Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
2.
ASN Neuro ; 8(5)2016 Oct.
Article in English | MEDLINE | ID: mdl-27683878

ABSTRACT

The transcriptional program that controls oligodendrocyte maturation and central nervous system (CNS) myelination has not been fully characterized. In this study, we use high-throughput RNA sequencing to analyze how the loss of a key transcription factor, zinc finger protein 191 (ZFP191), results in oligodendrocyte development abnormalities and CNS hypomyelination. Using a previously described mutant mouse that is deficient in ZFP191 protein expression (Zfp191null), we demonstrate that key transcripts are reduced in the whole brain as well as within oligodendrocyte lineage cells cultured in vitro To determine whether the loss of myelin seen in Zfp191null mice contributes indirectly to these perturbations, we also examined the transcriptome of a well-characterized mouse model of hypomyelination, in which the myelin structural protein myelin basic protein (MBP) is deficient. Interestingly, Mbpshi (shiverer) mice had far fewer transcripts perturbed with the loss of myelin alone. This study demonstrates that the loss of ZFP191 disrupts expression of genes involved in oligodendrocyte maturation and myelination, largely independent from the loss of myelin. Nevertheless, hypomyelination in both mouse mutants results in the perturbation of lipid synthesis pathways, suggesting that oligodendrocytes have a feedback system that allows them to regulate myelin lipid synthesis depending on their myelinating state. The data presented are of potential clinical relevance as the human orthologs of the Zfp191 and MBP genes reside on a region of Chromosome 18 that is deleted in childhood leukodystrophies.

3.
Cell Res ; 26(9): 1048-61, 2016 09.
Article in English | MEDLINE | ID: mdl-27502025

ABSTRACT

Gene editing in non-human primates may lead to valuable models for exploring the etiologies and therapeutic strategies of genetically based neurological disorders in humans. However, a monkey model of neurological disorders that closely mimics pathological and behavioral deficits in humans has not yet been successfully generated. Microcephalin 1 (MCPH1) is implicated in the evolution of the human brain, and MCPH1 mutation causes microcephaly accompanied by mental retardation. Here we generated a cynomolgus monkey (Macaca fascicularis) carrying biallelic MCPH1 mutations using transcription activator-like effector nucleases. The monkey recapitulated most of the important clinical features observed in patients, including marked reductions in head circumference, premature chromosome condensation (PCC), hypoplasia of the corpus callosum and upper limb spasticity. Moreover, overexpression of MCPH1 in mutated dermal fibroblasts rescued the PCC syndrome. This monkey model may help us elucidate the role of MCPH1 in the pathogenesis of human microcephaly and better understand the function of this protein in the evolution of primate brain size.


Subject(s)
Microcephaly/pathology , Transcription Activator-Like Effector Nucleases/metabolism , Animals , Base Sequence , Behavior, Animal , Brain/pathology , Disease Models, Animal , Female , Genotype , Humans , Macaca fascicularis , Magnetic Resonance Imaging , Mutation , Phenotype
4.
Biomaterials ; 50: 56-66, 2015 May.
Article in English | MEDLINE | ID: mdl-25736496

ABSTRACT

Renal resident mesenchymal stem cells (MSCs) are important regulators of kidney homeostasis, repair or regeneration. However, natural distribution and the starting population properties of these cells remain elusive because of the lack of specific markers. Here, we identified post-natal kidney derived Nestin(+) cells that fulfilled all of the criteria as a mesenchymal stem cell. These isolated Nestin(+) cells expressed the typical cell-surface marker of MSC, including Sca-1, CD44, CD106, NG2 and PDGFR-α. They were capable of self-renewal, possessed high clonogenic potential and extensive proliferation for more than 30 passages. Under appropriate differentiation conditions, these cells could differentiate into adipocytes, osteocytes, chondrocytes and podocytes. After intravenous injection into acute kidney injury mice, Nestin(+) cells contributed to functional improvement by significantly decreasing the peak level of serum creatinine and BUN, and reducing the damaged cell apoptosis. Furthermore, conditioned medium from Nestin(+) cells could protect against ischemic acute renal failure partially through paracrine factor VEGF. Taken together, our findings indicate that renal resident Nestin(+) MSCs can be derived, propagated, differentiated, and repair the acute kidney injury, which may shed new light on understanding MSCs biology and developing cell replacement therapies for kidney disease.


