Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cell Mol Life Sci ; 76(4): 627-635, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30341460

ABSTRACT

Hearing loss is a common affection mainly resulting from irreversible loss of the sensory hair cells of the cochlea; therefore, developing therapies to replace missing hair cells is essential. Understanding the mechanisms that drive their formation will not only help to unravel the molecular basis of deafness, but also give a roadmap for recapitulating hair cells development from cultured pluripotent stem cells. In this review, we provide an overview of the molecular mechanisms involved in hair cell production from both human and mouse embryonic stem cells. We then provide insights how this knowledge has been applied to differentiate induced pluripotent stem cells into otic progenitors and hair cells. Finally, we discuss the current limitations for properly obtaining functional hair cell in a Petri dish, as well as the difficulties that have to be overcome prior to consider stem cell therapy as a potential treatment for hearing loss.


Subject(s)
Cell Differentiation , Cochlea/cytology , Hair Cells, Auditory/cytology , Pluripotent Stem Cells/cytology , Stem Cells/cytology , Animals , Hearing Loss/therapy , Humans , Mice , Stem Cell Transplantation/methods
2.
Eur J Ophthalmol ; 28(4): NP11-NP14, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29623723

ABSTRACT

PURPOSE: To report a case of macular toxicity and blind spot enlargement during voriconazole treatment. METHODS: This is a case report. RESULTS: We describe a 77-year-old man treated by voriconazole for pulmonary aspergillosis, who complained of visual disorders such as dyschromatopsia and visual hallucinations 3 days after voriconazole initiation. Initial ophthalmological examination found no loss of visual acuity. The anterior and posterior segments presented no anomalies. The chromatic vision evaluated with the Lanthony 15-Hue Desaturated Test demonstrated dyschromatopsia in the left eye along the tritan axis, and the Goldmann visual field examination found a blind spot enlargement in both eyes. The multifocal electroretinogram found a global decrease in the foveal peak in both eyes. Visual evoked potential showed asymmetric data and lower amplitudes of the P(100) wave on the left eye. No anomalies were observed on spectral domain macular optical coherence tomography. As a first step, based on presumed dose-dependent toxicity, voriconazole dose was reduced. No improvements were noted. The voriconazole treatment was then discontinued and replaced with itraconazole. After 1 month, visual field and multifocal electroretinogram had improved and visual hallucinations had disappeared. CONCLUSION: Voriconazole can cause potentially serious visual side effects. Adapting treatment based on plasma concentrations of voriconazole did not prevent the appearance of visual side effects in this case. Therapeutic drug switching within the same drug family seems to be an effective alternative to preserve ocular function.


Subject(s)
Macula Lutea/drug effects , Retinal Diseases/chemically induced , Tomography, Optical Coherence/methods , Visual Acuity , Voriconazole/adverse effects , Aged , Antifungal Agents/adverse effects , Electroretinography , Evoked Potentials, Visual/drug effects , Humans , Macula Lutea/diagnostic imaging , Male , Pulmonary Aspergillosis/drug therapy , Retinal Diseases/diagnosis , Retinal Diseases/physiopathology
3.
Front Genet ; 9: 32, 2018.
Article in English | MEDLINE | ID: mdl-29472946

ABSTRACT

While the risks of maternal alcohol abuse during pregnancy are well-established, several preclinical studies suggest that chronic preconception alcohol consumption by either parent may also have significance consequences for offspring health and development. Notably, since isogenic male mice used in these studies are not involved in gestation or rearing of offspring, the cross-generational effects of paternal alcohol exposure suggest a germline-based epigenetic mechanism. Many recent studies have demonstrated that the effects of paternal environmental exposures such as stress or malnutrition can be transmitted to the next generation via alterations to small noncoding RNAs in sperm. Therefore, we used high throughput sequencing to examine the effect of preconception ethanol on small noncoding RNAs in sperm. We found that chronic intermittent ethanol exposure altered several small noncoding RNAs from three of the major small RNA classes in sperm, tRNA-derived small RNA (tDR), mitochondrial small RNA, and microRNA. Six of the ethanol-responsive small noncoding RNAs were evaluated with RT-qPCR on a separate cohort of mice and five of the six were confirmed to be altered by chronic ethanol exposure, supporting the validity of the sequencing results. In addition to altered sperm RNA abundance, chronic ethanol exposure affected post-transcriptional modifications to sperm small noncoding RNAs, increasing two nucleoside modifications previously identified in mitochondrial tRNA. Furthermore, we found that chronic ethanol reduced epididymal expression of a tRNA methyltransferase, Nsun2, known to directly regulate tDR biogenesis. Finally, ethanol-responsive sperm tDR are similarly altered in extracellular vesicles of the epididymis (i.e., epididymosomes), supporting the hypothesis that alterations to sperm tDR emerge in the epididymis and that epididymosomes are the primary source of small noncoding RNAs in sperm. These results add chronic ethanol to the growing list of paternal exposures that can affect small noncoding RNA abundance and nucleoside modifications in sperm. As small noncoding RNAs in sperm have been shown to causally induce heritable phenotypes in offspring, additional research is warranted to understand the potential effects of ethanol-responsive sperm small noncoding RNAs on offspring health and development.

