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










Publication year range
2.
Epigenetics Chromatin ; 16(1): 12, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37101286

ABSTRACT

BACKGROUND: Diabetic cardiomyopathy (DCM) is a leading cause of death in diabetic patients. Hyperglycemic myocardial microenvironment significantly alters chromatin architecture and the transcriptome, resulting in aberrant activation of signaling pathways in a diabetic heart. Epigenetic marks play vital roles in transcriptional reprogramming during the development of DCM. The current study is aimed to profile genome-wide DNA (hydroxy)methylation patterns in the hearts of control and streptozotocin (STZ)-induced diabetic rats and decipher the effect of modulation of DNA methylation by alpha-ketoglutarate (AKG), a TET enzyme cofactor, on the progression of DCM. METHODS: Diabetes was induced in male adult Wistar rats with an intraperitoneal injection of STZ. Diabetic and vehicle control animals were randomly divided into groups with/without AKG treatment. Cardiac function was monitored by performing cardiac catheterization. Global methylation (5mC) and hydroxymethylation (5hmC) patterns were mapped in the Left ventricular tissue of control and diabetic rats with the help of an enrichment-based (h)MEDIP-sequencing technique by using antibodies specific for 5mC and 5hmC. Sequencing data were validated by performing (h)MEDIP-qPCR analysis at the gene-specific level, and gene expression was analyzed by qPCR. The mRNA and protein expression of enzymes involved in the DNA methylation and demethylation cycle were analyzed by qPCR and western blotting. Global 5mC and 5hmC levels were also assessed in high glucose-treated DNMT3B knockdown H9c2 cells. RESULTS: We found the increased expression of DNMT3B, MBD2, and MeCP2 with a concomitant accumulation of 5mC and 5hmC, specifically in gene body regions of diabetic rat hearts compared to the control. Calcium signaling was the most significantly affected pathway by cytosine modifications in the diabetic heart. Additionally, hypermethylated gene body regions were associated with Rap1, apelin, and phosphatidyl inositol signaling, while metabolic pathways were most affected by hyperhydroxymethylation. AKG supplementation in diabetic rats reversed aberrant methylation patterns and restored cardiac function. Hyperglycemia also increased 5mC and 5hmC levels in H9c2 cells, which was normalized by DNMT3B knockdown or AKG supplementation. CONCLUSION: This study demonstrates that reverting hyperglycemic damage to cardiac tissue might be possible by erasing adverse epigenetic signatures by supplementing epigenetic modulators such as AKG along with an existing antidiabetic treatment regimen.


Subject(s)
Diabetes Mellitus, Experimental , Epigenesis, Genetic , Male , Rats , Animals , Ketoglutaric Acids , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Rats, Wistar , DNA Methylation , DNA
3.
Stem Cell Res ; 67: 103041, 2023 03.
Article in English | MEDLINE | ID: mdl-36764067

ABSTRACT

A pair of Down syndrome (DS) human iPSCs (hiPSCs) and isogenic euploid hiPSCs generated by using an integration-free Sendai viral vector system showed trisomy 21 (47; XY) and typical (46; XY) karyotype respectively. Pluripotency of both hiPSC lines was confirmed by pluripotency marker expression and three germ layer differentiation potentials.


Subject(s)
Down Syndrome , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Down Syndrome/metabolism , Cell Differentiation , Karyotype , Sendai virus
4.
Front Genet ; 13: 1007519, 2022.
Article in English | MEDLINE | ID: mdl-36313423

ABSTRACT

Impaired neurogenesis in Down syndrome (DS) is characterized by reduced neurons, increased glial cells, and delayed cortical lamination. However, the underlying cause for impaired neurogenesis in DS is not clear. Using both human and mouse iPSCs, we demonstrate that DS impaired neurogenesis is due to biphasic cell cycle dysregulation during the generation of neural progenitors from iPSCs named the "neurogenic stage" of neurogenesis. Upon neural induction, DS cells showed reduced proliferation during the early phase followed by increased proliferation in the late phase of the neurogenic stage compared to control cells. While reduced proliferation in the early phase causes reduced neural progenitor pool, increased proliferation in the late phase leads to delayed post mitotic neuron generation in DS. RNAseq analysis of late-phase DS progenitor cells revealed upregulation of S phase-promoting regulators, Notch, Wnt, Interferon pathways, and REST, and downregulation of several genes of the BAF chromatin remodeling complex. NFIB and POU3F4, neurogenic genes activated by the interaction of PAX6 and the BAF complex, were downregulated in DS cells. ChIPseq analysis of late-phase neural progenitors revealed aberrant PAX6 binding with reduced promoter occupancy in DS cells. Together, these data indicate that impaired neurogenesis in DS is due to biphasic cell cycle dysregulation during the neurogenic stage of neurogenesis.