Subject(s)
Acute Kidney Injury/therapy , Ischemia/therapy , Kidney/blood supply , Kidney/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Nestin/metabolism , Acute Kidney Injury/complications , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Separation , Clone Cells , Colony-Forming Units Assay , Culture Media, Conditioned/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Green Fluorescent Proteins/metabolism , Ischemia/complications , Ischemia/pathology , Ischemia/physiopathology , Kidney/metabolism , Kidney/physiopathology , Kidney Function Tests , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Multipotent Stem Cells/cytology , Multipotent Stem Cells/drug effects , Protective Agents/metabolism , Recovery of Function/drug effects , Time Factors , Vascular Endothelial Growth Factor A/metabolism
5.
Cell Res ; 24(12): 1466-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25418539

ABSTRACT

The ability to identify and isolate lineage-specific stem cells from adult tissues could facilitate cell replacement therapy. Leydig cells (LCs) are the primary source of androgen in the mammalian testis, and the prospective identification of stem Leydig cells (SLCs) may offer new opportunities for treating testosterone deficiency. Here, in a transgenic mouse model expressing GFP driven by the Nestin (Nes) promoter, we observed Nes-GFP+ cells located in the testicular interstitial compartment where SLCs normally reside. We showed that these Nes-GFP+ cells expressed LIFR and PDGFR-α, but not LC lineage markers. We further observed that these cells were capable of clonogenic self-renewal and extensive proliferation in vitro and could differentiate into neural or mesenchymal cell lineages, as well as LCs, with the ability to produce testosterone, under defined conditions. Moreover, when transplanted into the testes of LC-disrupted or aging models, the Nes-GFP+ cells colonized the interstitium and partially increased testosterone production, and then accelerated meiotic and post-meiotic germ cell recovery. In addition, we further demonstrated that CD51 might be a putative cell surface marker for SLCs, similar with Nestin. Taken together, these results suggest that Nes-GFP+ cells from the testis have the characteristics of SLCs, and our study would shed new light on developing stem cell replacement therapy for testosterone deficiency.


Subject(s)
Adult Stem Cells/transplantation , Leydig Cells/pathology , Leydig Cells/transplantation , Nestin/analysis , Testis/cytology , Testis/pathology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Aging , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Expression , Integrin alphaV/analysis , Leydig Cells/cytology , Leydig Cells/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Nestin/genetics , Prospective Studies , Spermatogenesis , Testis/physiology , Testosterone/metabolism
6.
Neuroreport ; 25(10): 782-7, 2014 Jul 09.
Article in English | MEDLINE | ID: mdl-24918459

ABSTRACT

Nestin knockout leads to embryonic lethality and self-renewal deficiency in neural stem cells (NSCs). However, how nestin maintains self-renewal remains uncertain. Here, we used the dosage effect of nestin in heterozygous mice (Nes+/-) to study self-renewal of NSCs. With existing extracellular signaling in vivo or in vitro, nestin levels do not affect proliferation ability or apoptosis when compared between Nes+/- and Nes+/+ NSCs. However, self-renewal ability of Nes+/- NSCs is impaired when plated at a low cell density and completely lost at a clonal density. This deficiency in self-renewal at a clonal density is rescued using a medium conditioned by Nes+/+ NSCs. In addition, the Akt signaling pathway is altered at low density and reversed by conditioned medium. Our data show that secreted factors contribute toward maintaining self-renewal of NSCs by nestin, potentially through Akt signaling.


Subject(s)
Cell Proliferation/genetics , Nestin/genetics , Neural Stem Cells/physiology , Animals , Apoptosis , Cell Count , Cells, Cultured , Extracellular Space/metabolism , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
7.
Genome Res ; 24(2): 267-80, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24310002