4.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 152-161, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29038051

ABSTRACT

Alzheimer's disease (AD) is a multifactorial neurodegenerative disorder that is influenced by genetic and environmental risk factors, such as inheritance of ε4 allele of APOE (APOE4), sex and diet. Here, we examined the effect of high fat diet (HFD) on amyloid pathology and expression profile in brains of AD model mice expressing human APOE isoforms (APP/E3 and APP/E4 mice). APP/E3 and APP/E4 mice were fed HFD or Normal diet for 3months. We found that HFD significantly increased amyloid plaques in male and female APP/E4, but not in APP/E3 mice. To identify differentially expressed genes and gene-networks correlated to diet, APOE isoform and sex, we performed RNA sequencing and applied Weighted Gene Co-expression Network Analysis. We determined that the immune response network with major hubs Tyrobp/DAP12, Csf1r, Tlr2, C1qc and Laptm5 correlated significantly and positively to the phenotype of female APP/E4-HFD mice. Correspondingly, we found that in female APP/E4-HFD mice, microglia coverage around plaques, particularly of larger size, was significantly reduced. This suggests altered containment of the plaque growth and sex-dependent vulnerability in response to diet. The results of our study show concurrent impact of diet, APOE isoform and sex on the brain transcriptome and AD-like phenotype.


Subject(s)
Apolipoproteins E/genetics , Diet , Immunity, Innate/physiology , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Female , Gene Regulatory Networks , Gene-Environment Interaction , Genotype , Immunity, Innate/genetics , Male , Mice , Mice, Transgenic , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Sex Factors , Systems Biology/methods
5.
Cell Death Differ ; 24(12): 2054-2065, 2017 12.
Article in English | MEDLINE | ID: mdl-28777373

ABSTRACT

MicroRNAs are important regulators of gene expression and are involved in cellular processes such as proliferation or differentiation, particularly during development of numerous organs including the inner ear. However, it remains unknown if miRNAs are required during the earliest stages of otocyst and cochlear duct development. Here, we report that a conditional loss of Dicer expression in the otocyst impairs the early development of the inner ear as a result of the accumulation of DNA damage that trigger p53-mediated apoptosis. Moreover, cochlear progenitors in the prosensory domain do not exit the cell cycle. Our unbiased approach identified ItgA3 as a target of miR-183, which are both enriched in the otic vesicle. We observed that the repression of integrin alpha 3 by miR-183 controls cell proliferation in the developing cochlea. Collectively, our results reveal that Dicer and miRNAs play essential roles in the regulation of early inner ear development.


Subject(s)
Ear, Inner/embryology , Integrin alpha3/physiology , MicroRNAs/physiology , Animals , Cell Differentiation/physiology , Cell Line , Cochlea/cytology , Cochlea/embryology , DEAD-box RNA Helicases/genetics , Female , Mice , Mice, Knockout , Pregnancy , Ribonuclease III/genetics , Signal Transduction
6.
Sci Rep ; 7(1): 4307, 2017 06 27.
Article in English | MEDLINE | ID: mdl-28655926

ABSTRACT

We examined the effect of chronic high fat diet (HFD) on amyloid deposition and cognition of 12-months old APP23 mice, and correlated the phenotype to brain transcriptome and lipidome. HFD significantly increased amyloid plaques and worsened cognitive performance compared to mice on normal diet (ND). RNA-seq results revealed that in HFD mice there was an increased expression of genes related to immune response, such as Trem2 and Tyrobp. We found a significant increase of TREM2 immunoreactivity in the cortex in response to HFD, most pronounced in female mice that correlated to the amyloid pathology. Down-regulated by HFD were genes related to neuron projections and synaptic transmission in agreement to the significantly deteriorated neurite morphology and cognition in these mice. To examine the effect of the diet on the brain lipidome, we performed Shotgun Lipidomics. While there was no difference in the total amounts of phospholipids of each class, we revealed that the levels of 24 lipid sub-species in the brain were significantly modulated by HFD. Network visualization of correlated lipids demonstrated overall imbalance with most prominent effect on cardiolipin molecular sub-species. This integrative approach demonstrates that HFD elicits a complex response at molecular, cellular and system levels in the CNS.


