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










Publication year range
1.
Stem Cell Res ; 65: 102976, 2022 12.
Article in English | MEDLINE | ID: mdl-36434993

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by abnormal glutamine (Q) expansion in the huntingtin protein due to elongated CAG repeats in the gene HTT. We used non-integrative episomal plasmids to generate induced pluripotent stem cells (iPSCs) from three individuals affected by HD: CH1 (58Q), and two twin brothers CH3 (44Q) and CH4 (44Q). The iPSC lines exhibited one healthy HTT allele and one with elongated CAG repeats, as confirmed by PCR and sequencing. All iPSC lines expressed pluripotency markers, exhibited a normal karyotype, and generated cells of the three germ layers in vitro.


Subject(s)
Huntingtin Protein , Huntington Disease , Induced Pluripotent Stem Cells , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Induced Pluripotent Stem Cells/pathology , Siblings , Cell Line , Huntingtin Protein/genetics , Alleles , Male
2.
Stem Cell Res ; 61: 102742, 2022 05.
Article in English | MEDLINE | ID: mdl-35279592

ABSTRACT

We report the generation of four human iPSC lines (8993-A12, 8993-B12, 8993-C11, and 8993-D7) from fibroblasts of four patients affected by maternally inherited Leigh syndrome (MILS) carrying homoplasmic mutations m.8993T > G or m.8993T > C in the mitochondrial gene MT-ATP6. We used Sendai viruses to deliver reprogramming factors OCT4, SOX2, KLF4, and c-MYC. The established iPSC lines expressed pluripotency markers, exhibited a normal karyotype, were capable to form cells of the three germ layers in vitro, and retained the MT-ATP6 mutations at the same homoplasmic level of the parental fibroblasts.


Subject(s)
Induced Pluripotent Stem Cells , Leigh Disease , Fibroblasts , Genes, Mitochondrial , Humans , Leigh Disease/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Mutation/genetics
4.
Nat Commun ; 12(1): 1929, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33771987

ABSTRACT

Leigh syndrome (LS) is a severe manifestation of mitochondrial disease in children and is currently incurable. The lack of effective models hampers our understanding of the mechanisms underlying the neuronal pathology of LS. Using patient-derived induced pluripotent stem cells and CRISPR/Cas9 engineering, we developed a human model of LS caused by mutations in the complex IV assembly gene SURF1. Single-cell RNA-sequencing and multi-omics analysis revealed compromised neuronal morphogenesis in mutant neural cultures and brain organoids. The defects emerged at the level of neural progenitor cells (NPCs), which retained a glycolytic proliferative state that failed to instruct neuronal morphogenesis. LS NPCs carrying mutations in the complex I gene NDUFS4 recapitulated morphogenesis defects. SURF1 gene augmentation and PGC1A induction via bezafibrate treatment supported the metabolic programming of LS NPCs, leading to restored neuronal morphogenesis. Our findings provide mechanistic insights and suggest potential interventional strategies for a rare mitochondrial disease.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Leigh Disease/genetics , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Mutation , Neurons/metabolism , Organoids/metabolism , Cells, Cultured , Child, Preschool , Humans , Induced Pluripotent Stem Cells/cytology , Leigh Disease/metabolism , Male , Metabolomics/methods , Mitochondria/genetics , Mitochondria/metabolism , Morphogenesis/genetics , Neurons/cytology , Proteomics/methods , Single-Cell Analysis/methods , Exome Sequencing
5.
EMBO Rep ; 19(5)2018 05.
Article in English | MEDLINE | ID: mdl-29661859

ABSTRACT

The maintenance of cellular identity requires continuous adaptation to environmental changes. This process is particularly critical for stem cells, which need to preserve their differentiation potential over time. Among the mechanisms responsible for regulating cellular homeostatic responses, mitochondria are emerging as key players. Given their dynamic and multifaceted role in energy metabolism, redox, and calcium balance, as well as cell death, mitochondria appear at the interface between environmental cues and the control of epigenetic identity. In this review, we describe how mitochondria have been implicated in the processes of acquisition and loss of stemness, with a specific focus on pluripotency. Dissecting the biological functions of mitochondria in stem cell homeostasis and differentiation will provide essential knowledge to understand the dynamics of cell fate modulation, and to establish improved stem cell-based medical applications.