5.
Stem Cell Res ; 64: 102890, 2022 10.
Article in English | MEDLINE | ID: mdl-35961104

ABSTRACT

Human mouse chimeric models are valuable tools to develop in-vivo disease models. However, in-vivo detection of human cells limits their analysis. To facilitate in-vivo modeling of Down syndrome (DS), we generated a stable AAVS1-EGFP isogenic pair of DS human iPSCs by zinc finger mediated genetic engineering of the AAVS1 locus. These lines overcome the limitation of reporter human iPSCs generated using random integration, which may not express reporter gene in all tissues due to heterochromatin-induced gene silencing. These reporter cell lines provide a valuable tool to facilitate in-vivo tracking of the graft cell integration, differentiation, and distinction from host cells.


Subject(s)
Down Syndrome , Induced Pluripotent Stem Cells , Mice , Animals , Humans , Down Syndrome/genetics , Down Syndrome/metabolism , Heterochromatin/metabolism , Transfection , Induced Pluripotent Stem Cells/metabolism , Cell Line , Genes, Reporter , Cell Differentiation
6.
Stem Cell Res ; 61: 102771, 2022 05.
Article in English | MEDLINE | ID: mdl-35381521

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) clones NSi001-A, NSi001-B, and NSi001-C were generated from a female individual of Indian origin having Robertsonian translocation down syndrome (DS) by reprogramming peripheral blood mononuclear cells (PBMCs) using integration-free Sendai viral vectors. The established hiPSCs clones had karyotype similar to the patient sample with Robertsonian translocation [46, XX rob (14;21)], normal ES-like morphology, expression of pluripotency markers, and potential for three germ layer differentiation, i.e., ectoderm, mesoderm, and endoderm.


Subject(s)
Down Syndrome , Induced Pluripotent Stem Cells , Cells, Cultured , Clone Cells , Down Syndrome/genetics , Down Syndrome/metabolism , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear , Translocation, Genetic
7.
PLoS Genet ; 13(5): e1006757, 2017 May.
Article in English | MEDLINE | ID: mdl-28498870

ABSTRACT

Zygotic gene expression programs control cell differentiation in vertebrate development. In Xenopus, these programs are initiated by local induction of regulatory genes through maternal signaling activities in the wake of zygotic genome activation (ZGA) at the midblastula transition (MBT). These programs lay down the vertebrate body plan through gastrulation and neurulation, and are accompanied by massive changes in chromatin structure, which increasingly constrain cellular plasticity. Here we report on developmental functions for Brahma related gene 1 (Brg1), a key component of embyronic SWI/SNF chromatin remodeling complexes. Carefully controlled, global Brg1 protein depletion in X. tropicalis and X. laevis causes embryonic lethality or developmental arrest from gastrulation on. Transcriptome analysis at late blastula, before development becomes arrested, indicates predominantly a role for Brg1 in transcriptional activation of a limited set of genes involved in pattern specification processes and nervous system development. Mosaic analysis by targeted microinjection defines Brg1 as an essential amplifier of gene expression in dorsal (BCNE/Nieuwkoop Center) and ventral (BMP/Vent) signaling centers. Moreover, Brg1 is required and sufficient for initiating axial patterning in cooperation with maternal Wnt signaling. In search for a common denominator of Brg1 impact on development, we have quantitatively filtered global mRNA fluctuations at MBT. The results indicate that Brg1 is predominantly required for genes with the highest burst of transcriptional activity. Since this group contains many key developmental regulators, we propose Brg1 to be responsible for raising their expression above threshold levels in preparation for embryonic patterning.


Subject(s)
Adenosine Triphosphatases/genetics , DNA Helicases/genetics , Transcription, Genetic , Animals , Blastula/growth & development , Blastula/metabolism , Cell Differentiation/genetics , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA Helicases/biosynthesis , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Genome , Maternal Inheritance/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Xenopus/genetics , Xenopus/growth & development , Zygote/growth & development , Zygote/metabolism
8.
PLoS One ; 11(3): e0152471, 2016.
Article in English | MEDLINE | ID: mdl-27023444

