Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Dev Neurosci ; 36(5): 381-95, 2014.
Article in English | MEDLINE | ID: mdl-25138526

ABSTRACT

Spock3/Testican-3 is a nervous system-expressed heparan sulfate proteoglycan belonging to a subgroup of the BM-40/SPARC/osteonectin family, the role of which in brain development is unclear. Because Spock1, a member of the Spock family, inhibits their attachment to substrates and the neurite outgrowth of cultured neuronal cells, Spock3 is also thought to be similarly involved in the neuronal development. In the present study, we established a Spock3-mutant mouse harboring a deletion extending from the presumptive upstream regulatory region to exon 4 of the Spock3 locus and performed histological and behavioral studies on these mutant mice. In wild-type (WT) mice, all Spock members were clearly expressed during brain development. In adults, intense Spock1 and Spock2 expressions were observed throughout the entire brain; whereas, Spock3 expression was no longer visible except in the thalamic nuclei. Thus, Spock3 expression is mostly confined to the developmental stage of the brain. In adult mutant mice, the cells of all cortical layers were swollen. The corpus callosum was narrowed around the central region along the rostral-caudal axis and many small spaces were observed without myelin sheaths throughout the entire corpus callosum. In addition, the cortical input and output fibers did not form into thick bundled fibers as well as the WT counterparts did. Moreover, a subpopulation of corticospinal axonal fibers penetrated into the dorsal striatum with moderately altered orientations. Consistent with these modifications of brain structures, the mutant mice exhibited decreased anxiety-like behavior and lowered sociability. Together, these results demonstrate that Spock3 plays an important role in the formation or maintenance of major neuronal structures in the brain.


Subject(s)
Agenesis of Corpus Callosum/genetics , Anxiety/genetics , Axons/metabolism , Behavior, Animal/physiology , Corpus Callosum/metabolism , Proteoglycans/genetics , Social Behavior , Agenesis of Corpus Callosum/metabolism , Agenesis of Corpus Callosum/pathology , Animals , Anxiety/metabolism , Anxiety/pathology , Axons/pathology , Corpus Callosum/pathology , Male , Mice , Neurons/metabolism , Neurons/pathology , Proteoglycans/metabolism
2.
Dev Growth Differ ; 56(9): 653-68, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25389047

ABSTRACT

A mouse line carrying a lacZ transgene driven by the human EEF1A1/EF1 alpha promoter was established. Although the promoter is known to show ubiquitous activity, only paternal transgene alleles were expressed, resulting in a transgene imprinting. At mid-gestation, the promoter sequence was differentially methylated, hypomethylated for paternal and hypermethylated for maternal alleles. In germline, the promoter was a typical differentially methylated region. After fertilization, however, both alleles were hypermethylated. Thus, the differential methylation of the promoter required for transgene imprinting was re-established during later embryonic development independently of the germline differential methylation. Furthermore, also a retroelement promoter closely-flanking imprinted transgene and its wild type counterpart displayed similar differential methylation during early development. The retroelement promoter was methylated differentially also in germline, but in an opposite pattern to the embryonic differential methylation. These results suggest that there might be an unknown epigenetic regulation inducing transgene imprinting independently of DNA methylation in the transgene insertion site. Then, besides CpG dinucleotides, non-CpG cytosines of the retroelement promoter were highly methylated especially in the transgene-active mid-gestational embryos, suggesting that an unusual epigenetic regulation might protect the active transgene against de novo methylation occurring generally in mid-gestational embryo.