Subject(s)
Homeostasis , Mitochondria/physiology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Energy Metabolism , Humans , Oxidation-Reduction
6.
EMBO J ; 37(2): 282-299, 2018 01 17.
Article in English | MEDLINE | ID: mdl-29212816

ABSTRACT

Huntington's disease (HD) is a neurodegenerative disorder caused by an expanded CAG trinucleotide repeat in the huntingtin gene (HTT). Molecular chaperones have been implicated in suppressing or delaying the aggregation of mutant Htt. Using in vitro and in vivo assays, we have identified a trimeric chaperone complex (Hsc70, Hsp110, and J-protein) that completely suppresses fibrilization of HttExon1Q48 The composition of this chaperone complex is variable as recruitment of different chaperone family members forms distinct functional complexes. The trimeric chaperone complex is also able to resolubilize Htt fibrils. We confirmed the biological significance of these findings in HD patient-derived neural cells and on an organismal level in Caenorhabditis elegans Among the proteins in this chaperone complex, the J-protein is the concentration-limiting factor. The single overexpression of DNAJB1 in HEK293T cells is sufficient to profoundly reduce HttExon1Q97 aggregation and represents a target of future therapeutic avenues for HD.


Subject(s)
HSC70 Heat-Shock Proteins , HSP110 Heat-Shock Proteins , HSP40 Heat-Shock Proteins , Huntingtin Protein , Multiprotein Complexes , Neurons/metabolism , Animals , Caenorhabditis elegans , HEK293 Cells , HSC70 Heat-Shock Proteins/chemistry , HSC70 Heat-Shock Proteins/genetics , HSC70 Heat-Shock Proteins/metabolism , HSP110 Heat-Shock Proteins/chemistry , HSP110 Heat-Shock Proteins/genetics , HSP110 Heat-Shock Proteins/metabolism , HSP40 Heat-Shock Proteins/chemistry , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Huntington Disease/pathology , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Neurons/pathology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology
7.
Sci Rep ; 7(1): 7516, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28790359

ABSTRACT

Nijmegen Breakage Syndrome (NBS) is associated with cancer predisposition, premature aging, immune deficiency, microcephaly and is caused by mutations in the gene coding for NIBRIN (NBN) which is involved in DNA damage repair. Dermal-derived fibroblasts from NBS patients were reprogrammed into induced pluripotent stem cells (iPSCs) in order to bypass premature senescence. The influence of antioxidants on intracellular levels of ROS and DNA damage were screened and it was found that EDHB-an activator of the hypoxia pathway, decreased DNA damage in the presence of high oxidative stress. Furthermore, NBS fibroblasts but not NBS-iPSCs were found to be more susceptible to the induction of DNA damage than their healthy counterparts. Global transcriptome analysis comparing NBS to healthy fibroblasts and NBS-iPSCs to embryonic stem cells revealed regulation of P53 in NBS fibroblasts and NBS-iPSCs. Cell cycle related genes were down-regulated in NBS fibroblasts. Furthermore, oxidative phosphorylation was down-regulated and glycolysis up-regulated specifically in NBS-iPSCs compared to embryonic stem cells. Our study demonstrates the utility of NBS-iPSCs as a screening platform for anti-oxidants capable of suppressing DNA damage and a cellular model for studying NBN de-regulation in cancer and microcephaly.


Subject(s)
Antioxidants/pharmacology , Fibroblasts/drug effects , Induced Pluripotent Stem Cells/drug effects , Nijmegen Breakage Syndrome/genetics , Signal Transduction/genetics , Transcriptome , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cellular Reprogramming , DNA Damage , Dermis/drug effects , Dermis/metabolism , Dermis/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation , Glycolysis/genetics , High-Throughput Screening Assays , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Male , Nijmegen Breakage Syndrome/metabolism , Nijmegen Breakage Syndrome/pathology , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Primary Cell Culture , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
Stem Cells ; 35(7): 1655-1662, 2017 07.
Article in English | MEDLINE | ID: mdl-28544378

ABSTRACT

High attrition rates and loss of capital plague the drug discovery process. This is particularly evident for mitochondrial disease that typically involves neurological manifestations and is caused by nuclear or mitochondrial DNA defects. This group of heterogeneous disorders is difficult to target because of the variability of the symptoms among individual patients and the lack of viable modeling systems. The use of induced pluripotent stem cells (iPSCs) might significantly improve the search for effective therapies for mitochondrial disease. iPSCs can be used to generate patient-specific neural cell models in which innovative compounds can be identified or validated. Here we discuss the promises and challenges of iPSC-based drug discovery for mitochondrial disease with a specific focus on neurological conditions. We anticipate that a proper use of the potent iPSC technology will provide critical support for the development of innovative therapies against these untreatable and detrimental disorders. Stem Cells 2017;35:1655-1662.