ABSTRACT

Both diffusible factors acting in trans and chromatin components acting in cis are implicated in gene regulation, but the extent to which either process causally determines a cell's transcriptional identity is unclear. We recently used cell fusion to define a class of silent genes termed "cis-silenced" (or "occluded") genes, which remain silent even in the presence of trans-acting transcriptional activators. We further showed that occlusion of lineage-inappropriate genes plays a critical role in maintaining the transcriptional identities of somatic cells. Here, we present, for the first time, a comprehensive map of occluded genes in somatic cells. Specifically, we mapped occluded genes in mouse fibroblasts via fusion to a dozen different rat cell types followed by whole-transcriptome profiling. We found that occluded genes are highly prevalent and stable in somatic cells, representing a sizeable fraction of silent genes. Occluded genes are also highly enriched for important developmental regulators of alternative lineages, consistent with the role of occlusion in safeguarding cell identities. Alongside this map, we also present whole-genome maps of DNA methylation and eight other chromatin marks. These maps uncover a complex relationship between chromatin state and occlusion. Furthermore, we found that DNA methylation functions as the memory of occlusion in a subset of occluded genes, while histone deacetylation contributes to the implementation but not memory of occlusion. Our data suggest that the identities of individual cell types are defined largely by the occlusion status of their genomes. The comprehensive reference maps reported here provide the foundation for future studies aimed at understanding the role of occlusion in development and disease.


Subject(s)
Gene Expression Regulation , Gene Silencing , Regulatory Sequences, Nucleic Acid , Trans-Activators/genetics , Transcription, Genetic , Animals , Cell Fusion , Cell Line , Chromatin/genetics , DNA Methylation/genetics , Genome , Histones/genetics , Histones/metabolism , Mice , Rats
8.
Cell Rep ; 6(1): 231-244, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24373966

ABSTRACT

Hematopoietic stem cell differentiation involves the silencing of self-renewal genes and induction of a specific transcriptional program. Identification of multiple covalent cytosine modifications raises the question of how these derivatized bases influence stem cell commitment. Using a replicative primary human hematopoietic stem/progenitor cell differentiation system, we demonstrate dynamic changes of 5-hydroxymethylcytosine (5-hmC) during stem cell commitment and differentiation to the erythroid lineage. Genomic loci that maintain or gain 5-hmC density throughout erythroid differentiation contain binding sites for erythroid transcription factors and several factors not previously recognized as erythroid-specific factors. The functional importance of 5-hmC was demonstrated by impaired erythroid differentiation, with augmentation of myeloid potential, and disrupted 5-hmC patterning in leukemia patient-derived CD34+ stem/early progenitor cells with TET methylcytosine dioxygenase 2 (TET2) mutations. Thus, chemical conjugation and affinity purification of 5-hmC-enriched sequences followed by sequencing serve as resources for deciphering functional implications for gene expression during stem cell commitment and differentiation along a particular lineage.


Subject(s)
DNA Methylation , Erythroid Cells/metabolism , Erythropoiesis/genetics , Regulatory Sequences, Nucleic Acid , 5-Methylcytosine/analogs & derivatives , Antigens, CD34/genetics , Antigens, CD34/metabolism , Cells, Cultured , Cytosine/analogs & derivatives , Cytosine/analysis , Dioxygenases/genetics , Dioxygenases/metabolism , Erythroid Cells/cytology , Erythroid Cells/drug effects , Histones/metabolism , Humans , Mutation , Transcription Factors/metabolism
9.
Biomaterials ; 34(6): 1701-11, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23218839

ABSTRACT

Pluripotent stem cells, including embryonic stem (ES) and induced pluripotent stem (iPS) cells, serve as unlimited resources for cell replacement therapy and tissue engineering because such cells are capable of extensive proliferation in vitro and can give rise to lineages that represent any of the three embryonic germ layers. However, in the context of the in vivo behavior of cell transplants, key challenges need to be addressed and essential strategies should be developed before stem cells can be used in clinical practice. In the present study, we modified mouse ES/iPS cells to contain a suicide gene, deltaTK or CodA, under the transcriptional control of the EF1α or Nanog promoter. The suicide gene was introduced via lentivirus transduction without interfering with their self-renewal and pluripotency characteristics. We found that EF1α promoter-controlled deltaTK/CodA expression efficiently eliminated pluripotent stem cells and their derivatives both in vitro and in vivo. When the suicide gene was under the control of the Nanog promoter, tumor-initiating undifferentiated pluripotent stem cells were selectively ablated in vitro after prodrug treatment. These results indicate that modification of pluripotent stem cells with a suicide gene prior to transplantation offers a safe manner by which wayward stem cells, and their progeny, can be controlled in vivo. Our approach will render the clinical application of human pluripotent stem cells increasingly possible.


