Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Front Genet ; 12: 627050, 2021.
Article in English | MEDLINE | ID: mdl-33889176

ABSTRACT

Gene editing of the mitochondrial genome using the CRISPR-Cas9 system is highly challenging mainly due to sub-efficient delivery of guide RNA and Cas9 enzyme complexes into the mitochondria. In this study, we were able to perform gene editing in the mitochondrial DNA by appending an NADH-ubiquinone oxidoreductase chain 4 (ND4) targeting guide RNA to an RNA transport-derived stem loop element (RP-loop) and expressing the Cas9 enzyme with a preceding mitochondrial localization sequence. We observe mitochondrial colocalization of RP-loop gRNA and a marked reduction of ND4 expression in the cells carrying a 11205G variant in their ND4 sequence coincidently decreasing the mtDNA levels. This proof-of-concept study suggests that a stem-loop element added sgRNA can be transported to the mitochondria and functionally interact with Cas9 to mediate sequence-specific mtDNA cleavage. Using this novel approach to target the mtDNA, our results provide further evidence that CRISPR-Cas9-mediated gene editing might potentially be used to treat mitochondrial-related diseases.

2.
JCI Insight ; 3(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30429376

ABSTRACT

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant or digenic disorder linked to derepression of the toxic DUX4 gene in muscle. There is currently no pharmacological treatment. The emergence of DUX4 enabled development of cell and animal models that could be used for basic and translational research. Since DUX4 is toxic, animal model development has been challenging, but progress has been made, revealing that tight regulation of DUX4 expression is critical for creating viable animals that develop myopathy. Here, we report such a model - the tamoxifen-inducible FSHD mouse model called TIC-DUX4. Uninduced animals are viable, born in Mendelian ratios, and overtly indistinguishable from WT animals. Induced animals display significant DUX4-dependent myopathic phenotypes at the molecular, histological, and functional levels. To demonstrate the utility of TIC-DUX4 mice for therapeutic development, we tested a gene therapy approach aimed at improving muscle strength in DUX4-expressing muscles using adeno-associated virus serotype 1.Follistatin (AAV1.Follistatin), a natural myostatin antagonist. This strategy was not designed to modulate DUX4 but could offer a mechanism to improve muscle weakness caused by DUX4-induced damage. AAV1.Follistatin significantly increased TIC-DUX4 muscle mass and strength even in the presence of DUX4 expression, suggesting that myostatin inhibition may be a promising approach to treat FSHD-associated weakness. We conclude that TIC-DUX4 mice are a relevant model to study DUX4 toxicity and, importantly, are useful in therapeutic development studies for FSHD.


Subject(s)
Disease Models, Animal , Follistatin/genetics , Genetic Therapy , Homeodomain Proteins/genetics , Muscular Dystrophy, Facioscapulohumeral/therapy , Myostatin/antagonists & inhibitors , Animals , Female , Follistatin/therapeutic use , Male , Mice, Transgenic , Muscular Dystrophy, Facioscapulohumeral/chemically induced , Muscular Dystrophy, Facioscapulohumeral/genetics , Phenotype , Tamoxifen
3.
J Vis Exp ; (136)2018 06 14.
Article in English | MEDLINE | ID: mdl-29985369

ABSTRACT

CRISPR/Cas9 technology is accelerating genome engineering in many cell types, but so far, gene delivery and stable gene modification have been challenging in primary NK cells. For example, transgene delivery using lentiviral or retroviral transduction resulted in a limited yield of genetically-engineered NK cells due to substantial procedure-associated NK cell apoptosis. We describe here a DNA-free method for genome editing of human primary and expanded NK cells using Cas9 ribonucleoprotein complexes (Cas9/RNPs). This method allowed efficient knockout of the TGFBR2 and HPRT1 genes in NK cells. RT-PCR data showed a significant decrease in gene expression level, and a cytotoxicity assay of a representative cell product suggested that the RNP-modified NK cells became less sensitive to TGFß. Genetically modified cells could be expanded post-electroporation by stimulation with irradiated mbIL21-expressing feeder cells.