Subject(s)
Drug Discovery/methods , Induced Pluripotent Stem Cells/drug effects , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Cell Differentiation , DNA, Mitochondrial/genetics , Gene Expression , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/agonists , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Mutation , Neurons/metabolism , Neurons/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Precision Medicine
9.
Cell Stem Cell ; 20(5): 659-674.e9, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28132834

ABSTRACT

Mitochondrial DNA (mtDNA) mutations frequently cause neurological diseases. Modeling of these defects has been difficult because of the challenges associated with engineering mtDNA. We show here that neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (iPSCs) retain the parental mtDNA profile and exhibit a metabolic switch toward oxidative phosphorylation. NPCs derived in this way from patients carrying a deleterious homoplasmic mutation in the mitochondrial gene MT-ATP6 (m.9185T>C) showed defective ATP production and abnormally high mitochondrial membrane potential (MMP), plus altered calcium homeostasis, which represents a potential cause of neural impairment. High-content screening of FDA-approved drugs using the MMP phenotype highlighted avanafil, which we found was able to partially rescue the calcium defect in patient NPCs and differentiated neurons. Overall, our results show that iPSC-derived NPCs provide an effective model for drug screening to target mtDNA disorders that affect the nervous system.


Subject(s)
DNA, Mitochondrial/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mitochondria/genetics , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Calcium/metabolism , Cell Line , Drug Discovery/methods , Humans , Mutation
10.
Cell Stem Cell ; 19(4): 419-420, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27716519

ABSTRACT

Glycolysis is an essential component of cellular metabolism associated with pluripotent stem cells (PSCs). Two new papers, one by Gu et al. (2016) in this issue of Cell Stem Cell and one by Zhang et al. (2016) in Cell Reports, demonstrate that glycolytic flux is dynamically increased in human primed PSCs upon feeder-free cultivation or conversion into the naive state.


Subject(s)
Glycolysis , Pluripotent Stem Cells , Humans
11.
Cell Rep ; 16(9): 2499-511, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27545893

ABSTRACT

Nijmegen breakage syndrome (NBS) results from the absence of the NBS1 protein, responsible for detection of DNA double-strand breaks (DSBs). NBS is characterized by microcephaly, growth retardation, immunodeficiency, and cancer predisposition. Here, we show successful reprogramming of NBS fibroblasts into induced pluripotent stem cells (NBS-iPSCs). Our data suggest a strong selection for karyotypically normal fibroblasts to go through the reprogramming process. NBS-iPSCs then acquire numerous chromosomal aberrations and show a delayed response to DSB induction. Furthermore, NBS-iPSCs display slower growth, mitotic inhibition, a reduced apoptotic response to stress, and abnormal cell-cycle-related gene expression. Importantly, NBS neural progenitor cells (NBS-NPCs) show downregulation of neural developmental genes, which seems to be mediated by P53. Our results demonstrate the importance of NBS1 in early human development, shed light on the molecular mechanisms underlying this severe syndrome, and further expand our knowledge of the genomic stress cells experience during the reprogramming process.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomal Instability , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Nijmegen Breakage Syndrome/genetics , Nuclear Proteins/genetics , Acid Anhydride Hydrolases , Base Sequence , Cell Cycle Proteins/metabolism , Cell Proliferation , Cellular Reprogramming , DNA Breaks, Double-Stranded , DNA Repair , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblasts/pathology , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/pathology , Karyotyping , MRE11 Homologue Protein , Mitosis , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/pathology , Nijmegen Breakage Syndrome/metabolism , Nijmegen Breakage Syndrome/pathology , Nuclear Proteins/metabolism , Primary Cell Culture , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Semin Cell Dev Biol ; 52: 102-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26877213

ABSTRACT

The metabolic switch associated with the reprogramming of somatic cells to pluripotency has received increasing attention in recent years. However, the impact of mitochondrial and metabolic modulation on stem cell differentiation into neuronal/glial cells and related brain disease modeling still remains to be fully addressed. Here, we seek to focus on this aspect by first addressing brain energy metabolism and its inter-cellular metabolic compartmentalization. We then review the findings related to the mitochondrial and metabolic reconfiguration occurring upon neuronal/glial specification from pluripotent stem cells (PSCs). Finally, we provide an update of the PSC-based models of mitochondria-related brain disorders and discuss the challenges and opportunities that may exist on the road to develop a new era of brain disease modeling and therapy.