Subject(s)
Allelic Imbalance , DNA Methylation/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Repetitive Sequences, Nucleic Acid/genetics , Transgenes , Animals , Embryo, Mammalian/cytology , Humans , Lac Operon/genetics , Mice , Mice, Transgenic , Peptide Elongation Factor 1/genetics , Promoter Regions, Genetic/genetics
3.
Genesis ; 49(10): 803-10, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21956886

ABSTRACT

The homeobox gene Lbx1 not only plays critical roles in myogenesis and neurogenesis during embryonic development but is also expressed in activated satellite cells of adult mice. To address the potential postnatal functions of Lbx1, we generated conditional Lbx1-null mice using the Cre-loxP system. We generated a mouse in which Exon 2 of Lbx1 was floxed (Lbx1flox/flox), followed by cross-breeding between the Lbx1flox/flox mouse and either a transgenic mouse where a tamoxifen-inducible Cre-recombinase (Cre) was ubiquitously expressed, or a Myf5Cre mouse where Cre was inserted into the Myf5 locus. In both Lbx1-null mouse lines generated, Pax3-expressing limb muscle precursor cells were seriously reduced during embryonic development and eventually the limb extensor muscles were lost after birth. Since the conditional Lbx1-null mice generated were viable for a prolonged time, they will be useful in the investigation of Lbx1 function throughout the lifespan of the mouse.


Subject(s)
Integrases/genetics , Muscle Proteins/genetics , Muscle Proteins/physiology , Myogenic Regulatory Factor 5/genetics , Paired Box Transcription Factors/biosynthesis , Alleles , Animals , Crosses, Genetic , Extremities/embryology , Female , Integrases/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/cytology , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics
4.
Stem Cells ; 28(12): 2205-16, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20960513

ABSTRACT

Satellite cells are skeletal muscle stem cells responsible for growth, maintenance, and repair of postnatal skeletal muscle. Although several studies have demonstrated that Notch signaling plays a critical role in muscle regeneration through promoting proliferation and self-renewal of satellite cells, the function of Notch3 is yet to be elucidated. We analyzed muscle regeneration in Notch3-deficient mutant mice. We found a remarkable overgrowth of muscle mass in the Notch3-deficient mice but only when they suffered repetitive muscle injuries. Immunochemical analysis found that Notch3 was expressed in Pax7(+)/MyoD(-) quiescent satellite cells and also in Pax7(+)/MyoD(+)-activated satellite cells, but the expression was restricted to around half the population of each cell type. In Notch3-deficient mice, the number of sublaminar quiescent satellite cells was significantly increased compared with those in control mice. We also found that primary cultured myoblasts isolated from the Notch3-deficient mice proliferated faster than those from control mice. Analysis of cultured myofibers revealed that the number of self-renewing Pax7-positive satellite cells attached to the myofiber was increased in the Notch3-deficient mice when compared with control mice. The data obtained in this study suggested that Notch3 pathway might be distinct from Notch1 in muscle regeneration. Because overexpression of Notch3 activated the expression of Nrarp, a negative feedback regulator of Notch signaling, Notch3 might act as a Notch1 repressor by activating Nrarp.


Subject(s)
Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Mutation/genetics , Receptors, Notch/genetics , Regeneration/physiology , Animals , Cell Proliferation , Cells, Cultured , Hyperplasia , Mice , Mice, Inbred C57BL , Muscle, Skeletal/injuries , Myoblasts/metabolism , Myoblasts/pathology , Organ Size , Receptor, Notch3 , Receptors, Notch/deficiency , Receptors, Notch/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Satellite Cells, Skeletal Muscle/pathology
5.
Dev Growth Differ ; 52(2): 169-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20039927

ABSTRACT

Heparan sulfate (HS) comprises a structurally diverse group of glycosaminoglycans present ubiquitously on cell surfaces and in the extracellular matrix. The spatially and temporally regulated expression of specific HS structures is essential for various developmental processes in the nervous system but their distributions in the mouse olfactory system have not been explored. Here, we examined the spatiotemporal distribution of particular HS species in the developing mouse olfactory system using three structure-specific monoclonal antibodies (HepSS-1, JM403 and NAH46). The major findings were as follows. (i) During olfactory bulb morphogenesis, the HepSS-1 epitope was strongly expressed in anterior telencephalic cells and coexpressed with fibroblast growth factor receptor 1. (ii) In early postnatal glomeruli, the JM403 epitope was expressed at different levels among individual glomeruli. The expression pattern and levels of the JM403 epitope were both associated with those of ephrin-A3. (iii) In the vomeronasal system, the JM403 epitope was expressed in all vomeronasal axons but became increasingly restricted to vomeronasal axons terminating in the anterior region of the accessory olfactory bulb by 3 weeks of age. Our results demonstrate that each HS epitope exhibits a unique expression pattern during the development of the mouse olfactory system. Thus, each HS epitope is closely associated with particular developmental processes of the olfactory system and might have a particular role in developmental events.