Subject(s)
Neoplasms/pathology , Pluripotent Stem Cells/cytology , Animals , Base Sequence , Cell Death , DNA Primers , Fluorescent Antibody Technique , Mice , Reverse Transcriptase Polymerase Chain Reaction
10.
Cancer Res ; 72(18): 4714-23, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22815530

ABSTRACT

Epigenetic changes in pediatric neuroblastoma may contribute to the aggressive pathophysiology of this disease, but little is known about the basis for such changes. In this study, we examined a role for the DNA methyltransferase DNMT3B, in particular, the truncated isoform DNMT3B7, which is generated frequently in cancer. To investigate if aberrant DNMT3B transcripts alter DNA methylation, gene expression, and phenotypic character in neuroblastoma, we measured DNMT3B expression in primary tumors. Higher levels of DNMT3B7 were detected in differentiated ganglioneuroblastomas compared to undifferentiated neuroblastomas, suggesting that expression of DNMT3B7 may induce a less aggressive clinical phenotype. To test this hypothesis, we investigated the effects of enforced DNMT3B7 expression in neuroblastoma cells, finding a significant inhibition of cell proliferation in vitro and angiogenesis and tumor growth in vivo. DNMT3B7-positive cells had higher levels of total genomic methylation and a dramatic decrease in expression of the FOS and JUN family members that comprise AP1 transcription factors. Consistent with an established antagonistic relationship between AP1 expression and retinoic acid receptor activity, increased differentiation was seen in the DNMT3B7-expressing neuroblastoma cells following treatment with all-trans retinoic acid (ATRA) compared to controls. Our results indicate that DNMT3B7 modifies the epigenome in neuroblastoma cells to induce changes in gene expression, inhibit tumor growth, and increase sensitivity to ATRA.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methylation/genetics , Gene Expression Regulation, Neoplastic/genetics , Neuroblastoma/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/metabolism , Drug Resistance, Neoplasm/genetics , Epigenesis, Genetic , Female , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Nude , Neuroblastoma/metabolism , Neuroblastoma/pathology , Tretinoin/pharmacology , Xenograft Model Antitumor Assays , DNA Methyltransferase 3B
11.
Mol Cell ; 46(2): 159-70, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22445485

ABSTRACT

It is a long-held paradigm that cell fusion reprograms gene expression but the extent of reprogramming and whether it is affected by the cell types employed remain unknown. We recently showed that the silencing of somatic genes is attributable to either trans-acting cellular environment or cis-acting chromatin context. Here, we examine how trans- versus cis-silenced genes in a somatic cell type behave in fusions to another somatic cell type or to embryonic stem cells (ESCs). We demonstrate that while reprogramming of trans-silenced somatic genes occurs in both cases, reprogramming of cis-silenced somatic genes occurs only in somatic-ESC fusions. Importantly, ESCs reprogram the somatic genome in two distinct phases: trans-reprogramming occurs rapidly, independent of DNA replication, whereas cis-reprogramming occurs with slow kinetics requiring DNA replication. We also show that pluripotency genes Oct4 and Nanog are cis-silenced in somatic cells. We conclude that cis-reprogramming capacity is a fundamental feature distinguishing ESCs from somatic cells.


Subject(s)
Cell Fusion , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , DNA Replication , Gene Silencing , Kinetics , Mice
12.
Biomaterials ; 33(11): 3195-204, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22269649

ABSTRACT

The generation of human induced pluripotent stem cells (hiPSCs) opens a prospect for regenerative medicine. However, transplantation of somatic cells derived from hiPSCs still harbor many risks such as cells' incorrect differentiation or over-proliferation, and the worst, tumor formation. Therefore, it's essential to ravel out these obstacles before their clinical application. Herein, we genetically modified hiPSCs and human embryonic stem cells (hESCs) with a truncated herpes simplex virus delta thymidine kinase (deltaTK) gene driven by EF1α or Nanog promoter to selectively ablate wayward pluripotent stem cells. The results showed that insertion of deltaTK gene did not alter their pluripotency and self-renewal capacity but rendered them sensitive to ganciclovir, which induced elimination of deltaTK(+) cells in vitro in a dose and time-dependent manner, most importantly, facilitated both prevention and ablation of tumors in vivo. Furthermore, comparative analysis between transduced hiPSCs and hESCs showed that there was no difference in ganciclovir sensitivity between them. This approach may help to develop safety strategies for clinical application of hiPSCs in regenerative medicine in the future.