Subject(s)
Brain Diseases/metabolism , Induced Pluripotent Stem Cells/metabolism , Neuroglia/metabolism , Neurons/metabolism , Animals , Brain Diseases/pathology , Cell Differentiation/physiology , Energy Metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Neuroglia/cytology , Neurons/cytology
13.
Stem Cell Res ; 15(3): 629-32, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26987925

ABSTRACT

Human dermal fibroblasts from a Nijmegen Breakage Syndrome (NBS) patient bearing the 657del5 mutation within the DNA repair gene NIBRIN were used to generate two iPSC-lines (vNBS8-iPS-c1, vNBS8-iPS-c2) by retroviral transduction of OCT4, SOX2, c-MYC and KLF4. Pluripotency was confirmed both in vivo and in vitro.


Subject(s)
Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Nijmegen Breakage Syndrome , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Mutation
14.
Stem Cells ; 32(2): 364-76, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24123565

ABSTRACT

Reprogramming somatic cells to a pluripotent state drastically reconfigures the cellular anabolic requirements, thus potentially inducing cancer-like metabolic transformation. Accordingly, we and others previously showed that somatic mitochondria and bioenergetics are extensively remodeled upon derivation of induced pluripotent stem cells (iPSCs), as the cells transit from oxidative to glycolytic metabolism. In the attempt to identify possible regulatory mechanisms underlying this metabolic restructuring, we investigated the contributing role of hypoxia-inducible factor one alpha (HIF1α), a master regulator of energy metabolism, in the induction and maintenance of pluripotency. We discovered that the ablation of HIF1α function in dermal fibroblasts dramatically hampers reprogramming efficiency, while small molecule-based activation of HIF1α significantly improves cell fate conversion. Transcriptional and bioenergetic analysis during reprogramming initiation indicated that the transduction of the four factors is sufficient to upregulate the HIF1α target pyruvate dehydrogenase kinase (PDK) one and set in motion the glycolytic shift. However, additional HIF1α activation appears critical in the early upregulation of other HIF1α-associated metabolic regulators, including PDK3 and pyruvate kinase (PK) isoform M2 (PKM2), resulting in increased glycolysis and enhanced reprogramming. Accordingly, elevated levels of PDK1, PDK3, and PKM2 and reduced PK activity could be observed in iPSCs and human embryonic stem cells in the undifferentiated state. Overall, the findings suggest that the early induction of HIF1α targets may be instrumental in iPSC derivation via the activation of a glycolytic program. These findings implicate the HIF1α pathway as an enabling regulator of cellular reprogramming.


Subject(s)
Carrier Proteins/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Thyroid Hormones/genetics , Carrier Proteins/metabolism , Cell Differentiation/genetics , Cell Lineage , Cellular Reprogramming/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Glycolysis/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Induced Pluripotent Stem Cells/metabolism , Membrane Proteins/metabolism , Mitochondria/genetics , Neoplasms/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
15.
Adv Mater ; 25(20): 2765-71, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23529911

ABSTRACT

The groundbreaking discovery of reprogramming fibroblasts towards pluripotency merely by introducing four transcription factors (OCT4, SOX2, KLF4 and c-MYC) by means of retroviral transduction has created a promising revolution in the field of regenerative medicine. These so-called induced pluripotent stem cells (iPSCs) can provide a cell source for disease-modelling, drug-screening platforms, and transplantation strategies to treat incurable degenerative diseases, while circumventing the ethical issues and immune rejections associated with the use of non-autologous embryonic stem cells. The risk of insertional mutagenesis, caused both by the viral and transgene nature of the technique has proven to be the major limitation for iPSCs to be used in a clinical setting. In view of this, a variety of alternative techniques have been developed to induce pluripotency in somatic cells. This review provides an overview on current reprogramming protocols, discusses their pros and cons and future challenges to provide safe and transgene-free iPSCs.