Subject(s)
CRISPR-Cas Systems/genetics , Genetic Engineering/methods , Genetic Therapy/methods , Immunotherapy/methods , Killer Cells, Natural/metabolism , Ribonucleoproteins/metabolism , Humans
4.
Stem Cells ; 36(2): 192-205, 2018 02.
Article in English | MEDLINE | ID: mdl-29044892

ABSTRACT

One strategy for stem cell-based therapy of the cerebral cortex involves the generation and transplantation of functional, histocompatible cortical-like neurons from embryonic stem cells (ESCs). Diploid parthenogenetic Pg-ESCs have recently emerged as a promising source of histocompatible ESC derivatives for organ regeneration but their utility for cerebral cortex therapy is unknown. A major concern with Pg-ESCs is genomic imprinting. In contrast with biparental Bp-ESCs derived from fertilized oocytes, Pg-ESCs harbor two maternal genomes but no sperm-derived genome. Pg-ESCs are therefore expected to have aberrant expression levels of maternally expressed (MEGs) and paternally expressed (PEGs) imprinted genes. Given the roles of imprinted genes in brain development, tissue homeostasis and cancer, their deregulation in Pg-ESCs might be incompatible with therapy. Here, we report that, unexpectedly, only one gene out of 7 MEGs and 12 PEGs was differentially expressed between Pg-ESCs and Bp-ESCs while 13 were differentially expressed between androgenetic Ag-ESCs and Bp-ESCs, indicating that Pg-ESCs but not Ag-ESCs, have a Bp-like imprinting compatible with therapy. In vitro, Pg-ESCs generated cortical-like progenitors and electrophysiologically active glutamatergic neurons that maintained the Bp-like expression levels for most imprinted genes. In vivo, Pg-ESCs participated to the cortical lineage in fetal chimeras. Finally, transplanted Pg-ESC derivatives integrated into the injured adult cortex and sent axonal projections in the host brain. In conclusion, mouse Pg-ESCs generate functional cortical-like neurons with Bp-like imprinting and their derivatives properly integrate into both the embryonic cortex and the injured adult cortex. Collectively, our data support the utility of Pg-ESCs for cortical therapy. Stem Cells 2018;36:192-205.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Animals , DNA Methylation/genetics , DNA Methylation/physiology , Electrophysiology , Genomic Imprinting/genetics , Genomic Imprinting/physiology , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Parthenogenesis/genetics , Parthenogenesis/physiology
5.
PLoS One ; 11(12): e0166822, 2016.
Article in English | MEDLINE | ID: mdl-27926922

ABSTRACT

Transferring mouse mutations into specific mouse strain backgrounds can be critical for appropriate analysis of phenotypic effects of targeted genomic alterations and quantitative trait loci. Speed congenic breeding strategies incorporating marker-assisted selection of progeny with the highest percentage target background as breeders for the next generation can produce congenic strains within approximately 5 generations. When mating selected donor males to target strain females, this may require more than 1 year, with each generation lasting 10 to 11 weeks including 3 weeks of gestation and 7 to 8 weeks until the males reach sexual maturity. Because ovulation can be induced in female mice as early as 3 weeks of age, superovulation-aided backcrossing of marker-selected females could accelerate the production of congenic animals by approximately 4 weeks per generation, reducing time and cost. Using this approach, we transferred a transgenic strain of undefined genetic background to >99% C57BL/6J within 10 months, with most generations lasting 7 weeks. This involved less than 60 mice in total, with 9 to 18 animals per generation. Our data demonstrate that high-speed backcrossing through the female germline is feasible and practical with small mouse numbers.