Subject(s)
Cell Transformation, Neoplastic/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Genes, Transgenic, Suicide/genetics , Genetic Enhancement/methods , Thymidine Kinase/genetics , Animals , Apoptosis/genetics , Cell Survival/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Homeodomain Proteins/genetics , Humans , Mice , Mice, SCID , Nanog Homeobox Protein
13.
Cell Res ; 22(5): 848-58, 2012 May.
Article in English | MEDLINE | ID: mdl-22124232

ABSTRACT

The progressive restriction of cell fate during lineage differentiation is a poorly understood phenomenon despite its ubiquity in multicellular organisms. We recently used a cell fusion assay to define a mode of epigenetic silencing that we termed "occlusion", wherein affected genes are silenced by cis-acting chromatin mechanisms irrespective of whether trans-acting transcriptional activators are present. We hypothesized that occlusion of lineage-inappropriate genes could contribute to cell fate restriction. Here, we test this hypothesis by introducing bacterial artificial chromosomes (BACs), which are devoid of chromatin modifications necessary for occlusion, into mouse fibroblasts. We found that BAC transgenes corresponding to occluded endogenous genes are expressed in most cases, whereas BAC transgenes corresponding to silent but non-occluded endogenous genes are not expressed. This indicates that the cellular milieu in trans supports the expression of most occluded genes in fibroblasts, and that the silent state of these genes is solely the consequence of occlusion in cis. For the BAC corresponding to the occluded myogenic master regulator Myf5, expression of the Myf5 transgene on the BAC triggered fibroblasts to acquire a muscle-like phenotype. These results provide compelling evidence for a critical role of gene occlusion in cell fate restriction.


Subject(s)
Gene Silencing , Animals , Cell Line , Cell Lineage , Chromatin/genetics , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Bacterial/metabolism , Fibroblasts/metabolism , Mice , Models, Genetic , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Trans-Activators/genetics
14.
Int J Biochem Cell Biol ; 44(2): 377-84, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22120652

ABSTRACT

Mesenchymal stem cells (MSCs) are the common progenitors of osteoblasts and adipocytes. A reciprocal relationship exists between osteogenesis and adipogenesis in the bone marrow, and the identification of signaling pathways that stimulate MSC osteogenesis at the expense of adipogenesis is of great importance from the viewpoint of developing new therapeutic treatments for bone loss. The adipogenic transcription factor peroxisome proliferator-activated receptor γ (PPARγ) has been reported to play a vital role in modulating mesenchymal lineage allocation within the bone marrow compartment, stimulating adipocyte development at the expense of osteoblast differentiation. Hence, PPARγ may be a valuable target for drugs intended to enhance bone mass. However, little direct evidence is available for the role played by PPARγ in human mesenchymal lineage allocation. In this study, using human MSCs as an in vitro model, we showed that the two isoforms of PPARγ, PPARγ1 and PPARγ2, were differentially induced during hMSC adipogenesis, whereas only PPARγ1 was detected during osteogenesis. BADGE and GW9662, two potential antagonists of PPARγ, as well as lentivirus-mediated knockdown of PPARγ, inhibited hMSC adipogenesis but did not significantly affect osteogenesis. PPARγ knockdown did not significantly influence the expression level of the osteogenic transcription factor Runx2. Together, these results suggest that PPARγ is not the master factor regulating mesenchymal lineage determination in human bone marrow.