Subject(s)
Epitopes/biosynthesis , Heparitin Sulfate/biosynthesis , Olfactory Bulb/chemistry , Olfactory Bulb/embryology , Animals , Antibodies, Monoclonal/immunology , Antigen-Antibody Reactions , Epitopes/immunology , Heparitin Sulfate/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Olfactory Bulb/immunology , Olfactory Bulb/ultrastructure
6.
Biochem Biophys Res Commun ; 381(4): 482-6, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19233126

ABSTRACT

NaPi-IIb encodes a Na(+)-dependent Pi co-transporter, which is expressed in various adult tissues and mediates transport of extracellular Pi ions coupling with Na(+) ion. To define the role of NaPi-IIbin vivo, NaPi-IIb gene deficient mice were generated utilizing targeted mutagenesis, yielding viable, heterozygous NaPi-IIb mice. In contrast, homozygous NaPi-IIb mice died in utero soon after implantation, indicating that NaPi-IIb was essential for early embryonic development. In situ hybridization revealed NaPi-IIb mRNA expression in the parietal endoderm, followed by the visceral endoderm, at a time point prior to establishment of a functioning chorio-allantoic placenta. At the time point of functional placenta development, the main site of NaPi-IIb production resided in the labyrinthine zone, where embryonic and maternal circulations were in closest contact. Expression patterns of NaPi-IIb suggest that NaPi-IIb plays an important role in Pi absorption from maternal circulation.


Subject(s)
Embryo Loss/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIb/physiology , Animals , Embryonic Development/genetics , Female , Gene Deletion , Gene Expression , Mice , Mice, Mutant Strains , Sodium-Phosphate Cotransporter Proteins, Type IIb/genetics
7.
Bone ; 43(5): 901-9, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18692165

ABSTRACT

In mammals and birds, most of the skeletal bones develop via endochondral ossification. Chondrocytes in the cartilaginous anlagen undergo processes of maturation such as hypertrophy, calcification and apoptosis. Concomitantly, osteoblasts are recruited to replace the cartilage scaffold gradually with bone matrix and become osteocytes in the trabecular bones. Throughout the successive development of bones, several gene products have been identified as being the components of the molecular mechanism regulating bone development. Transcription factor SOX9 plays essential roles during developmental steps from undifferentiated mesenchymal cells to proliferating chondrocytes, meanwhile, it inhibits transition of proliferating chondrocytes to hypertrophy. Other transcription factors RUNX2 and OSTERIX are critical in osteoblast differentiation, and RUNX2 is also essential for chondrocyte maturation such as hypertrophy and matrix mineralization. GDF5, a protein belonging to the transforming growth factor beta superfamily, is involved in joint formation and chondrogenesis. The limb skeleton of one of the ancestral tetrapod, anuran amphibians also develops through cartilaginous anlagen to bones, but their skeletogenesis has some unique characteristics compared with that of mammals and birds. Anuran amphibians develop and grow with less bone trabeculae and poor epiphyseal growth plates, and its endochondral ossification was found to be a delayed process. In order to address the characteristic skeletal development of anuran amphibians, we cloned Xenopus tropicalis RUNX2 (Xt-runx2), OSTERIX (Xt-osterix) and GDF5 (Xt-gdf5) homologue, and observed expression patterns together with Xt-sox9. In X. tropicalis limbs, histological observation and section in situ hybridization analysis suggest that Xt-SOX9 is involved in chondrogenesis, Xt-RUNX2 and Xt-OSTERIX are involved in osteogenesis, and Xt-GDF5 is involved in joint formation. In the cartilaginous anlagen, Xt-runx2 expression was found in perichondrium and immature chondrocytes as seen in other vertebrates. However, Xt-runx2 expression in enlarged chondrocytes was weak and dissimilar to common hypertrophic chondrocytes. These observations suggest that weak Xt-runx2 expression in maturing chondrocytes affects characteristic bone development in X. tropicalis long bones.