Subject(s)
Germ Cells/physiology , Reproduction/physiology , Animals , Animals, Congenic/physiology , Female , Inbreeding/methods , Male , Mice , Mice, Inbred C57BL , Phenotype , Quantitative Trait Loci/physiology
6.
Mol Med ; 21: 185-96, 2015 Mar 23.
Article in English | MEDLINE | ID: mdl-25811991

ABSTRACT

Parent-of-origin imprints have been implicated in the regulation of neural differentiation and brain development. Previously we have shown that, despite the lack of a paternal genome, human parthenogenetic (PG) embryonic stem cells (hESCs) can form proliferating neural stem cells (NSCs) that are capable of differentiation into physiologically functional neurons while maintaining allele-specific expression of imprinted genes. Since biparental ("normal") hESC-derived NSCs (N NSCs) are targeted by immune cells, we characterized the immunogenicity of PG NSCs. Flow cytometry and immunocytochemistry revealed that both N NSCs and PG NSCs exhibited surface expression of human leukocyte antigen (HLA) class I but not HLA-DR molecules. Functional analyses using an in vitro mixed lymphocyte reaction assay resulted in less proliferation of peripheral blood mononuclear cells (PBMC) with PG compared with N NSCs. In addition, natural killer (NK) cells cytolyzed PG less than N NSCs. At a molecular level, expression analyses of immune regulatory factors revealed higher HLA-G levels in PG compared with N NSCs. In line with this finding, MIR152, which represses HLA-G expression, is less transcribed in PG compared with N cells. Blockage of HLA-G receptors ILT2 and KIR2DL4 on natural killer cell leukemia (NKL) cells increased cytolysis of PG NSCs. Together this indicates that PG NSCs have unique immunological properties due to elevated HLA-G expression.


Subject(s)
Cell Differentiation , Cytotoxicity, Immunologic , Embryonic Stem Cells/cytology , Gene Expression , HLA-G Antigens/genetics , Killer Cells, Natural/immunology , Neural Stem Cells/immunology , Neural Stem Cells/metabolism , Apoptosis/genetics , Apoptosis/immunology , Cell Line , Gene Expression Regulation , HLA-DR Antigens/genetics , HLA-DR Antigens/immunology , HLA-DR Antigens/metabolism , HLA-G Antigens/immunology , HLA-G Antigens/metabolism , Humans , Killer Cells, Natural/metabolism , MicroRNAs/genetics , Neural Stem Cells/cytology
7.
Stem Cells ; 32(7): 1983-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24740448

ABSTRACT

Parthenogenesis is the development of an oocyte without fertilization. Mammalian parthenogenetic (PG) embryos are not viable, but can develop into blastocysts from which embryonic stem cells (ESCs) have been derived in mouse and human. PG ESCs are frequently homozygous for alleles encoding major histocompatibility complex (MHC) molecules. MHC homozygosity permits much more efficient immune matching than MHC heterozygosity found in conventional ESCs, making PG ESCs a promising cell source for cell therapies requiring no or little immune suppression. However, findings of restricted differentiation and proliferation of PG cells in developmental chimeras have cast doubt on the potential of PG ESC derivatives for organ regeneration. To address this uncertainty, we determined whether PG ESC derivatives are effective in rescuing mice with lethal liver failure due to deficiency of fumarylacetoacetate hydrolase (Fah). In developmental chimeras generated by injecting wild-type PG ESCs into Fah-deficient blastocysts, PG ESCs differentiated into hepatocytes that could repopulate the liver, provide normal liver function, and facilitate long-term survival of adult mice. Moreover, after transplantation into adult Fah-deficient mice, PG ESC-derived hepatocytes efficiently engrafted and proliferated, leading to high-level liver repopulation. Our results show that--despite the absence of a paternal genome--PG ESCs can form therapeutically effective hepatocytes.