Subject(s)
Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Brain/metabolism , Diet, High-Fat/adverse effects , Lipid Metabolism , Metabolome , Phenotype , Transcriptome , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Apoptosis , Brain/pathology , Cell Differentiation/genetics , Cognition , Computational Biology/methods , Disease Models, Animal , Female , Gene Expression Profiling , Maze Learning , Mice , Mice, Transgenic , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Plaque, Amyloid/pathology , Protein Aggregation, Pathological
7.
Neurobiol Dis ; 105: 1-14, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28502803

ABSTRACT

Traumatic brain injury (TBI) is strongly linked to an increased risk of developing dementia, including chronic traumatic encephalopathy and possibly Alzheimer's disease (AD). APOEε4 allele of human Apolipoprotein E (APOE) gene is the major genetic risk factor for late onset AD and has been associated with chronic traumatic encephalopathy and unfavorable outcome following TBI. To determine if there is an APOE isoform-specific response to TBI we performed controlled cortical impact on 3-month-old mice expressing human APOE3 or APOE4 isoforms. Following injury, we used several behavior paradigms to test for anxiety and learning and found that APOE3 and APOE4 targeted replacement mice demonstrate cognitive impairments following moderate TBI. Transcriptional profiling 14days following injury revealed a significant effect of TBI, which was similar in both genotypes. Significantly upregulated by injury in both genotypes were mRNA expression and protein level of ABCA1 transporter and APOJ, but not APOE. To identify gene-networks correlated to injury and APOE isoform, we performed Weighted Gene Co-expression Network Analysis. We determined that the network mostly correlated to TBI in animals expressing both isoforms is immune response with major hub genes including Trem2, Tyrobp, Clec7a and Cd68. We also found a significant increase of TREM2, IBA-1 and GFAP protein levels in the brains of injured mice. We identified a network representing myelination that correlated significantly with APOE isoform in both injury groups. This network was significantly enriched in oligodendrocyte signature genes, such as Mbp and Plp1. Our results demonstrate unique and distinct gene networks at this acute time point for injury and APOE isoform, as well as a network driven by APOE isoform across TBI groups.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apolipoproteins E/metabolism , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/physiopathology , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , Up-Regulation/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Anxiety/etiology , Apolipoproteins E/genetics , Astrocytes/metabolism , Astrocytes/pathology , Brain Injuries, Traumatic/complications , Cognition Disorders/etiology , Cognition Disorders/genetics , Disease Models, Animal , Gene Regulatory Networks , Glial Fibrillary Acidic Protein/metabolism , Humans , Maze Learning/physiology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Principal Component Analysis , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/metabolism , Receptors, Immunologic/genetics
8.
PLoS One ; 12(2): e0172161, 2017.
Article in English | MEDLINE | ID: mdl-28241068

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) controls cholesterol and phospholipid efflux to lipid-poor apolipoprotein E (APOE) and is transcriptionally controlled by Liver X receptors (LXRs) and Retinoic X Receptors (RXRs). In APP transgenic mice, lack of Abca1 increased Aß deposition and cognitive deficits. Abca1 haplo-deficiency in mice expressing human APOE isoforms, increased level of Aß oligomers and worsened memory deficits, preferentially in APOE4 mice. In contrast upregulation of Abca1 by LXR/RXR agonists significantly ameliorated pathological phenotype of those mice. The goal of this study was to examine the effect of LXR agonist T0901317 (T0) on the phenotype and brain transcriptome of APP/E3 and APP/E4 Abca1 haplo-deficient (APP/E3/Abca1+/- and APP/E4/Abca1+/-) mice. Our data demonstrate that activated LXRs/RXR ameliorated APOE4-driven pathological phenotype and significantly affected brain transcriptome. We show that in mice expressing either APOE isoform, T0 treatment increased mRNA level of genes known to affect brain APOE lipidation such as Abca1 and Abcg1. In both APP/E3/Abca1+/- and APP/E4/Abca1+/- mice, the application of LXR agonist significantly increased ABCA1 protein level accompanied by an increased APOE lipidation, and was associated with restoration of APOE4 cognitive deficits, reduced levels of Aß oligomers, but unchanged amyloid load. Finally, using Gene set enrichment analysis we show a significant APOE isoform specific response to LXR agonist treatment: Gene Ontology categories "Microtubule Based Process" and "Synapse Organization" were differentially affected in T0-treated APP/E4/Abca1+/- mice. Altogether, the results are suggesting that treatment of APP/E4/Abca1+/- mice with LXR agonist T0 ameliorates APOE4-induced AD-like pathology and therefore targeting the LXR-ABCA1-APOE regulatory axis could be effective as a potential therapeutic approach in AD patients, carriers of APOEε4.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , Apolipoprotein E3/genetics , Apolipoprotein E4/genetics , Liver X Receptors/agonists , Transcriptome , Amyloid beta-Peptides/metabolism , Animals , Behavior, Animal , Brain/metabolism , Cluster Analysis , Fear , Female , Haploinsufficiency , Heterozygote , Humans , Male , Maze Learning , Memory Disorders/metabolism , Mice , Mice, Transgenic , Microtubules/metabolism , Phenotype , Software , Up-Regulation
9.
Sci Rep ; 6: 24048, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27051978