Subject(s)
Bone and Bones/embryology , Bone and Bones/physiology , Xenopus Proteins/metabolism , Xenopus , Amino Acid Sequence , Animals , Bone and Bones/anatomy & histology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Molecular Sequence Data , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Xenopus/anatomy & histology , Xenopus/physiology , Xenopus Proteins/genetics
8.
Nucleic Acids Res ; 33(9): e85, 2005 May 24.
Article in English | MEDLINE | ID: mdl-15914664

ABSTRACT

A major challenge of the post-genomic era is the functional characterization of anonymous open reading frames (ORFs) identified by the Human Genome Project. In this context, there is a strong requirement for the development of technologies that enhance our ability to analyze gene functions at the level of the whole organism. Here, we describe a rapid and efficient procedure to generate transgenic chimaeric mice that continuously secrete a foreign protein into the systemic circulation. The transgene units were inserted into the genomic site adjacent to the endogenous immunoglobulin (Ig) kappa locus by homologous recombination, using a modified mouse embryonic stem (ES) cell line that exhibits a high frequency of homologous recombination at the Igkappa region. The resultant ES clones were injected into embryos derived from a B-cell-deficient host strain, thus producing chimaerism-independent, B-cell-specific transgene expression. This feature of the system eliminates the time-consuming breeding typically implemented in standard transgenic strategies and allows for evaluating the effect of ectopic transgene expression directly in the resulting chimaeric mice. To demonstrate the utility of this system we showed high-level protein expression in the sera and severe phenotypes in human EPO (hEPO) and murine thrombopoietin (mTPO) transgenic chimaeras.


Subject(s)
Mice, Transgenic/genetics , Proteins/genetics , Proteins/metabolism , Animals , B-Lymphocytes/metabolism , Cell Line , Chimera , Clone Cells , Embryo, Mammalian/cytology , Erythropoietin/blood , Erythropoietin/genetics , Gene Targeting , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic/metabolism , Phenotype , Recombination, Genetic , Stem Cells/cytology , Thrombopoietin/blood , Thrombopoietin/genetics
9.
Sci Rep ; 6: 26342, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27197874

ABSTRACT

Disease-specific induced pluripotent stem cells (iPSCs) have been used as a model to analyze pathogenesis of disease. In this study, we generated iPSCs derived from a fibroblastic cell line of xeroderma pigmentosum (XP) group A (XPA-iPSCs), a rare autosomal recessive hereditary disease in which patients develop skin cancer in the areas of skin exposed to sunlight. XPA-iPSCs exhibited hypersensitivity to ultraviolet exposure and accumulation of single-nucleotide substitutions when compared with ataxia telangiectasia-derived iPSCs that were established in a previous study. However, XPA-iPSCs did not show any chromosomal instability in vitro, i.e. intact chromosomes were maintained. The results were mutually compensating for examining two major sources of mutations, nucleotide excision repair deficiency and double-strand break repair deficiency. Like XP patients, XPA-iPSCs accumulated single-nucleotide substitutions that are associated with malignant melanoma, a manifestation of XP. These results indicate that XPA-iPSCs may serve a monitoring tool (analogous to the Ames test but using mammalian cells) to measure single-nucleotide alterations, and may be a good model to clarify pathogenesis of XP. In addition, XPA-iPSCs may allow us to facilitate development of drugs that delay genetic alteration and decrease hypersensitivity to ultraviolet for therapeutic applications.