Subject(s)
Embryonic Stem Cells/transplantation , Liver Failure/therapy , Tyrosinemias/therapy , Animals , Cell Differentiation , Embryonic Stem Cells/physiology , Hepatocytes/physiology , Humans , Liver/pathology , Liver/physiopathology , Liver Regeneration , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Parthenogenesis
8.
PLoS One ; 9(2): e89471, 2014.
Article in English | MEDLINE | ID: mdl-24586802

ABSTRACT

Tudor domain containing (Tdrd) proteins that are expressed in germ cells are divided into two groups. One group, consisting of TDRD1, TDRKH, TDRD9 and TDRD12, function in piRNA biogenesis and retrotransposon silencing, while the other group including RNF17/TDRD4 and TDRD5-7 are required for spermiogenesis. These Tdrd proteins play distinct roles during male germ cell development. Here, we report the characterization of STK31/TDRD8 in mice. STK31 contains a tudor domain and a serine/threonine kinase domain. We find that STK31 is a cytoplasmic protein in germ cells. STK31 is expressed in embryonic gonocytes of both sexes and postnatal spermatocytes and round spermatids in males. Disruption of the tudor domain and kinase domain of STK31 respectively does not affect fertility in mice. Our data suggest that the function of STK31 may be redundant with other Tdrd proteins in germ cell development.


Subject(s)
Germ Cells/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Reproduction/genetics , Animals , Blotting, Western , Cytoplasm/metabolism , DNA Primers/genetics , Fluorescent Antibody Technique , Gene Expression Profiling , Histological Techniques , Male , Mice , Protein Structure, Tertiary/genetics , Reproduction/physiology
9.
Cell Med ; 5(1): 29-42, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-26858862

ABSTRACT

Uniparental zygotes with two paternal (androgenetic, AG) or two maternal genomes (gynogenetic, GG) cannot develop into viable offsprings but form blastocysts from which pluripotent embryonic stem (ES) cells can be derived. For most organs, it is unclear whether uniparental ES cells can give rise to stably expandable somatic stem cells that can repair injured tissues. Even if previous reports indicated that the capacity of AG ES cells to differentiate in vitro into pan-neural progenitor cells (pNPCs) and into cells expressing neural markers is similar to biparental [normal fertilized (N)] ES cells, their potential for functional neurogenesis is not known. Here we show that murine AG pNPCs give rise to neuron-like cells, which then generate sodium-driven action potentials while maintaining fidelity of imprinted gene expression. Neural engraftment after intracerebral transplantation was achieved only by late (22 days) AG and N pNPCs with in vitro low colony-forming cell (CFC) capacity. However, persisting CFC formation seen, in particular, in early (13 or 16 days) differentiation cultures of N and AG pNPCs correlated with a high incidence of trigerm layer teratomas. As AG ES cells display functional neurogenesis and in vivo stability similar to N ES cells, they represent a unique model system to study the roles of paternal and maternal genomes on neural development and on the development of imprinting-associated brain diseases.

10.
Cell Cycle ; 10(13): 2091-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21606677

ABSTRACT

Chimeras are organisms composed of at least two genetically distinct cell lineages originating from different zygotes. In the laboratory, mouse chimeras can be produced experimentally; various techniques allow combining different early stage mouse embryos with each other or with pluripotent stem cells. Identification of the progeny of the different lineages in chimeras permits to follow cell fate and function, enabling correlation of genotype with phenotype. Mouse chimeras have become a tool to investigate critical developmental processes, including cell specification, differentiation, patterning, and the function of specific genes. In addition, chimeras can also be generated to address biological processes in the adult, including mechanisms underlying diseases or tissue repair and regeneration. This review summarizes the different types of chimeras and how they have been generated and provides examples of how mouse chimeras offer a unique and powerful system to investigate questions pertaining to cell and tissue function in the developing and adult organism.


Subject(s)
Chimera , Disease , Embryo, Mammalian/physiology , Regeneration/physiology , Animals , Embryo, Mammalian/anatomy & histology , Humans , Mice , Organ Transplantation/methods , Stem Cells/cytology , Stem Cells/physiology
11.
Mol Reprod Dev ; 78(4): 241-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21308854

ABSTRACT

In eukaryotes, mRNA is actively exported to the cytoplasm by a family of nuclear RNA export factors (NXF). Four Nxf genes have been identified in the mouse: Nxf1, Nxf2, Nxf3, and Nxf7. Inactivation of Nxf2, a germ cell-specific gene, causes defects in spermatogenesis. Here we report that Nxf3 is expressed exclusively in Sertoli cells of the postnatal testis, in a developmentally regulated manner. Expression of Nxf3 coincides with the cessation of Sertoli cell proliferation and the beginning of their differentiation. Continued expression of Nxf3 in mature Sertoli cells of the adult is spermatogenesis stage-independent. Nxf3 is not essential for spermatogenesis, however, suggesting functional redundancy among Nxf family members. With its unique expression pattern in the testis, the promoter of Nxf3 can be used to drive postnatal Sertoli cell-specific expression of other proteins such as Cre recombinase.