Subject(s)
Batch Cell Culture Techniques/trends , Cell Culture Techniques/trends , Genetic Engineering/trends , Hybrid Cells/cytology , Hybrid Cells/physiology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Animals , Humans , Kruppel-Like Factor 4 , Tissue Engineering/trends
16.
PLoS One ; 7(11): e50393, 2012.
Article in English | MEDLINE | ID: mdl-23226273

ABSTRACT

The goal of our work has been to investigate the mechanisms of gender-independent human skin ageing and examine the hypothesis of skin being an adequate model of global ageing. For this purpose, whole genome gene profiling was employed in sun-protected skin obtained from European Caucasian young and elderly females (mean age 26.7±4 years [n1 = 7] and 70.75±3.3 years [n2 = 4], respectively) and males (mean age 25.8±5.2 years [n3 = 6] and 76±3.8 years [n4 = 7], respectively) using the Illumina array platform. Confirmation of gene regulation was performed by real-time RT-PCR and immunohistochemistry. 523 genes were significantly regulated in female skin and 401 genes in male skin for the chosen criteria. Of these, 183 genes exhibited increased and 340 decreased expression in females whereas 210 genes showed increased and 191 decreased expression in males with age. In total, 39 genes were common in the target lists of significant regulated genes in males and females. 35 of these genes showed increased (16) or decreased (19) expression independent of gender. Only 4 overlapping genes (OR52N2, F6FR1OP2, TUBAL3 and STK40) showed differential regulation with age. Interestingly, Wnt signalling pathway showed to be significantly downregulated in aged skin with decreased gene and protein expression for males and females, accordingly. In addition, several genes involved in central nervous system (CNS) ageing (f.i. APP, TAU) showed to be expressed in human skin and were significanlty regulated with age. In conclusion, our study provides biomarkers of endogenous human skin ageing in both genders and highlight the role of Wnt signalling in this process. Furthermore, our data give evidence that skin could be used as a good alternative to understand ageing of different tissues such as CNS.


Subject(s)
Aging/genetics , Skin Aging/genetics , Transcriptome , Wnt Signaling Pathway/genetics , Adult , Aged , Aged, 80 and over , Aging/metabolism , Aging/radiation effects , Biomarkers/metabolism , Central Nervous System/metabolism , Central Nervous System/radiation effects , Female , Gene Expression Profiling , Gene Expression Regulation/radiation effects , Genome-Wide Association Study , Humans , Immunohistochemistry , Male , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Sex Factors , Skin Aging/radiation effects , Sunlight , Ultraviolet Rays , Wnt Signaling Pathway/radiation effects
17.
Int J Dev Biol ; 56(10-12): 789-97, 2012.
Article in English | MEDLINE | ID: mdl-23417401

ABSTRACT

Primordial germ cells (PGCs) are precursors of gametes and share several features in common with pluripotent stem cells, such as alkaline phosphatase activity and the expression of pluripotency-associated genes such as OCT4 and NANOG. PGCs are able to differentiate into oocytes and spermatogonia and establish totipotency after fertilization. However, our knowledge of human germ cell development is still fragmentary. In this study, we have carried out genome-wide comparisons of the transcriptomes and molecular portraits of human male PGCs (mPGCs), female PGCs (fPGCs) and unfertilized oocytes. We detected 9210 genes showing elevated expression in fPGCs, 9184 in mPGCs and 9207 in oocytes, with 6342 of these expressed in common. As well as known germ cell-related genes such as BLIMP1/PRDM1, PIWIL2, VASA/DDX4, DAZL, STELLA/DPPA3 and LIN28, we also identified 465 novel non-annotated genes with orthologs in the mouse. A plethora of olfactory receptor-encoding genes were detected in all samples, which would suggest their involvement not only in sperm chemotaxis, but also in the development of female germ cells and oocytes. We anticipate that our data might increase our meagre knowledge of the genes and associated signaling pathways operative during germ cell development. This in turn might aid in the development of strategies enabling better differentiation and molecular characterisation of germ cells derived from either embryonic or induced pluripotent stem cells. Ultimately, this would have a profound relevance for reproductive as well as regenerative medicine.