ABSTRACT

In Down syndrome (DS) or trisomy of chromosome 21, the ß-amyloid (Aß) peptide product of the amyloid precursor protein (APP) is present in excess. Evidence points to increased APP gene dose and Aß as playing a critical role in cognitive difficulties experienced by people with DS. Particularly, Aß is linked to the late-life emergence of dementia as associated with neuropathological markers of Alzheimer's disease (AD). At present, no treatment targets Aß-related pathogenesis in people with DS. Herein we used a vaccine containing the Aß 1-15 peptide embedded into liposomes together with the adjuvant monophosphoryl lipid A (MPLA). Ts65Dn mice, a model of DS, were immunized with the anti-Aß vaccine at 5 months of age and were examined for cognitive measures at 8 months of age. The status of basal forebrain cholinergic neurons and brain levels of APP and its proteolytic products were measured. Immunization of Ts65Dn mice resulted in robust anti-Aß IgG titers, demonstrating the ability of the vaccine to break self-tolerance. The vaccine-induced antibodies reacted with Aß without detectable binding to either APP or its C-terminal fragments. Vaccination of Ts65Dn mice resulted in a modest, but non-significant reduction in brain Aß levels relative to vehicle-treated Ts65Dn mice, resulting in similar levels of Aß as diploid (2N) mice. Importantly, vaccinated Ts65Dn mice showed resolution of memory deficits in the novel object recognition and contextual fear conditioning tests, as well as reduction of cholinergic neuron atrophy. No treatment adverse effects were observed; vaccine did not result in inflammation, cellular infiltration, or hemorrhage. These data are the first to show that an anti-Aß immunotherapeutic approach may act to target Aß-related pathology in a mouse model of DS.


Subject(s)
Amyloid beta-Peptides/immunology , Cognition Disorders/complications , Cognition Disorders/drug therapy , Down Syndrome/complications , Down Syndrome/drug therapy , Vaccines/therapeutic use , Amyloid beta-Peptides/genetics , Animals , Animals, Newborn , Antibodies/metabolism , Atrophy , Behavior, Animal , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Cholinergic Neurons/metabolism , Disease Models, Animal , Gene Expression Regulation , Hemorrhage/pathology , Inflammation/pathology , Male , Memory , Mice, Transgenic , Septal Nuclei/pathology , Vaccination
9.
F1000Res ; 4: 269, 2015.
Article in English | MEDLINE | ID: mdl-26664707

ABSTRACT

Recently, in studies examining fibroblasts obtained from the tissues of one set of monozygotic twins (i.e. fetuses derived from the same egg) discordant for trisomy 21 (Down syndrome; DS), Letourneau et al., ( )reported the presence of a defined pattern of dysregulation within specific genomic domains they referred to as Gene Expression Dysregulated Domains (GEDDs). GEDDs were described as alternating segments of increased or decreased gene expression affecting all chromosomes. Strikingly, GEDDs in fibroblasts were largely conserved in induced pluripotent cells (iPSCs) generated from the twin's fibroblasts as well as in fibroblasts from the Ts65Dn mouse model of DS. Our recent analysis failed to find GEDDs. We reexamined the human iPSCs RNAseq data from Letourneau et al., and data from this same research group published earlier examining iPSCs from the same monozygotic twins. An independent analysis of RNAseq data from Ts65Dn fibroblasts also failed to confirm presence of GEDDs. Our analysis questions the validity of GEDDs in DS.

10.
Biores Open Access ; 3(1): 1-8, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24570840

ABSTRACT

BAF chromatin remodeling complexes containing the BRG1 protein have been shown to be not only essential for early embryonic development, but also paramount in enhancing the efficiency of reprogramming somatic cells to pluripotency mediated by four transcription factors. To investigate the role of BRG1 in regulating pluripotency, we found that Oct4 and Nanog levels were increased immediately after BRG1 knockdown. While Nanog levels remained elevated over the investigated time period, Oct4 levels decreased at later time points. Additionally, OCT4 target genes were also found to be upregulated upon Brg1 knockdown. SiRNA-mediated BRG1 knockdown in embryonic stem (ES) cells led to Oct4 and Nanog upregulation, whereas F9 cells showed primarily Oct4 upregulation. BRG1 knockdown upregulated the expression of differentiation markers in mouse ES cells as well as differentiated morphology under reduced leukemia inhibitory factor conditions. Our results show that BRG1 plays an important role in maintaining pluripotency by fine-tuning the expression level of Oct4 and other pluripotency-associated genes.