Subject(s)
Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Sertoli Cells/metabolism , Spermatogenesis/physiology , Active Transport, Cell Nucleus , Animals , Cell Differentiation , Female , Integrases/metabolism , Male , Mice , Mice, Transgenic , RNA Transport , RNA, Messenger/metabolism , Sertoli Cells/cytology , Testis/cytology , Testis/metabolism
12.
J Clin Invest ; 121(2): 623-7, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21293060

ABSTRACT

To be of therapeutic use, autologous stem cells derived from patients with inherited genetic disorders require genetic modification via gene repair or insertion. Here, we present proof of principle that, for diseases associated with dominant alleles (gain-of-function or haploinsufficient loss-of-function), disease allele­free ES cells can be derived from afflicted individuals without genome manipulation. This approach capitalizes on the derivation of uniparental cells, such as parthenogenetic (PG) ES cell lines from disease allele­free gametes. Diploid mammalian uniparental embryos with only maternally (oocyte-) or paternally (sperm-)derived genomes fail early in development due to the nonequivalence of parental genomes caused by genomic imprinting. However, these uniparental embryos develop to the blastocyst stage, allowing the derivation of ES cell lines. Using a mouse model for dominant beta-thalassemia, we developed disease allele­free PG ES cell lines from the oocytes of affected animals. Phenotype correction was obtained in donor-genotype recipients after transplantation of in vitro hematopoietic ES cell derivatives. This genetic correction strategy without gene targeting is potentially applicable to any dominant disease. It could also be the sole approach for larger or more complex mutations that cannot be corrected by homologous recombination.


Subject(s)
Alleles , Disease Models, Animal , Genetic Therapy/methods , beta-Thalassemia/genetics , Animals , Blastocyst/cytology , Blastocyst/physiology , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Humans , Mice , Mice, Inbred C57BL
13.
Mol Cell Biol ; 31(6): 1275-87, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21245380

ABSTRACT

Histone H1 is an abundant and essential component of chromatin whose precise role in regulating gene expression is poorly understood. Here, we report that a major target of H1-mediated regulation in embryonic stem (ES) cells is the X-linked Rhox homeobox gene cluster. To address the underlying mechanism, we examined the founding member of the Rhox gene cluster-Rhox5-and found that its distal promoter (Pd) loses H1, undergoes demethylation, and is transcriptionally activated in response to loss of H1 genes in ES cells. Demethylation of the Pd is required for its transcriptional induction and we identified a single cytosine in the Pd that, when methylated, is sufficient to inhibit Pd transcription. Methylation of this single cytosine prevents the Pd from binding GA-binding protein (GABP), a transcription factor essential for Pd transcription. Thus, H1 silences Rhox5 transcription by promoting methylation of one of its promoters, a mechanism likely to extend to other H1-regulated Rhox genes, based on analysis of ES cells lacking DNA methyltransferases. The Rhox cluster genes targeted for H1-mediated transcriptional repression are also subject to another DNA methylation-regulated process: Xp imprinting. Remarkably, we found that only H1-regulated Rhox genes are imprinted, not those immune to H1-mediated repression. Together, our results indicate that the Rhox gene cluster is a major target of H1-mediated transcriptional repression in ES cells and that H1 is a candidate to have a role in Xp imprinting.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Genomic Imprinting , Histones/metabolism , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Cells, Cultured , Cytosine/metabolism , DNA Modification Methylases/metabolism , GA-Binding Protein Transcription Factor/metabolism , Gene Deletion , Genes, Homeobox , Histones/genetics , Homeodomain Proteins/metabolism , Mice , Multigene Family , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic
14.
J Biol Chem ; 285(53): 41337-47, 2010 Dec 31.
Article in English | MEDLINE | ID: mdl-21047779