Subject(s)
Gene Expression Regulation, Developmental , Germ Cells/metabolism , Oocytes/metabolism , Transcriptome/genetics , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Germ Cells/cytology , Gestational Age , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis , Oocytes/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
18.
Proc Natl Acad Sci U S A ; 105(27): 9284-9, 2008 Jul 08.
Article in English | MEDLINE | ID: mdl-18591665

ABSTRACT

RNAi is widely applied to inhibit expression of specific genes, but it is limited by variable efficiency and specificity of empirically designed siRNA or shRNA constructs. This complicates studies targeting individual genes and significantly impairs large-scale screens using genome-wide knockdown libraries. Here, we show that ectopic expression of the RISC slicer Argonaute-2 (Ago2, eIF2C2) dramatically enhances RNAi specifically for mRNA targets with perfectly matched binding sites. This effect depends on its endonuclease activity and is uncoupled from its regulation of microRNA expression. To model the application of Ago2 coexpression with shRNA knockdown, we targeted the EGF receptor (EGFR) in lung cancer cells exhibiting oncogene addiction to EGFR. Whereas multiple empirically designed shRNA constructs exhibited highly divergent efficiencies in mediating EGFR knockdown and cell killing, coexpression of Ago2 resulted in uniform and highly specific target gene suppression and apoptosis in EGFR-dependent cells. Codelivery of Ago2 with shRNA constructs or siRNA duplexes thus provides a strategy to enhance the efficacy and the specificity of RNAi in experimental and potentially therapeutic settings.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , RNA Interference , Argonaute Proteins , Binding Sites , Cell Death , Cell Line , ErbB Receptors/metabolism , Humans , RNA, Small Interfering/metabolism
19.
Int J Oncol ; 26(2): 493-8, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15645135

ABSTRACT

The p53 tumor suppressor protein mediates cell cycle arrest and apoptosis through transactivation of downstream target genes. While many target genes have been identified to date, the mechanisms and time course of their induction are still unclear. We investigated the kinetics of p53 binding to the p21CIP1, MDM2, BAX and PIG3 promoters in vivo using a novel quantitative real-time chromatin immunoprecipitation-PCR assay. Our results demonstrate distinct kinetics of p53 promoter binding dependent on the target gene promoters. The timed induction of target genes due to genotoxic stress is likely to play a pivotal role for the divergent functions of p53.


Subject(s)
Genes, p53 , Genetic Techniques , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction/methods , Blotting, Southern , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Cyclin-Dependent Kinase Inhibitor p21 , DNA/metabolism , Humans , Immunoprecipitation , Kinetics , Polymerase Chain Reaction , Protein Binding , Time Factors , Ultraviolet Rays
20.
Oncogene ; 23(57): 9162-72, 2004 Dec 09.
Article in English | MEDLINE | ID: mdl-15516979

ABSTRACT

Microarray analyses were performed to identify target genes that are shared by the acute myeloid leukemia (AML) translocation products PML-RARalpha, PLZF-RARalpha and AML1-ETO in inducibly transfected U937 cell lines. The cytoplasmic serine and threonine kinase MNK1 was identified as one of the target genes. At the protein level, MNK1 was significantly induced by each of the three fusion proteins. Protein half-life analyses showed that PML-RARalpha enhanced MNK1 protein stability in U937 cells and ATRA exposure decreased MNK1 half-life in NB4 cells. EIF4E, the main MNK1 substrate, plays a role in the pathogenesis of a variety of cancers. Upon MNK1 overexpression, eIF4E phosphorylation increased as a sign of functional activation. Interestingly, MNK1 protein expression decreased during myeloid differentiation. Inhibition of MNK1 activity by a specific inhibitor (CGP57380) enhanced differentiation of HL60 and 32D cells, further suggesting a role for MNK1 in the myeloid differentiation. In addition, kinase dead mutants of MNK1 significantly impaired proliferation of 32D cells. Immunohistochemistry of primary AML bone marrow biopsies showed strong cytoplasmic MNK1 expression in 25 of 99 AML specimens (25%). MNK1 expression was associated with high levels of c-myc expression. Taken together, we identified MNK1 as a target gene of several leukemogenic fusion proteins in AML. MNK1 plays a role in myeloid differentiation. These data suggest a role for MNK1 in the AML fusion protein-associated differentiation block.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/physiology , Protein Processing, Post-Translational/physiology , Protein Serine-Threonine Kinases/metabolism , Base Sequence , Cell Line, Tumor , DNA Primers , Eukaryotic Initiation Factor-4E/metabolism , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...