in guinea-pig cultured endometrial glandular epithelial cells (GEC). Guinea-pig and human gec1/GABARAPL1 proteins share 87% identity with GABARAP, which acts as a protein linker between microtubules and the GABA(A) receptor. To investigate the molecular mechanisms regulating gec1/GABARAPL1 gene expression, the 1.5-kbp region upstream of the translation initiation codon of the guinea-pig gec1/GABARAPL1 gene was cloned. A 300-bp fragment encompassing a pyrimidine-rich initiator element (INR) and the transcription start site (+1) was sufficient to initiate transcription. Transfection and gel shift experiments showed that a sequence located at +36/+50 in the first exon permitted induction of expression of this gene by estradiol acting via ERalpha. This sequence (GGGTCAACGTGACGT) differs only by one base pair from the consensus estrogen response element ERE (GGGTCAACGTGACCT). It can be concluded that the ERE located in the first exon encoding the 5'-untranslated region is sufficient for E2 activation of gec1/GABARAPL1 transcription.


Subject(s)
Estrogens/pharmacology , Microtubule-Associated Proteins/genetics , Promoter Regions, Genetic/genetics , Response Elements/genetics , Adaptor Proteins, Signal Transducing , Animals , Base Sequence , CHO Cells , Cricetinae , Estradiol/pharmacology , Estrogen Receptor alpha/biosynthesis , Estrogen Receptor alpha/metabolism , Exons , Female , Gene Expression Regulation , Guinea Pigs , Microtubule-Associated Proteins/biosynthesis , Molecular Sequence Data , Transcription Initiation Site
13.
Exp Cell Res ; 296(2): 109-22, 2004 Jun 10.
Article in English | MEDLINE | ID: mdl-15149842

ABSTRACT

In normal endometrial glandular epithelial cells (GEC), 17beta-estradiol (E2) enhances proliferation and c-fos expression only in the presence of growth factors. On the contrary, growth factors are not required for the E2 effects in cancerous cells. Thus, a repression of E2 action could exist in normal cells and be turned off in cancerous cells, allowing a direct estrogen-dependent proliferation. To verify this hypothesis, we established immortalized and transformed cell models, then investigated alterations of E2 effects during oncogenesis. SV40 large T-antigen was used to generate immortalized GEC model (IGEC). After observation of telomerase reactivation, IGEC model was transfected by activated c-Ha-ras to obtain transformed cell lines (TGEC1 and TGEC2). The phenotypic, morphological, and genetic characteristics of these models were determined before studying the E2 effects. In IGEC, the E2 action on proliferation and c-fos expression required the presence of growth factors, as observed in GECs. In TGECs, this action arose in the absence of growth factors. After IGEC transformation, the activation of ras pathway would substitute the priming events required for the release of repression in GEC and IGEC and thus permit direct E2 effects. Our cell models are particularly suitable to investigate alterations of gene regulation by E2 during oncogenesis.


Subject(s)
Cell Transformation, Neoplastic/chemically induced , Epithelial Cells/drug effects , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Antigens, Viral, Tumor , Cell Division/drug effects , Cell Line, Transformed , Endometrial Neoplasms/etiology , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Epithelial Cells/pathology , Female , Genes, ras , Guinea Pigs , Proto-Oncogene Proteins c-fos/genetics , Transfection
14.
Brain Res Mol Brain Res ; 119(2): 216-9, 2003 Nov 26.
Article in English | MEDLINE | ID: mdl-14625090

ABSTRACT

GABARAP and gec1/GABARAPL1 genes encode very similar proteins belonging to a new microtubule-associated protein (MAP) family. These proteins could participate in a complex clustering, targeting and/or degrading the GABA(A) receptors on post-synaptic membrane of neurons. Using specific cDNA probes, we investigated the differential expression of both genes in 76 human tissues. Against all odds, gec1/GABARAPL1 was more expressed than GABARAP in the central nervous system (CNS), while GABARAP was more expressed in endocrine glands.


Subject(s)
Central Nervous System/metabolism , Endocrine Glands/metabolism , Gene Expression/genetics , Microtubule-Associated Proteins/genetics , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins , Cell Membrane/metabolism , Chromosomes, Human, Pair 12/genetics , Chromosomes, Human, Pair 17/genetics , Fetus , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Receptor Aggregation/genetics , Receptors, GABA-A/metabolism , Synaptic Transmission/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...