ABSTRACT

Glycogen synthase kinase-3 (Gsk-3) isoforms, Gsk-3α and Gsk-3ß, are constitutively active, largely inhibitory kinases involved in signal transduction. Underscoring their biological significance, altered Gsk-3 activity has been implicated in diabetes, Alzheimer disease, schizophrenia, and bipolar disorder. Here, we demonstrate that deletion of both Gsk-3α and Gsk-3ß in mouse embryonic stem cells results in reduced expression of the de novo DNA methyltransferase Dnmt3a2, causing misexpression of the imprinted genes Igf2, H19, and Igf2r and hypomethylation of their corresponding imprinted control regions. Treatment of wild-type embryonic stem cells and neural stem cells with the Gsk-3 inhibitor, lithium, phenocopies the DNA hypomethylation at these imprinted loci. We show that inhibition of Gsk-3 by phosphatidylinositol 3-kinase (PI3K)-mediated activation of Akt also results in reduced DNA methylation at these imprinted loci. Finally, we find that N-Myc is a potent Gsk-3-dependent regulator of Dnmt3a2 expression. In summary, we have identified a signal transduction pathway that is capable of altering the DNA methylation of imprinted loci.


Subject(s)
DNA Methylation , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Enzymologic , Genomic Imprinting , Glycogen Synthase Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Insulin-Like Growth Factor II/metabolism , Mice , Mice, Transgenic , Models, Biological , Oligonucleotide Array Sequence Analysis , RNA, Long Noncoding , RNA, Untranslated/metabolism , Receptor, IGF Type 2/metabolism , Signal Transduction
15.
PLoS One ; 5(11): e14017, 2010 Nov 16.
Article in English | MEDLINE | ID: mdl-21103378

ABSTRACT

Ubiquitin E3 ligases target their substrates for ubiquitination, leading to proteasome-mediated degradation or altered biochemical properties. The ubiquitin ligase Ubr2, a recognition E3 component of the N-end rule proteolytic pathway, recognizes proteins with N-terminal destabilizing residues and plays an important role in spermatogenesis. Tex19.1 (also known as Tex19) has been previously identified as a germ cell-specific protein in mouse testis. Here we report that Tex19.1 forms a stable protein complex with Ubr2 in mouse testes. The binding of Tex19.1 to Ubr2 is independent of the second position cysteine of Tex19.1, a putative target for arginylation by the N-end rule pathway R-transferase. The Tex19.1-null mouse mutant phenocopies the Ubr2-deficient mutant in three aspects: heterogeneity of spermatogenic defects, meiotic chromosomal asynapsis, and embryonic lethality preferentially affecting females. In Ubr2-deficient germ cells, Tex19.1 is transcribed, but Tex19.1 protein is absent. Our results suggest that the binding of Ubr2 to Tex19.1 metabolically stabilizes Tex19.1 during spermatogenesis, revealing a new function for Ubr2 outside the conventional N-end rule pathway.


Subject(s)
Nuclear Proteins/metabolism , Spermatogenesis , Testis/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Binding Sites/genetics , Blotting, Western , Cysteine/genetics , Female , Immunoprecipitation , Male , Methionine/genetics , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Nuclear Proteins/genetics , Protein Binding , Protein Stability , RNA-Binding Proteins , Signal Transduction , Testis/cytology , Ubiquitin-Protein Ligases/genetics
16.
J Clin Invest ; 120(9): 3120-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20739754