Subject(s)
Adipogenesis/physiology , Mesenchymal Stem Cells/metabolism , Osteogenesis/physiology , PPAR gamma/metabolism , Adipocytes/metabolism , Adipogenesis/drug effects , Cell Differentiation/physiology , Cell Line , Down-Regulation , Humans , Osteoblasts/metabolism , Osteogenesis/drug effects , PPAR gamma/antagonists & inhibitors , RNA, Small Interfering/pharmacology , Transcription Factors/metabolism , Transcription Factors/physiology
15.
Bioessays ; 33(1): 13-20, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20954221

ABSTRACT

A simple model, termed "occlusis", is presented here to account for both cell fate restriction during somatic development and reestablishment of pluripotency during reproduction. The model makes three assertions: (1) A gene's transcriptional potential can assume one of two states: the "competent" state, wherein the gene is responsive to, and can be activated by, trans-acting factors in the cellular milieu, and the "occluded" state, wherein the gene is blocked by cis-acting, chromatin-based mechanisms from responding to trans-acting factors such that it remains silent irrespective of whether transcriptional activators are present in the milieu. (2) As differentiation proceeds in somatic lineages, lineage-inappropriate genes shift progressively and irreversibly from competent to occluded state, thereby leading to the restriction of cell fate. (3) During reproduction, global deocclusion takes place in the germline and/or early zygotic cells to reset the genome to the competent state in order to facilitate a new round of organismal development.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Developmental , Gene Silencing , Trans-Activators/genetics , Transcription, Genetic/physiology , Transcriptional Activation , Animals , Cell Differentiation/genetics , Cell Growth Processes , Gene Expression Regulation, Developmental/physiology , Genome , Humans , Models, Biological , Pluripotent Stem Cells/physiology , Reproduction
16.
Nat Biotechnol ; 29(1): 68-72, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21151123

ABSTRACT

In contrast to 5-methylcytosine (5-mC), which has been studied extensively, little is known about 5-hydroxymethylcytosine (5-hmC), a recently identified epigenetic modification present in substantial amounts in certain mammalian cell types. Here we present a method for determining the genome-wide distribution of 5-hmC. We use the T4 bacteriophage ß-glucosyltransferase to transfer an engineered glucose moiety containing an azide group onto the hydroxyl group of 5-hmC. The azide group can be chemically modified with biotin for detection, affinity enrichment and sequencing of 5-hmC-containing DNA fragments in mammalian genomes. Using this method, we demonstrate that 5-hmC is present in human cell lines beyond those previously recognized. We also find a gene expression level-dependent enrichment of intragenic 5-hmC in mouse cerebellum and an age-dependent acquisition of this modification in specific gene bodies linked to neurodegenerative disorders.


Subject(s)
Biotin/chemistry , Cytosine/analogs & derivatives , DNA/chemistry , Genome, Human , Genome , Staining and Labeling/methods , 5-Methylcytosine/analogs & derivatives , Animals , Bacteriophage T4/enzymology , Cerebellum/chemistry , Cytosine/analysis , DNA/genetics , Glucosyltransferases/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Models, Molecular , Molecular Sequence Data , Sequence Analysis, DNA
17.
Stem Cells ; 28(12): 2162-71, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20963821

ABSTRACT

The intermediate filament protein, nestin, is a widely employed marker of multipotent neural stem cells (NSCs). Recent in vitro studies have implicated nestin in a number of cellular processes, but there is no data yet on its in vivo function. Here, we report the construction and functional characterization of Nestin knockout mice. We found that these mice show embryonic lethality, with neuroepithelium of the developing neural tube exhibiting significantly fewer NSCs and much higher levels of apoptosis. Consistent with this in vivo observation, NSC cultures derived from knockout embryos show dramatically reduced self-renewal ability that is associated with elevated apoptosis but no overt defects in cell proliferation or differentiation. Unexpectedly, nestin deficiency has no detectable effect on the integrity of the cytoskeleton. Furthermore, the knockout of Vimentin, which abolishes nestin's ability to polymerize into intermediate filaments in NSCs, does not lead to any apoptotic phenotype. These data demonstrate that nestin is important for the proper survival and self-renewal of NSCs, and that this function is surprisingly uncoupled from nestin's structural involvement in the cytoskeleton.