Subject(s)
Induced Pluripotent Stem Cells/pathology , Point Mutation , Skin Neoplasms/genetics , Xeroderma Pigmentosum/genetics , Cell Line, Tumor , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/radiation effects , Models, Biological , Sequence Analysis, DNA , Skin Neoplasms/etiology , Xeroderma Pigmentosum/complications , Xeroderma Pigmentosum/pathology , Xeroderma Pigmentosum Group A Protein/genetics
10.
Biochim Biophys Acta ; 1588(3): 195-202, 2002 Dec 12.
Article in English | MEDLINE | ID: mdl-12393173

ABSTRACT

Aceruloplasminemia is an autosomal recessive disorder caused by mutations in the ceruloplasmin (CP) gene, and is characterized by a unique combination of neurovisceral iron overload and iron deficiency anemia. We generated CP-deficient (CP(-/-)) mice to investigate the functional involvement of CP in iron metabolism. The mice showed a marked iron overload in the liver and mild iron deficiency anemia. We examined the expression of iron-metabolism genes in the duodenum and liver using TaqMan RT-PCR. The divalent metal transporter 1 (DMT1), ferroportin 1 (FPN1), and hephaestin (HEPH) genes were not up-regulated in the duodenum from CP(-/-) mice. These data suggest that the mechanism of hepatic iron overload in aceruloplasminemia is quite different from that in hemochromatoses and atransferrinemia. In the liver, CP(-/-) mice showed no increase of gene expression for DMT1 and transferrin receptors (TFR and TFR2), indicating that none of the known pathways of iron uptake is activated in hepatocytes of CP(-/-) mice. This result supports the hypothesis that CP mainly acts to release iron from cells in the liver.


Subject(s)
Anemia, Iron-Deficiency/genetics , Ceruloplasmin/deficiency , Iron Overload/genetics , Iron/metabolism , Anemia, Iron-Deficiency/metabolism , Animals , Cation Transport Proteins/genetics , Ceruloplasmin/genetics , Disease Models, Animal , Duodenum/metabolism , Gene Expression , Hepatocytes/metabolism , Iron/analysis , Iron/blood , Iron-Binding Proteins/genetics , Kupffer Cells/metabolism , Liver/metabolism , Membrane Proteins/genetics , Mice , Reverse Transcriptase Polymerase Chain Reaction
11.
Gene Expr Patterns ; 5(1): 43-9, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15533817

ABSTRACT

Two de novo-type DNA methyltransferases, Dnmt3a and Dnmt3b, are responsible for the creation of DNA methylation patterns during development. Dnmt3b is specifically expressed in the totipotent cells of mouse early embryos and Dnmt3a, a longer form of the two isoforms, is ubiquitously expressed in mesenchyme cells after the 10 day embryo stage [Mech. Dev. 118 (2002) 187]. In the present study, we demonstrated that Dnmt3b was expressed in the nuclei of specific cells in certain tissues after the 10 day embryo stage. In fetal liver, dorsal aorta and portal vein, Dnmt3b was expressed in cells expressing CD34, indicating that the cells were hematopoietic progenitor cells. However, Dnmt3b was not expressed in the hematopoietic progenitor cells in yolk sac blood islands at 8 day embryo stage and in adult bone marrow cells. Dnmt3b was also expressed in type-A spermatogonia after birth. Dnmt3b was expressed not only in the totipotent stem cells but also in the progenitor cells the direction of differentiation of which had been already determined. On the other hand, the long form of Dnmt3a was not expressed in these hematopoietic progenitor cells in fetal liver or type-A spermatogonia, but was expressed in hepatocytes in fetal liver and type-B spermatogonia. While Dnmt3b was distributed in both the heterochromatin and euchromatin regions, Dnmt3a was specifically localized to the euchromatin region.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Hematopoietic Stem Cells/enzymology , Spermatogonia/enzymology , Animals , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methyltransferase 3A , Fluorescent Antibody Technique , Gene Expression Profiling , Male , Mice
12.
Dev Growth Differ ; 28(3): 223-231, 1986 May.
Article in English | MEDLINE | ID: mdl-37281494