ABSTRACT

The ability to generate induced pluripotent stem (iPS) cells from a patient's somatic cells has provided a foundation for organ regeneration without the need for immune suppression. However, it has not been established that the differentiated progeny of iPS cells can effectively reverse failure of a vital organ. Here, we examined whether iPS cell-derived hepatocytes have both the functional and proliferative capabilities needed for liver regeneration in mice with fumarylacetoacetate hydrolase deficiency. To avoid biases resulting from random genomic integration, we used iPS cells generated without viruses. To exclude compensation by hepatocytes not derived from iPS cells, we generated chimeric mice in which all hepatocytes were iPS cell derived. In vivo analyses showed that iPS cells were intrinsically able to differentiate into fully mature hepatocytes that provided full liver function. The iPS cell-derived hepatocytes also replicated the unique proliferative capabilities of normal hepatocytes and were able to regenerate the liver after transplantation and two-thirds partial hepatectomy. Thus, our results establish the feasibility of using iPS cells generated in a clinically acceptable fashion for rapid and stable liver regeneration.


Subject(s)
Hepatocytes/cytology , Hepatocytes/metabolism , Induced Pluripotent Stem Cells , Liver Regeneration/physiology , Animals , Cell Differentiation , Chimera , Female , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Mice , Mice, Inbred C57BL
17.
Proc Natl Acad Sci U S A ; 107(26): 11841-6, 2010 Jun 29.
Article in English | MEDLINE | ID: mdl-20534472

ABSTRACT

Piwi-interacting RNAs (piRNAs) are essential for silencing of transposable elements in the germline, but their biogenesis is poorly understood. Here we demonstrate that MOV10L1, a germ cell-specific putative RNA helicase, is associated with Piwi proteins. Genetic disruption of the MOV10L1 RNA helicase domain in mice renders both MILI and MIWI2 devoid of piRNAs. Absence of a functional piRNA pathway in Mov10l1 mutant testes causes loss of DNA methylation and subsequent derepression of retrotransposons in germ cells. The Mov10l1 mutant males are sterile owing to complete meiotic arrest. This mouse mutant expresses Piwi proteins but lacks piRNAs, suggesting that MOV10L1 is required for piRNA biogenesis and/or loading to Piwi proteins.


Subject(s)
RNA Helicases/genetics , RNA Helicases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Animals , Argonaute Proteins , Base Sequence , Cell Cycle Proteins , DNA Methylation , DNA Primers/genetics , Fertility , Male , Meiosis , Mice , Mice, Knockout , Mutation , Proteins/metabolism , RNA Helicases/deficiency , Retroelements/genetics , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Spermatocytes/metabolism , Spermatogenesis , Spermatogonia/metabolism , Testis/metabolism
18.
Int J Dev Biol ; 54(11-12): 1755-62, 2010.
Article in English | MEDLINE | ID: mdl-21404193

ABSTRACT

Patient derived stem cell-based therapies are considered a future treatment option for Parkinson´s disease, a chronic and progressive brain neurodegenerative disorder characterized by depletion of dopaminergic neurons in the basal ganglia. While many aspects of the in vitro and in vivo differentiation potential of uniparental parthenogenetic (PG) and gynogenetic (GG) embryonic stem (ES) cells of several species have been studied, the capacity of androgenetic (AG) ES cells to develop into neuronal subtypes remains unclear. Here, we investigated the potential of murine AG ES cells to undergo dopaminergic differentiation both via directed in vitro differentiation, and in vivo, in ES cell-chimeric E12.5 and E16.5 brains. We show that similar to normal (N; developed from a zygote with maternal and paternal genomes) ES cells, AG cells generated dopaminergic neurons in vitro and in E12.5 and E16.5 chimeric brains following blastocyst injection. Expression of brain-specific imprinted genes was maintained in AG and normal dopaminergic cell cultures. Our results indicate that AG ES cells have dopaminergic differentiation potential in vitro and in vivo. This contrasts with previous reports of limited neural in vivo differentiation of AG cells in later brain development, and suggests that AG ES cells could be therapeutically relevant for future cellular replacement strategies for brain disease.