Subject(s)
Intermediate Filament Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Animals , Cell Death , Cell Proliferation , Cell Survival , Cells, Cultured , Cytoskeleton/metabolism , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Gene Deletion , Gene Expression Regulation , Gene Targeting , Intermediate Filament Proteins/deficiency , Intermediate Filament Proteins/genetics , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nestin , Neural Tube/metabolism , Neural Tube/pathology , Phenotype
18.
PLoS One ; 5(9): e13014, 2010 Sep 27.
Article in English | MEDLINE | ID: mdl-20885962

ABSTRACT

Techniques for small molecule screening are widely used in biological mechanism study and drug discovery. Here, we reported a novel adipocyte differentiation assay for small molecule selection, based on human mesenchymal stem cells (hMSCs) transduced with fluorescence reporter gene driven by adipogenic specific promoter--adipocyte Protein 2 (aP2; also namely Fatty Acid Binding Protein 4, FABP4). During normal adipogenic induction as well as adipogenic inhibition by Ly294002, we confirmed that the intensity of green fluorescence protein corresponded well to the expression level of aP2 gene. Furthermore, this variation of green fluorescence protein intensity can be read simply through fluorescence spectrophotometer. By testing another two small molecules in adipogenesis--Troglitazone and CHIR99021, we proved that this is a simple and sensitive method, which could be applied in adipocyte biology, drug discovery and toxicological study in the future.


Subject(s)
Adipocytes/drug effects , Adipogenesis/drug effects , Drug Evaluation, Preclinical/methods , Mesenchymal Stem Cells/cytology , Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation/drug effects , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism
19.
BMC Cell Biol ; 11: 42, 2010 Jun 19.
Article in English | MEDLINE | ID: mdl-20565897

ABSTRACT

BACKGROUND: Embryonic stem (ES) cells have attracted significant attention from researchers around the world because of their ability to undergo indefinite self-renewal and produce derivatives from the three cell lineages, which has enormous value in research and clinical applications. Until now, many ES cell lines of different mammals have been established and studied. In addition, recently, AS-ES1 cells derived from Apodemus sylvaticus were established and identified by our laboratory as a new mammalian ES cell line. Hence further research, in the application of AS-ES1 cells, is warranted. RESULTS: Herein we report the generation of multiple mesodermal AS-ES1 lineages via embryoid body (EB) formation by the hanging drop method and the addition of particular reagents and factors for induction at the stage of EB attachment. The AS-ES1 cells generated separately in vitro included: adipocytes, osteoblasts, chondrocytes and cardiomyocytes. Histochemical staining, immunofluorescent staining and RT-PCR were carried out to confirm the formation of multiple mesodermal lineage cells. CONCLUSIONS: The appropriate reagents and culture milieu used in mesodermal differentiation of mouse ES cells also guide the differentiation of in vitro AS-ES1 cells into distinct mesoderm-derived cells. This study provides a better understanding of the characteristics of AS-ES1 cells, a new species ES cell line and promotes the use of Apodemus ES cells as a complement to mouse ES cells in future studies.


Subject(s)
Antigens, Differentiation/metabolism , Cell Culture Techniques , Mesoderm/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Antigens, Differentiation/immunology , Cell Differentiation , Cell Lineage , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Embryonic Stem Cells , Immunohistochemistry , Mesoderm/cytology , Murinae/embryology , Murinae/growth & development , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Reverse Transcriptase Polymerase Chain Reaction
20.
PLoS One ; 5(5): e10611, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20485554

ABSTRACT

Constitutive promoters are used routinely to drive ectopic gene expression. Here, we carried out a systematic comparison of eight commonly used constitutive promoters (SV40, CMV, UBC, EF1A, PGK and CAGG for mammalian systems, and COPIA and ACT5C for Drosophila systems). We also included in the comparison the TRE promoter, which can be activated by the rtTA transcriptional activator in a doxycycline-inducible manner. To make our findings representative, we conducted the comparison in a variety of cell types derived from several species. We found that these promoters vary considerably from one another in their strength. Most promoters have fairly consistent strengths across different cell types, but the CMV promoter can vary considerably from cell type to cell type. At maximal induction, the TRE promoter is comparable to a strong constitutive promoter. These results should facilitate more rational choices of promoters in ectopic gene expression studies.


Subject(s)
Doxycycline/pharmacology , Promoter Regions, Genetic , Animals , Cell Line , Drosophila melanogaster/cytology , Drosophila melanogaster/drug effects , Drosophila melanogaster/genetics , Flow Cytometry , Genes, Reporter , Genetic Vectors/genetics , Green Fluorescent Proteins/metabolism , Humans , Lentivirus/drug effects , Lentivirus/genetics , Mice , Response Elements/genetics , Tetracycline/pharmacology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...