ABSTRACT

Bexarotene, a selective agonist for Retinoid X receptors (RXR) improves cognitive deficits and amyloid-ß (Aß) clearance in mice. Here we examine if the effect of bexarotene on RXR cistrome and transcriptomes depend on APOE isoform and Aß deposition. We found bexarotene increased RXR binding to promoter regions in cortex of APOE3 mice. Rho family GTPases and Wnt signaling pathway were highly enriched in ChIP-seq and RNA-seq datasets and members of those pathways - Lrp1, Lrp5, Sfrp5 and Sema3f were validated. The effect of APOE isoform was compared in APOE3 and APOE4 mice and we found significant overlapping in affected pathways. ChIP-seq using mouse embryonic stem cells and enrichment levels of histone marks H3K4me3 and H3K27me3 revealed that, bexarotene induced epigenetic changes, consistent with increased neuronal differentiation and in correlation with changes in transcription. Comparison of transcriptome in APOE3 and APP/APOE3 mice revealed that amyloid deposition significantly affects the response to bexarotene. In primary neurons, bexarotene ameliorated the damaged dendrite complexity and loss of neurites caused by Aß42. Finally, we show that the disruption of actin cytoskeleton induced by Aß42 in vitro was inhibited by bexarotene treatment. Our results suggest a mechanism to establish RXR therapeutic targets with significance in neurodegeneration.


Subject(s)
Amyloid beta-Peptides/toxicity , Brain/metabolism , Gene Regulatory Networks , Protein Multimerization , Retinoid X Receptors/metabolism , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Animals , Apolipoprotein E3/metabolism , Axon Guidance/drug effects , Bexarotene , Brain/drug effects , Cell Differentiation/drug effects , Chromatin Immunoprecipitation , Epigenesis, Genetic/drug effects , Gene Regulatory Networks/drug effects , Genome , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Neurites/drug effects , Neurites/metabolism , Protein Binding/drug effects , Protein Multimerization/drug effects , Tetrahydronaphthalenes/pharmacology , Transcriptome/drug effects , Transcriptome/genetics
10.
J Neurosci ; 35(34): 11862-76, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311769

ABSTRACT

Bexarotene-activated retinoid X receptors (RXRs) ameliorate memory deficits in Alzheimer's disease mouse models, including mice expressing human apolipoprotein E (APOE) isoforms. The goal of this study was to gain further insight into molecular mechanisms whereby ligand-activated RXR can affect or restore cognitive functions. We used an unbiased approach to discover genome-wide changes in RXR cistrome (ChIP-Seq) and gene expression profile (RNA-Seq) in response to bexarotene in the cortex of APOE4 mice. Functional categories enriched in both datasets revealed that bexarotene-liganded RXR affected signaling pathways associated with neurogenesis and neuron projection development. To further validate the significance of RXR for these functions, we used mouse embryonic stem (ES) cells, primary neurons, and APOE3 and APOE4 mice treated with bexarotene. In vitro data from ES cells confirmed that bexarotene-activated RXR affected neuronal development at different levels, including proliferation of neural progenitors and neuronal differentiation, and stimulated neurite outgrowth. This effect was validated in vivo by demonstrating an increased number of neuronal progenitors after bexarotene treatment in the dentate gyrus of APOE3 and APOE4 mice. In primary neurons, bexarotene enhanced the dendritic complexity characterized by increased branching, intersections, and bifurcations. This effect was confirmed by in vivo studies demonstrating that bexarotene significantly improved the compromised dendritic structure in the hippocampus of APOE4 mice. We conclude that bexarotene-activated RXRs promote genetic programs involved in the neurogenesis and development of neuronal projections and these results have significance for the improvement of cognitive deficits. SIGNIFICANCE STATEMENT: Bexarotene-activated retinoid X receptors (RXRs) ameliorate memory deficits in Alzheimer's disease mouse models, including mice expressing human apolipoprotein E (APOE) isoforms. The goal of this study was to gain further insight into molecular mechanisms whereby ligand-activated RXR can affect or restore cognitive functions. We used an unbiased approach to discover genome-wide changes in RXR cistrome (ChIP-Seq) and gene expression profile (RNA-Seq) in response to bexarotene in the cortex of APOE4 mice. Functional categories enriched in both datasets revealed that liganded RXR affected signaling pathways associated with neurogenesis and neuron projection development. The significance of RXR for these functions was validated in mouse embryonic stem cells, primary neurons, and APOE3 and APOE4 mice treated with bexarotene.