Subject(s)
Brain/embryology , Cell Differentiation , Dopamine/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome , Neurons/cytology , Animals , Blastocyst , Brain/cytology , Cell Line , Chimera/embryology , Chimera/genetics , Gene Expression Regulation, Developmental , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction , Stem Cell Transplantation , Zygote
19.
Mol Cell Biol ; 29(24): 6401-12, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19805514

ABSTRACT

Phosphorylation of the degron of the IFNAR1 chain of the type I interferon (IFN) receptor triggers ubiquitination and degradation of this receptor and, therefore, plays a crucial role in negative regulation of IFN-alpha/beta signaling. Besides the IFN-stimulated and Jak activity-dependent pathways, a basal ligand-independent phosphorylation of IFNAR1 has been described and implicated in downregulating IFNAR1 in response to virus-induced endoplasmic reticulum (ER) stress. Here we report purification and characterization of casein kinase 1alpha (CK1alpha) as a bona fide major IFNAR1 kinase that confers basal turnover of IFNAR1 and cooperates with ER stress stimuli to mediate phosphorylation-dependent degradation of IFNAR1. Activity of CK1alpha was required for phosphorylation and downregulation of IFNAR1 in response to ER stress and viral infection. While many forms of CK1 were capable of phosphorylating IFNAR1 in vitro, human CK1alpha and L-CK1 produced by the protozoan Leishmania major were also capable of increasing IFNAR1 degron phosphorylation in cells. Expression of leishmania CK1 in mammalian cells stimulated the phosphorylation-dependent downregulation of IFNAR1 and attenuated its signaling. Infection of mammalian cells with L. major modestly decreased IFNAR1 levels and attenuated cellular responses to IFN-alpha in vitro. We propose a role for mammalian and parasite CK1 enzymes in regulating IFNAR1 stability and type I IFN signaling.


Subject(s)
Casein Kinase Ialpha/metabolism , Interferon Type I/metabolism , Leishmania major/enzymology , Protein Isoforms/metabolism , Protozoan Proteins/metabolism , Receptor, Interferon alpha-beta/metabolism , Signal Transduction/physiology , Amino Acid Sequence , Animals , Casein Kinase Ialpha/genetics , Cell Line , Humans , Molecular Sequence Data , Protein Isoforms/genetics , Protozoan Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Interferon alpha-beta/genetics
20.
Dev Biol ; 330(1): 167-74, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19345203

ABSTRACT

In eukaryotes, mRNA is actively transported from nucleus to cytoplasm by a family of nuclear RNA export factors (NXF). While yeast harbors only one such factor (Mex67p), higher eukaryotes encode multiple NXFs. In mouse, four Nxf genes have been identified: Nxf1, Nxf2, Nxf3, and Nxf7. To date, the function of mouse Nxf genes has not been studied by targeted gene deletion in vivo. Here we report the generation of Nxf2 null mutant mice by homologous recombination in embryonic stem cells. Nxf2-deficient male mice exhibit fertility defects that differ between mouse strains. One third of Nxf2-deficient males on a mixed (C57BL/6x129) genetic background exhibit meiotic arrest and thus are sterile, whereas the remaining males are fertile. Disruption of Nxf2 in inbred (C57BL/6J) males impairs spermatogenesis, resulting in male subfertility, but causes no meiotic arrest. Testis weight and sperm output in C57BL/6J Nxf2(-/Y) mice are sharply reduced. Mutant epididymal sperm exhibit diminished motility. Importantly, proliferation of spermatogonia in Nxf2(-/Y) mice is significantly decreased. As a result, inactivation of Nxf2 causes depletion of germ cells in a substantial fraction of seminiferous tubules in aged mice. These studies demonstrate that Nxf2 plays a dual function in spermatogenesis: regulation of meiosis and maintenance of spermatogonial stem cells.


Subject(s)
Meiosis , Nucleocytoplasmic Transport Proteins/genetics , RNA-Binding Proteins/genetics , Spermatogonia/metabolism , Age Factors , Animals , Cell Proliferation , Cytoplasm/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Nucleocytoplasmic Transport Proteins/metabolism , RNA Transport , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Sperm Motility/genetics , Spermatogenesis/genetics , Spermatogonia/cytology , Spermatogonia/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...