ABSTRACT

Various embryonal carcinoma cells of different origins were compared as to the ability to form chimeric blastocysts by means of aggregating with normal 8-cell stage mouse embryos. The teratocarcinoma lines examined were OTT6050 and five newly established ones including a spontaneous testicular teratocarcinoma STT-2. The present results have revealed that distinct differences existed in the ability of colonizing blastocysts among teratocarcinomas and also among embryonal carcinoma cell lines. Since STT-2 stem cells were found to be incorporated into blastocysts most efficiently, further development of the blastocysts were examined in utero. It was found that STT-2 stem cells could be incorporated into the fetuses up to the 7-to 28-somite stages. This is the first case to demonstrate that testicular teratocarcinoma cells with the male germ cell origin have the developmental potency to participate into mouse embryogenesis.

13.
Dev Growth Differ ; 29(4): 307-315, 1987 Aug.
Article in English | MEDLINE | ID: mdl-37281862

ABSTRACT

In the present study, we examined in detail the process of forming chimeric blastocysts between B242g embryonal carcinoma (EC) cells and normal mouse embryos. Electron microscopic observations of the developing aggregates revealed that the embryonic cells spread over the surface of the EC cells, resulting in the internalization of EC cells in the aggregates. When a single blastomere of an 8-cell embryo was aggregated with EC cells, the blastomere spread over and engulfed the EC cells. These results strongly suggest that EC cells are segregated and become situated in the inside position as the development of an aggregate proceeds, and then they are incorporated into the ICM of a blastocyst.

14.
Dev Growth Differ ; 29(3): 263-270, 1987 Jun.
Article in English | MEDLINE | ID: mdl-37282067

ABSTRACT

An embryonal carcinoma (EC) cell line having the ability to form chimeric mice was isolated from embryo-derived teratocarcinomas experimentally induced in BALB/cCrSlc mice. This EC cell line, B242 g, was one of the 5 EC cell lines pre-selected based on the ability to incorporate into blastocysts by means of aggregating with 8-cell mouse embryos. Using the B242g EC cells, the effectiveness of producing chimeras was compared between two currently available techniques, aggregation and injection, by examining chimerism of the midgestationally recovered conceptuses and live-born mice. The present result revealed that EC cells studied here were able to form chimeras more efficiently by injection as compared to aggregation method.

15.
Dev Growth Differ ; 38(6): 707-716, 1996 Dec.
Article in English | MEDLINE | ID: mdl-37282333

ABSTRACT

Previous work has indicated that N-myc expression occurs widely in the developing central nervous system, but its level changes dynamically with region- and stage-specificities. We show in the present report that in the developing spinal cord of the mouse, N-myc protein expression takes place in the ventricular zone and reaches its maximum at the outermost layer, but is extinct in the intermediate zone, indicating that N-myc protein is not expressed in mature neurons. We examined the effect of forced, persistent N-myc expression in development of the spinal cord in order to understand the functional significance of N-myc down-regulation. We made embryonic stem (ES) cell lines that constitutively expressed N-myc at a high level, then produced mouse embryo chimeras with a high contribution of the ES cells. The majority of the chimeras developed to day 12 with normal gross morphology, but in these chimeras neuronal differentiation in the spinal cord was perturbed at the histological level. Intermediate zones and ventral horns were formed, but the expression of N-CAM and neurofilaments was diminished. Chimeras using ß-galactosidase-expressing recipient embryos indicated that inhibition of the neuronal differentiation was a cell-autonomous effect of persistent N-myc expression. These observations indicate that N-myc down-regulation in individual cells is required for full differentiation of neurons.