Subject(s)
Cell Differentiation/physiology , Dendrites/metabolism , Neurogenesis/physiology , Retinoid X Receptors/metabolism , Tetrahydronaphthalenes/pharmacology , Animals , Bexarotene , Cell Differentiation/drug effects , Cells, Cultured , Dendrites/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/drug effects , Pregnancy , Rats , Rats, Sprague-Dawley , Retinoid X Receptors/agonists
11.
Neurobiol Dis ; 82: 132-140, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26071899

ABSTRACT

We have recently demonstrated that short term bexarotene treatment of APP/PS1 mice significantly improves their cognitive performance. While there were no changes in plaque load, or insoluble Aß levels in brain, biochemical analysis strongly suggested improved clearance of soluble Aß, including Aß oligomers. To get further insight into molecular mechanisms underlying this therapeutic effect, we explored genome-wide differential gene expression in brain of bexarotene and control treated APP/PS1 mice. We performed high throughput massively parallel sequencing on mRNA libraries generated from cortices of bexarotene or vehicle treated APP/PS1 mice and compared the expression profiles for differential gene expression. Gene Ontology (GO) Biological Process categories with the highest fold enrichment and lowest False Discovery Rate (FDR) are clustered in GO terms immune response, inflammatory response, oxidation-reduction and immunoglobulin mediated immune response. Chromatin immunoprecipitation (ChIP) followed by ChIP-QPCR, and RT-QPCR expression assays were used to validate select genes, including Trem2, Tyrobp, Apoe and Ttr, differentially expressed in response to Retinoid X Receptor (RXR) activation. We found that bexarotene significantly increased the phagocytosis of soluble and insoluble Aß in BV2 cells. The results of our study demonstrate that in AD model mice expressing human APP, gene networks up-regulated in response to RXR activation by the specific, small molecule, ligand bexarotene may influence diverse regulatory pathways that are considered critical for cognitive performance, inflammatory response and Aß clearance, and may provide an explanation of the bexarotene therapeutic effect at the molecular level. This study also confirms that unbiased massive parallel sequencing approaches are useful and highly informative for revealing brain molecular and cellular mechanisms underlying responses to activated nuclear hormone receptors in AD animal models.


Subject(s)
Anticarcinogenic Agents/pharmacology , Brain/drug effects , Membrane Glycoproteins/genetics , Receptors, Immunologic/genetics , Tetrahydronaphthalenes/pharmacology , Up-Regulation/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Bexarotene , Brain/metabolism , Disease Models, Animal , Gene Expression Regulation/drug effects , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Phagocytosis/drug effects , Receptors, Immunologic/metabolism , Retinoid X Receptors/metabolism , Sequence Analysis, RNA
12.
J Alzheimers Dis ; 21(3): 1013-21, 2010.
Article in English | MEDLINE | ID: mdl-20693631

ABSTRACT

Since previous observations indicated that the urea cycle may have a role in the Alzheimer's disease (AD) process, we set out to quantify the expression of each gene involved in the urea cycle in control and AD brains and establish whether these genes could be genetic determinants of AD. We first confirmed that all the urea cycle enzyme genes are expressed in the AD brain. The expression of arginase 2 was greater in the AD brain than in the control brain. The presence of the rare arginase 2 allele rs742869 was associated with an increase in the risk of AD in men and with an earlier age-at-onset for both genders. None of the other genes in the pathway appeared to be differentially expressed in the AD brain or act as genetic determinants of the disease.


Subject(s)
Alzheimer Disease/genetics , Arginase/genetics , Brain/metabolism , Urea/metabolism , Aged , Aged, 80 and over , Alleles , Alzheimer Disease/metabolism , Arginase/metabolism , Case-Control Studies , Chi-Square Distribution , Female , Genetic Association Studies , Genotype , Humans , Male , Middle Aged , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...