11.
Nat Cell Biol ; 15(9): 1089-97, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23934214

ABSTRACT

Oct4A is a core component of the regulatory network of pluripotent cells, and by itself can reprogram neural stem cells into pluripotent cells in mice and humans. However, its role in defining totipotency and inducing pluripotency during embryonic development is still unclear. We genetically eliminated maternal Oct4A using a Cre/loxP approach in mouse and found that the establishment of totipotency was not affected, as shown by the generation of live pups. After complete inactivation of both maternal and zygotic Oct4A expression, the embryos still formed Oct4-GFP- and Nanog-expressing inner cell masses, albeit non-pluripotent, indicating that Oct4A is not a determinant for the pluripotent cell lineage separation. Interestingly, Oct4A-deficient oocytes were able to reprogram fibroblasts into pluripotent cells. Our results clearly demonstrate that, in contrast to its role in the maintenance of pluripotency, maternal Oct4A is not crucial for either the establishment of totipotency in embryos, or the induction of pluripotency in somatic cells using oocytes.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Octamer Transcription Factor-3/genetics , Oocytes/metabolism , Totipotent Stem Cells/metabolism , Animals , Cell Lineage/genetics , Cells, Cultured , Cellular Reprogramming/genetics , Embryo, Mammalian , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fibroblasts/cytology , Genes, Reporter , Green Fluorescent Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Nanog Homeobox Protein , Octamer Transcription Factor-3/deficiency , Oocytes/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pregnancy , Protein Isoforms/deficiency , Protein Isoforms/genetics , Signal Transduction , Totipotent Stem Cells/cytology
12.
Hepatology ; 53(5): 1651-61, 2011 May.
Article in English | MEDLINE | ID: mdl-21400558

ABSTRACT

UNLABELLED: Death receptor-mediated apoptosis of hepatocytes contributes to hepatitis and fulminant liver failure. MicroRNAs (miRNAs), 19-25 nucleotide-long noncoding RNAs, have been implicated in the posttranscriptional regulation of the various apoptotic pathways. Here we report that global loss of miRNAs in hepatic cells leads to increased cell death in a model of FAS/CD95 receptor-induced apoptosis. miRNA profiling of murine liver identified 11 conserved miRNAs, which were up-regulated in response to FAS-induced fulminant liver failure. We show that ectopic expression of miR-221, one of the highly up-regulated miRNAs in response to apoptosis, protects primary hepatocytes and hepatoma cells from apoptosis. Importantly, in vivo overexpression of miR-221 by adeno-associated virus serotype 8 (AAV8) delays FAS-induced fulminant liver failure in mice. We additionally demonstrate that miR-221 regulates hepatic expression of p53 up-regulated modulator of apoptosis (Puma), a well-known proapoptotic member of the Bcl2 protein family. CONCLUSION: We identified miR-221 as a potent posttranscriptional regulator of FAS-induced apoptosis. miR-221 may serve as a potential therapeutic target for the treatment of hepatitis and liver failure.


Subject(s)
Liver Failure, Acute/etiology , MicroRNAs/physiology , fas Receptor/physiology , Animals , Apoptosis , Hepatocytes , Mice , Mice, Inbred BALB C
13.
Cell ; 141(6): 943-55, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20550931

ABSTRACT

Reprogramming of somatic cells achieved by combination of the four transcription factors Oct4, Sox2, Klf4, and c-Myc has very low efficiency. To increase the reprogramming efficiency and better understand the process, we sought to identify factors that mediate reprogramming with higher efficiency. We established an assay to screen nuclear fractions from extracts of pluripotent mouse cells based on Oct4 reactivation. Using proteomics, we identified components of the ATP-dependent BAF chromatin-remodeling complex, which significantly increases reprogramming efficiency when used together with the four factors. The reprogrammed cells could transmit to the germline and exhibited pluripotency. Reprogramming remained highly efficient when c-Myc was not present but BAF components were overexpressed. BAF complex components mediate this effect by facilitating enhanced Oct4 binding to target promoters during reprogramming. Thus, somatic cell reprogramming using chromatin-remodeling molecules represents an efficient method of generating reprogrammed cells.


Subject(s)
Cellular Reprogramming , Chromatin Assembly and Disassembly , Animals , Cell Line , Chromatin/metabolism , DNA Helicases/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Kruppel-Like Factor 4 , Mice , Nuclear Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcription Factors/metabolism
14.
Eur J Biochem ; 269(9): 2294-9, 2002 May.
Article in English | MEDLINE | ID: mdl-11985611

ABSTRACT

During the development of multicellular organisms, both transient and stable gene expression patterns have to be established in a precisely orchestrated sequence. Evidence from diverse model organisms indicates that this epigenetic program involves not only transcription factors, but also the local structure, composition, and modification of chromatin, which define and maintain the accessibility and transcriptional competence of the nucleosomal DNA template. A paradigm for the interdependence of development and chromatin is constituted by the mechanisms controlling the specification and differentiation of the skeletal muscle cell lineage in vertebrates, which is the topic of this review.


Subject(s)
Chromatin/metabolism , Muscle, Skeletal/embryology , Acetylation , Animals , Basic Helix-Loop-Helix Transcription Factors , Cell Differentiation , DNA Methylation , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Histones/metabolism , Humans , MyoD Protein/physiology , Transcription Factors/physiology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...