16.
Dev Growth Differ ; 17(3): 227-235, 1975.
Article in English | MEDLINE | ID: mdl-37280967

ABSTRACT

Two cyclic nucleotide-dependent protein kinases, designated as protein kinase-I and -II, were obtained from the eggs of the silkworm, Bombyx mori. Protein kinase-I is highly dependent on cGMP, whereas protein kinase-II is dependent on cAMP. In developing non-diapause eggs, the level of cyclic nucleotide-dependent protein kinase activity is quite high but that in the diapause eggs is not. The developmental changes in the two protein kinases and the level of cyclic nucleotides were also studied during the development of the eggs.

17.
PLoS One ; 9(2): e89652, 2014.
Article in English | MEDLINE | ID: mdl-24586938

ABSTRACT

In this study we generated RNA interference (RNAi)-mediated gene knockdown transgenic mice (transgenic RNAi mice) against the functional Inv gene. Inv mutant mice show consistently reversed internal organs (situs inversus), multiple renal cysts and neonatal lethality. The Inv::GFP-rescue mice, which introduced the Inv::GFP fusion gene, can rescue inv mutant mice phenotypes. This indicates that the Inv::GFP gene is functional in vivo. To analyze the physiological functions of the Inv gene, and to demonstrate the availability of transgenic RNAi mice, we introduced a short hairpin RNA expression vector against GFP mRNA into Inv::GFP-rescue mice and analyzed the gene silencing effects and Inv functions by examining phenotypes. Transgenic RNAi mice with the Inv::GFP-rescue gene (Inv-KD mice) down-regulated Inv::GFP fusion protein and showed hypomorphic phenotypes of inv mutant mice, such as renal cyst development, but not situs abnormalities or postnatal lethality. This indicates that shRNAi-mediated gene silencing systems that target the tag sequence of the fusion gene work properly in vivo, and suggests that a relatively high level of Inv protein is required for kidney development in contrast to left/right axis determination. Inv::GFP protein was significantly down-regulated in the germ cells of Inv-KD mice testis compared with somatic cells, suggesting the existence of a testicular germ cell-specific enhanced RNAi system that regulates germ cell development. The Inv-KD mouse is useful for studying Inv gene functions in adult tissue that are unable to be analyzed in inv mutant mice showing postnatal lethality. In addition, the shRNA-based gene silencing system against the tag sequence of the fusion gene can be utilized as a new technique to regulate gene expression in either in vitro or in vivo experiments.


Subject(s)
Kidney Diseases, Cystic/genetics , RNA Interference , Transcription Factors/genetics , Aging , Animals , Gene Fusion , Gene Knockdown Techniques , Gene Silencing , Green Fluorescent Proteins/metabolism , Kidney/embryology , Male , Mice, Transgenic , RNA, Small Interfering/genetics , Situs Inversus/genetics , Testis/cytology , Testis/metabolism
18.
Sci Rep ; 4: 5421, 2014 Jun 27.
Article in English | MEDLINE | ID: mdl-24970375

ABSTRACT

Ataxia telangiectasia is a neurodegenerative inherited disease with chromosomal instability and hypersensitivity to ionizing radiation. iPS cells lacking ATM (AT-iPS cells) exhibited hypersensitivity to X-ray irradiation, one of the characteristics of the disease. While parental ataxia telangiectasia cells exhibited significant chromosomal abnormalities, AT-iPS cells did not show any chromosomal instability in vitro for at least 80 passages (560 days). Whole exome analysis also showed a comparable nucleotide substitution rate in AT-iPS cells. Taken together, these data show that ATM is involved in protection from irradiation-induced cell death.


Subject(s)
Ataxia Telangiectasia/pathology , Chromosomal Instability/radiation effects , Exome/genetics , Induced Pluripotent Stem Cells/cytology , Radiation Tolerance/genetics , Teratoma/pathology , Animals , Apoptosis/radiation effects , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/radiotherapy , Ataxia Telangiectasia Mutated Proteins/genetics , Blotting, Western , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , Cellular Reprogramming , Child , Fluorescent Antibody Technique , High-Throughput Nucleotide Sequencing , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/radiation effects , Karyotyping , Male , Mice , Mice, Inbred NOD , Mice, SCID , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Teratoma/genetics , Teratoma/radiotherapy , X-Rays
19.
Exp Anim ; 62(3): 211-7, 2013.
Article in English | MEDLINE | ID: mdl-23903056

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive progressive muscle degenerative disorder that causes dilated cardiomyopathy in the second decade of life in affected males. Dystrophin, the gene responsible for DMD, encodes full-length dystrophin and various short dystrophin isoforms. In the mouse heart, full-length dystrophin Dp427 and a short dystrophin isoform, Dp71, are expressed. In this study, we intended to clarify the functions of these dystrophin isoforms in DMD-related cardiomyopathy. We used two strains of mice: mdx mice, in which Dp427 was absent but Dp71 was present, and DMD-null mice, in which both were absent. By immunohistochemical staining and density-gradient centrifugation, we found that Dp427 was located in the cardiac sarcolemma and also at the T-tubules, whereas Dp71 was specifically located at the T-tubules. In order to determine whether T tubule-associated Dp71 was involved in DMD-related cardiac disruption, we compared the cardiac phenotypes between DMD-null mice and mdx mice. Both DMD-null mice and mdx mice exhibited severe necrosis, which was followed by fibrosis in cardiac muscle. However, we could not detect a significant difference in myocardial fibrosis between mdx mice and DMD-null mice. Based on the present results, we have shown that cardiac myopathy is caused predominantly by a deficiency of full-length dystrophin Dp427.


Subject(s)
Cardiomyopathies/genetics , Dystrophin/deficiency , Dystrophin/genetics , Myocytes, Cardiac/metabolism , Phenotype , Animals , Fibrosis , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred Strains , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/pathology , Protein Isoforms/genetics , Sarcolemma/metabolism
20.
PLoS One ; 8(5): e63016, 2013.
Article in English | MEDLINE | ID: mdl-23671652

ABSTRACT

Muscle satellite cells (SCs) are stem cells that reside in skeletal muscles and contribute to regeneration upon muscle injury. SCs arise from skeletal muscle progenitors expressing transcription factors Pax3 and/or Pax7 during embryogenesis in mice. However, it is unclear whether these fetal progenitors possess regenerative ability when transplanted in adult muscle. Here we address this question by investigating whether fetal skeletal muscle progenitors (FMPs) isolated from Pax3(GFP/+) embryos have the capacity to regenerate muscle after engraftment into Dystrophin-deficient mice, a model of Duchenne muscular dystrophy. The capacity of FMPs to engraft and enter the myogenic program in regenerating muscle was compared with that of SCs derived from adult Pax3(GFP/+) mice. Transplanted FMPs contributed to the reconstitution of damaged myofibers in Dystrophin-deficient mice. However, despite FMPs and SCs having similar myogenic ability in culture, the regenerative ability of FMPs was less than that of SCs in vivo. FMPs that had activated MyoD engrafted more efficiently to regenerate myofibers than MyoD-negative FMPs. Transcriptome and surface marker analyses of these cells suggest the importance of myogenic priming for the efficient myogenic engraftment. Our findings suggest the regenerative capability of FMPs in the context of muscle repair and cell therapy for degenerative muscle disease.


Subject(s)
Dystrophin/deficiency , Muscle, Skeletal/cytology , Muscular Dystrophy, Duchenne/surgery , Myoblasts, Skeletal/transplantation , Stem Cell Transplantation/methods , Animals , Cells, Cultured , Dystrophin/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Injections, Intramuscular , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Skeletal/embryology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Animal/surgery , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/physiopathology , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts, Skeletal/metabolism , Myofibrils/genetics , Myofibrils/physiology , Myogenin/genetics , Myogenin/metabolism , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Regeneration/physiology , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/transplantation , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL