Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biochem Biophys Res Commun ; 510(1): 59-64, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30665718

ABSTRACT

Neural stem/progenitor cells (NSPCs) express higher levels of poly(ADP-ribose) polymerase 1 (PARP1) than mouse embryonic fibroblasts (MEFs). Inhibition of PARP induces the expression of several genes in the p53 signaling pathway, including p21, which is critical for cell cycle control at the G1/S phase, triggers apoptosis, and suppresses cell cycle progression in NSPCs. However, upon the up-regulation of p21, the cell cycle does not arrest at any specific phase. In the present study, the expression of genes specific to the G1/S and G2/M phases of the cell cycle were analyzed following treatment with PJ34 (N-[6-oxo-5,6-dihydro-phenanthridin-2-yl]-N,N-dimethylacetamide), an inhibitor of PARP. PJ34 treatment dramatically down-regulated cyclin B1 expression in NSPCs, but not in MEFs, which was confirmed by a promoter assay. Down-regulation of FoxM1 and B-MYB revealed that the down-regulation of cyclin B occurs at the transcriptional level. GADD45 was also specifically up-regulated in NSPCs. Taken together, the activation of p53 by PJ34 treatment in NSPCs induced changes in the expression of genes involved in the cell cycle. Fluorescence-activated cell sorting analysis revealed that PJ34 treatment suppressed G2/M to G1 progression in NSPCs, but not in MEFs. These data indicate that PJ34 treatment inhibits cyclin expression at the mRNA level and suppresses cell cycle progression in NSPCs.


Subject(s)
Cell Cycle/drug effects , Neural Stem Cells/cytology , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Animals , Cyclin B/drug effects , Cyclin B/genetics , Fibroblasts/drug effects , Genes, cdc/drug effects , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , RNA, Messenger/drug effects
2.
Biomacromolecules ; 20(5): 1849-1859, 2019 05 13.
Article in English | MEDLINE | ID: mdl-30893557

ABSTRACT

To facilitate the cytosolic delivery of larger molecules such as proteins, we developed a new cell-penetrating peptide sequence, named Pas2r12, consisting of a repeated Pas sequence (FFLIG-FFLIG) and d-dodeca-arginine (r12). This peptide significantly enhanced the cellular uptake and cytosolic release of enhanced green fluorescent protein and immunoglobulin G as cargos. We found that simply mixing Pas2r12 with cargos could generate cytosolic introducible forms. The cytosolic delivery of cargos by Pas2r12 was found to be an energy-requiring process, to rely on actin polymerization, and to be suppressed by caveolae-mediated endocytosis inhibitors (genistein and methyl-ß-cyclodextrin) and small interfering RNA against caveolin-1. These results suggest that Pas2r12 enhances membrane penetration of cargos without the need for cross-linking and that caveolae-mediated endocytosis may be the route by which cytosolic delivery is enhanced.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , Endocytosis , Arginine/analogs & derivatives , Caveolae/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell-Penetrating Peptides/chemistry , Cytosol/metabolism , Drug Carriers/chemistry , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunoglobulin G/metabolism
3.
BMC Neurosci ; 18(1): 14, 2017 01 17.
Article in English | MEDLINE | ID: mdl-28095779

ABSTRACT

BACKGROUND: Poly(ADP-ribose) polymerase 1 (PARP-1), which catalyzes poly(ADP-ribosyl)ation of proteins by using NAD+ as a substrate, plays a key role in several nuclear events, including DNA repair, replication, and transcription. Recently, PARP-1 was reported to participate in the somatic cell reprogramming process. Previously, we revealed a role for PARP-1 in the induction of neural apoptosis in a cellular model of cerebral ischemia and suggested the possible use of PARP inhibitors as a new therapeutic intervention. In the present study, we examined the effects of PARP inhibitors on neural stem/progenitor cells (NSPCs) of the mouse brain. RESULTS: PARP-1 was more abundant and demonstrated higher activity in NSPCs than in mouse embryonic fibroblasts. Treatment with PARP inhibitors suppressed the formation of neurospheres by NSPCs through the suppression of cell cycle progression and the induction of apoptosis. In order to identify the genes responsible for these effects, we investigated gene expression profiles by microarray analyses and found that several genes in the p53 signaling pathway were upregulated, including Cdkn1a, which is critical for cell cycle control, and Fas, Pidd, Pmaip1, and Bbc3, which are principal factors in the apoptosis pathway. Inhibition of poly(ADP-ribosyl)ation increased the levels of p53 protein, but not p53 mRNA, and enhanced the phosphorylation of p53 at Ser18. Experiments with specific inhibitors and also shRNA demonstrated that PARP-1, but not PARP-2, has a role in the regulation of p53. The effects of PARP inhibitors on NSPCs were not observed in Trp53 -/- NSPCs, suggesting a key role for p53 in these events. CONCLUSIONS: On the basis of the finding that PARP inhibitors facilitated the p53 signaling pathway, we propose that poly(ADP-ribosyl)ation contributes to the proliferation and self-renewal of NSPCs through the suppression of p53 activation.


Subject(s)
Neural Stem Cells/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/physiology , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Gene Expression Profiling , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred ICR , Mice, Knockout , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurogenesis/physiology , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Polymerase Chain Reaction , RNA, Messenger/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics
4.
Biol Reprod ; 86(6): 178, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22441799

ABSTRACT

Somatic cell hybridization is widely used to study the control of gene regulation and the stability of differentiated states. In contrast, the application of this method to germ cells has been limited in part because of an inability to culture germ cells. In this study, we produced germ cell hybrids using germ-line stem (GS) cells and multipotent germ-line stem (mGS) cells. While GS cells are enriched for spermatogonial stem cell (SSC) activity, mGS cells are similar to embryonic stem (ES) cells and originally derived from GS cells. Hybrids were successfully obtained between GS cells and ES cells, between GS cells and mGS cells, and between mGS cells and thymocytes. All exhibited ES cell markers and a behavior similar to ES cells, formed teratomas, and differentiated into somatic cell tissues. However, none of the hybrid cells were able to reconstitute spermatogenesis after microinjection into seminiferous tubules. Analyses of the DNA methylation patterns of imprinted genes also showed that mGS cells do not possess a DNA demethylation ability, which was found in embryonic germ cells derived from primordial germ cells. However, mGS cells reactivated the X chromosome and induced Pou5f1 expression in female thymocytes in a manner similar to ES cells. These data show that mGS cells possess ES-like reprogramming potential, which predominates over-SSC activity.


Subject(s)
Cell Culture Techniques , Embryonic Stem Cells , Germ Cells , Hybrid Cells , Multipotent Stem Cells , Animals , Cell Fusion , Female , Genomic Imprinting , Hybrid Cells/cytology , Hybrid Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Octamer Transcription Factor-3/metabolism , Phenotype , Teratoma/etiology , Testis/cytology , X Chromosome/metabolism
5.
Biol Reprod ; 85(1): 208-17, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21471297

ABSTRACT

Spermatogonial stem cells (SSCs) are the only stem cells in the body with germline potential, which makes them an attractive target for germline modification. We previously showed the feasibility of homologous recombination in mouse SSCs and produced knockout (KO) mice by exploiting germline stem (GS) cells, i.e., cultured spermatogonia with SSC activity. In this study, we report the successful homologous recombination in rat GS cells, which can be readily established by their ability to form germ cell colonies on culture plates whose surfaces are hydrophilic and neutrally charged and thus limit somatic cell binding. We established a drug selection protocol for GS cells under hypoxic conditions. The frequency of the homologous recombination of the Ocln gene was 4.2% (2 out of 48 clones). However, these GS cell lines failed to produce offspring following xenogeneic transplantation into mouse testes and microinsemination, suggesting that long-term culture and drug selection have a negative effect on GS cells. Nevertheless, our results demonstrate the feasibility of gene targeting in rat GS cells and pave the way toward the generation of KO rats.


Subject(s)
Recombination, Genetic , Spermatogonia/physiology , Stem Cells/physiology , Animals , Cell Culture Techniques , Male , Mice , Mice, Nude , Rats , Rats, Sprague-Dawley , Rats, Wistar , Selection, Genetic , Sperm Injections, Intracytoplasmic
6.
J Enzyme Inhib Med Chem ; 26(6): 883-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21395487

ABSTRACT

Carbon tetrachloride (CCl(4)) is routinely used as a model compound for eliciting centrilobular hepatotoxicity. It can be bioactivated to the trichloromethyl radical, which causes extensive lipid peroxidation and ultimately cell death by necrosis. Overactivation of poly(ADP-ribose) polymerase-1 (PARP-1) can rapidly reduce the levels of ß-nicotinamide adenine dinucleotide and adenosine triphosphate and ultimately promote necrosis. The aim of this study was to determine whether inhibition of PARP-1 could decrease CCl(4)-induced hepatotoxicity, as measured by degree of poly(ADP-ribosyl)ation, serum levels of lactate dehydrogenase (LDH), lipid peroxidation, and oxidative DNA damage. For this purpose, male ICR mice were administered intraperitoneally a hepatotoxic dose of CCl(4) with or without 6(5H)-phenanthridinone, a potent inhibitor of PARP-1. Animals treated with CCl(4) exhibited extensive poly(ADP-ribosyl)ation in centrilobular hepatocytes, elevated serum levels of LDH, and increased lipid peroxidation. In contrast, animals treated concomitantly with CCl(4) and 6(5H)-phenanthridinone showed significantly lower levels of poly(ADP-ribosyl)ation, serum LDH, and lipid peroxidation. No changes were observed in the levels of oxidative DNA damage regardless of treatment. These results demonstrated that the hepatotoxicity of CCl(4) is dependent on the overactivation of PARP-1 and that inhibition of this enzyme attenuates the hepatotoxicity of CCl(4).


Subject(s)
Carbon Tetrachloride Poisoning/drug therapy , Carbon Tetrachloride/toxicity , Enzyme Inhibitors/pharmacology , Liver/drug effects , Phenanthrenes/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Carbon Tetrachloride/administration & dosage , Injections, Intraperitoneal , Liver/enzymology , Liver/metabolism , Male , Mice , Mice, Inbred ICR , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/metabolism , Structure-Activity Relationship
7.
Dev Growth Differ ; 52(3): 303-10, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20148923

ABSTRACT

Spermatogonial stem cells (SSCs) provide the foundation for spermatogenesis, and are unique tissue-specific stem cells because of their ability to transmit genetic information to offspring. Generation of knockout mice using mouse SSCs became feasible after the successful establishment of protocols for the transplantation and long-term culture of these cells, called germline stem (GS) cells. Furthermore, SSCs can acquire pluripotentiality similar to that of embryonic stem (ES) cells, in addition to their highly differentiated spermatogenic potential. These ES-like cells, called multipotent GS (mGS) cells, are capable of generating knockout mice in a manner similar to that of ES cells. The use of GS and mGS cells for animal transgenesis has added a new dimension to gene-targeting technology using ES cells and somatic cell nuclear transfer, which has limited application. Furthermore, for regenerative medicine purposes, the use of mGS will settle problems such as ethics issues and immunological rejection associated with ES cells, as well as risks of insertional mutagenesis associated with integrated genes into induced pluripotent stem cells.


Subject(s)
Animals, Genetically Modified/genetics , Pluripotent Stem Cells/cytology , Spermatogonia/cytology , Animals , Cell Culture Techniques , Gene Targeting/methods , Humans , Male , Mice , Mice, Knockout/genetics , Pluripotent Stem Cells/transplantation , Regeneration , Spermatogonia/transplantation
8.
Dev Biol ; 312(1): 344-52, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17959167

ABSTRACT

Spermatogonial stem cells can convert into embryonic stem (ES) cell-like multipotent germline stem (mGS) cells in vitro and produce germline chimeras by blastocyst microinjection. Although homologous recombination was previously demonstrated in mGS cells, spermatogenesis was not found in chimeras, suggesting that they are not competent for germline modification. Here we conducted detailed analysis of chimeric animals to determine whether mGS cells retain germline potential after genetic manipulation. Spermatozoa that were deficient in the occludin gene could be recovered from animals that were chimeric with mGS cells that underwent homologous recombination. The phenotypes of the occludin knockout (KO) mice were similar to those reported for KO mice produced using ES cells, and the animals showed growth retardation, gastritis and male infertility. Furthermore, we found that heterozygous mGS cells acquire two copies of the G418-resistant genes and become homozygous for the targeted allele by culturing at high concentrations of G418. Cytogenetic analysis showed that the aneuploid mGS cells observed during genetic manipulation were trisomic for chromosome 8 or 11, which is a common chromosomal abnormality in ES cells. Thus, mGS cells can be used to produce KO animals, and this novel method of germline manipulation may prove useful in diverse mammalian species.


Subject(s)
Gene Targeting/methods , Germ Cells/cytology , Germ Cells/metabolism , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Animals , Chimera , Female , Fertility , Homozygote , Insemination , Male , Membrane Proteins/deficiency , Mice , Mice, Knockout , Occludin , Pedigree , Phenotype , Spermatogenesis , Spermatozoa/metabolism , Testis/metabolism
9.
Cancer Sci ; 98(12): 1845-52, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17888029

ABSTRACT

Recently Welch et al. reported that microRNA (miRNA)-34a functions as a potential tumor suppressor in neuroblastoma cells (Oncogene 26: 5017-22, 2007). Here, we conversely show that miRNA-34a supports cell proliferation in rat oxidative stress-induced renal carcinogenesis and is overexpressed in various types of human cancers. While searching for genetically unstable chromosomal areas in rat renal carcinogenesis, we found the miRNA-34 family reciprocally overexpressed in chromosomal areas with frequent allelic loss. By in situ hybridization and reverse transcription-polymerase chain reaction, cerebral neurons and Purkinje cells showed the highest expression of a major type, miRNA-34a, followed by a variety of endocrine cells and proliferating cells including germinal center lymphocytes and mouse embryonic fibroblasts and stem cells. In contrast, normal renal tubules, hepatocytes and myocardial cells showed faint expression. After 3 weeks of ferric nitrilotriacetate (Fe-NTA)-induced oxidative stress, regenerating renal proximal tubular cells showed high miRNA-34a expression. All of the Fe-NTA-induced rat renal carcinomas and an array of human cancers (151 positive cases of 177) showed high expression of miRNA-34a. Furthermore, knockdown of miRNA-34a with small interfering RNA significantly suppressed proliferation not only of renal carcinoma cells but also of HeLa and MCF7 cells. These results indicate that miRNA-34a overexpression, an acquired trait during carcinogenesis, supports cell proliferation in the majority of cancers suggesting an unexpected link in the cellular metabolism between cancer and neuronal and/or endocrine cells, which warrants further investigation.


Subject(s)
Carcinoma, Renal Cell/genetics , Gene Expression Regulation , Kidney Neoplasms/genetics , MicroRNAs/genetics , Animals , Breast Neoplasms , Carcinoma, Renal Cell/chemically induced , Carcinoma, Renal Cell/pathology , Cell Division , Female , Ferric Compounds/toxicity , HeLa Cells , Humans , In Situ Hybridization , Kidney Neoplasms/chemically induced , Kidney Neoplasms/pathology , Male , Nitrates/toxicity , Organ Specificity , RNA, Neoplasm/genetics , RNA, Small Interfering/genetics , Rats , Rats, Wistar , Transfection
10.
Neurosci Lett ; 327(2): 79-82, 2002 Jul 19.
Article in English | MEDLINE | ID: mdl-12098640

ABSTRACT

The association between (GT)n dinucleotide repeats in monoamine oxidase gene loci, monoamine oxidase A (MAOA) and B (MAOB), and Parkinson's disease (PD), Alzheimer's disease (AD), and Lewy body variant (LBV) of AD were determined. MAOA-GT polymorphisms were significantly associated with pure AD and LBV. MAOA-GT allele 113 was excessively represented in pure AD and LBV compared with controls. Furthermore, the frequency of females homozygous for MAOA-GT allele 113 was higher in pure AD and LBV than controls by 2.79- and 2.77-fold, respectively. In contrast, there was no association between MAOA-GT or MAOB-GT polymorphisms and PD. These results suggest that polymorphisms within the MAOA gene may have implication in AD pathology shared by pure AD and LBV.


Subject(s)
Alzheimer Disease/genetics , Lewy Body Disease/genetics , Monoamine Oxidase/genetics , Polymorphism, Genetic , Aged , Aged, 80 and over , Dinucleotide Repeats , Female , Gene Frequency , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , X Chromosome
11.
Gene Expr ; 11(3-4): 117-24, 2003.
Article in English | MEDLINE | ID: mdl-14686785

ABSTRACT

Clonal pheochromocytoma cell lines overexpressing cytochrome P450 2D6 (CYP2D6) were established. CYP2D6 was localized in the endoplasmic reticulum, and its enzymatic activity in the microsomal fraction was confirmed by using high performance liquid chromatography analysis with [guanidine-14C]debrisoquine as a substrate. Overexpression of CYP2D6 protected both actively dividing and differentiated cells against the toxic effects of 1-methyl-4-phenylpyridinium ion at the concentration range of 20-40 microM, as assessed by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. The production of reactive oxygen species in the mitochondria was suppressed. The cytotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine was unchanged in both actively dividing and differentiated cells overexpressing CYP2D6 versus mock-transfected controls at concentrations up to 500 microM. These results suggest that the lowered enzyme activity of CYP2D6 in individuals termed "poor metabolizers" may represent a risk factor from exposure to select neurotoxicants.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Cytochrome P-450 CYP2D6/genetics , Gene Expression Regulation, Enzymologic/physiology , Herbicides/toxicity , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cytochrome P-450 CYP2D6/metabolism , Humans , Mitochondria/metabolism , PC12 Cells/cytology , PC12 Cells/enzymology , Rats , Reactive Oxygen Species/metabolism
12.
Gene Expr ; 11(5-6): 263-70, 2004.
Article in English | MEDLINE | ID: mdl-15200238

ABSTRACT

Septin 3 is a novel member of the septin subfamily of GTPase domain proteins that was recently identified in human neuronal cells. These proteins are involved in vesicle trafficking, neurite outgrowth, and neurofibrillary tangle formation; however, the expression and functional role of septin 3 in normal neuronal tissues and as an etiological agent in neurological disorders is currently unclear. To further characterize these parameters, the present study analyzed the expression of three isoforms of septin 3 (A, B, and C) in fetal and adult human brains and polymorphism of the septin 3 exon 11 microsatellite in control, pure Alzheimer's disease (AD), Lewy body variant (LBV) of AD, and Parkinson's disease. Septin 3 mRNAs for isoforms A and B, but not C, were detected in the frontal cortex of fetus and adult human samples, as measured by reverse transcription-coupled polymerase chain reaction. Genotype analyses indicated that polymorphic septin 3 alleles were distributed in two peaks of frequency in both control and disease groups. Categorization of the alleles into short (S) and long (L) types revealed a significant difference between AD patients and controls (p = 0.034 by chi-square test). Furthermore, the S-allele homozygosity was significantly underrepresented in AD compared with control (p = 0.015 by chi-square test). These results suggest that polymorphism in exon 11 of septin 3 may have a determinative role in the pathogenesis of AD.


Subject(s)
Alzheimer Disease/genetics , GTP Phosphohydrolases/genetics , Polymorphism, Genetic , Aged , Aged, 80 and over , Alternative Splicing/genetics , Brain Chemistry/genetics , Chromosomes, Human, Pair 22/genetics , Cytochrome P-450 CYP2D6/genetics , Exons/genetics , Gene Expression/genetics , Gene Frequency , Humans , Lewy Body Disease/genetics , Middle Aged , Parkinson Disease/genetics , RNA, Messenger/analysis , Septins
13.
Gene Expr ; 11(5-6): 271-8, 2004.
Article in English | MEDLINE | ID: mdl-15200239

ABSTRACT

Septin 3 is a novel member of the septin subfamily of GTPase domain proteins. Human septin 3 was originally cloned during a screening of genes expressed in human teratocarcinoma cells induced to differentiate with retinoic acid. Alternative splicing of the septin 3 gene transcript produces two isoforms, A and B, in the human brain, though their regional expression and physiological function remain to be determined. The purpose of the present study was to identify the expression patterns of human septin 3 isoforms in normal human brain and a human neuroblastoma cell line, SH-SY5Y, after retinoic acid-induced differentiation. The expression and distribution patterns of septin 3 isoforms A and B were similar and resembled that of another septin, CDCrel-1. Septin 3A and 3B were expressed in normal human brain in a region-specific manner, with the highest level in the temporal cortex and hippocampus and the lowest level in the brainstem regions. Prominent immunoreactivity was observed diffusely in the neocortices in association with neuropils and punctate structures suggestive of synaptic junctions. Immunoprecipitation studies revealed that septin 3A, 3B, and CDCrel-1 form a complex in the frontal cortex of human brain. These findings, taken together, suggest that septin 3A and 3B, along with CDCrel-1, play some fundamental role(s) in synaptogenesis and neuronal development.


Subject(s)
Brain/metabolism , GTP Phosphohydrolases/metabolism , Antibodies/immunology , Brain Chemistry , Cell Cycle Proteins/analysis , Cell Differentiation , Cell Line , Frontal Lobe/chemistry , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/immunology , Gene Expression , Humans , Immunochemistry , Immunoprecipitation , Protein Binding , Protein Isoforms/analysis , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/analysis , Septins , Tretinoin/pharmacology , Up-Regulation
14.
Cell Stem Cell ; 11(4): 567-78, 2012 Oct 05.
Article in English | MEDLINE | ID: mdl-23040482

ABSTRACT

Spermatogonial stem cells (SSCs) reside in specific niches within seminiferous tubules. These niches are thought to secrete chemotactic factors for SSCs, because SSCs migrate to them upon transplantation. However, the identity of these chemotactic molecules remains unknown. Here, we established a testis feeder cell culture system and used it to identify SSC chemotactic factors. When seeded on testis cells from infertile mice, SSCs migrated beneath the Sertoli cells and formed colonies with a cobblestone appearance that were very similar to those produced by hematopoietic stem cells. Cultured cells maintained SSC activity and fertility for at least 5 months. Cobblestone colony formation depended on GDNF and CXCL12, and dominant-negative GDNF receptor transfection or CXCL12 receptor deficiency reduced SSC colonization. Moreover, GDNF upregulated CXCL12 receptor expression, and CXCL12 transfection in Sertoli cells increased homing efficiency. Overall, our findings identify GDNF and CXCL12 as SSC chemotactic factors in vitro and in vivo.


Subject(s)
Chemokine CXCL12/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Seminiferous Tubules/cytology , Sertoli Cells/physiology , Spermatogonia/physiology , Stem Cell Niche/physiology , Animals , Cell Movement/genetics , Cell Proliferation , Cell Survival/genetics , Cells, Cultured , Chemokine CXCL12/genetics , Coculture Techniques , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Male , Mice , Mice, Inbred C57BL , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Seminiferous Tubules/growth & development , Transgenes/genetics
15.
Cell Stem Cell ; 9(5): 463-75, 2011 Nov 04.
Article in English | MEDLINE | ID: mdl-22056142

ABSTRACT

The homing ability of spermatogonial stem cells (SSCs) allows them to migrate into niches after being transplantated into infertile testes. Transplanted SSCs attach to Sertoli cells and transmigrate through the blood-testis barrier (BTB), formed by inter-Sertoli tight junctions, toward niches on the basement membrane. The most critical step is the passage through the BTB, which limits the homing efficiency to <10%. Here we demonstrated the involvement of Rac1 in SSC transmigration. Rac1-deficient SSCs did not colonize the adult testes, but they reinitiated spermatogenesis when transplanted into pup testes without a BTB. Moreover, a dominant-negative Rac1 construct not only reduced the expression of several claudin proteins, which comprise the BTB, but also increased SSC proliferation both in vitro and in vivo. Short hairpin RNA (shRNA) -mediated suppression of claudin3, which was downregulated by Rac inhibition, reduced the SSC homing efficiency. Thus, Rac1 is a critical regulator of SSC homing and proliferation.


Subject(s)
Blood-Testis Barrier/metabolism , Neuropeptides/metabolism , Spermatogonia/cytology , Stem Cell Niche , Stem Cells/cytology , Transendothelial and Transepithelial Migration , rac GTP-Binding Proteins/metabolism , Animals , Blood-Testis Barrier/cytology , Cell Shape , Cells, Cultured , Colony-Forming Units Assay , Embryo, Mammalian/cytology , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Deletion , Gene Expression Regulation , Male , Mice , Mice, Knockout , Mutation/genetics , Neuropeptides/genetics , Spermatogonia/metabolism , Spermatogonia/transplantation , Stem Cells/metabolism , Tight Junctions/metabolism , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
16.
J Nutr Sci Vitaminol (Tokyo) ; 57(4): 298-305, 2011.
Article in English | MEDLINE | ID: mdl-22041913

ABSTRACT

Selenocysteine lyase (SCL) catalyzes the decomposition of L-selenocysteine to yield L-alanine and selenium by acting exclusively on l-selenocysteine. The X-ray structural analysis of rat SCL has demonstrated how SCL discriminates L-selenocysteine from L-cysteine on the molecular basis. SCL has been proposed to function in the recycling of the micronutrient selenium from degraded selenoproteins containing selenocysteine residues, but the role of SCL in selenium metabolism in vivo remains unclear. We here demonstrate that the (75)Se-labeling efficiency of selenoproteins with (75)Se-labeled selenoprotein P (Sepp1) as a selenium source was decreased in HeLa cells transfected with SCL siRNA as compared to the cells transfected with control siRNA. Immunocytochemical analyses showed high SCL expression in kidney and liver cells, where selenocysteine is recovered from selenoproteins. Mature testes of mice exhibited a specific staining pattern of SCL in spermatids that actively produce selenoproteins. However, SCL was weakly expressed in Sertoli cells, which receive Sepp1 and supply selenium to germ cells. These demonstrate that SCL occurs in the cells requiring selenoproteins, probably to recycle selenium derived from selenoproteins such as Sepp1.


Subject(s)
Kidney/metabolism , Liver/metabolism , Lyases/metabolism , Selenium/metabolism , Selenocysteine/metabolism , Selenoproteins/biosynthesis , Spermatids/metabolism , Acebutolol , Animals , HeLa Cells , Humans , Isotope Labeling , Male , Mice , RNA, Small Interfering/metabolism , Rats , Selenoprotein P/metabolism , Selenoproteins/metabolism , Sertoli Cells/metabolism , Substrate Specificity , Transfection
17.
PLoS One ; 4(11): e7909, 2009 Nov 19.
Article in English | MEDLINE | ID: mdl-19936070

ABSTRACT

BACKGROUND: Spermatogonial stem cells (SSCs) continuously undergo self-renewal division to support spermatogenesis. SSCs are thought to have a fixed phenotype, and development of a germ cell transplantation technique facilitated their characterization and prospective isolation in a deterministic manner; however, our in vitro SSC culture experiments indicated heterogeneity of cultured cells and suggested that they might not follow deterministic fate commitment in vitro. METHODOLOGY AND PRINCIPAL FINDINGS: In this study, we report phenotypic plasticity of SSCs. Although c-kit tyrosine kinase receptor (Kit) is not expressed in SSCs in vivo, it was upregulated when SSCs were cultured on laminin in vitro. Both Kit(-) and Kit(+) cells in culture showed comparable levels of SSC activity after germ cell transplantation. Unlike differentiating spermatogonia that depend on Kit for survival and proliferation, Kit expressed on SSCs did not play any role in SSC self-renewal. Moreover, Kit expression on SSCs changed dynamically once proliferation began after germ cell transplantation in vivo. CONCLUSIONS/SIGNIFICANCE: These results indicate that SSCs can change their phenotype according to their microenvironment and stochastically express Kit. Our results also suggest that activated and non-activated SSCs show distinct phenotypes.


Subject(s)
Gene Expression Regulation , Spermatogonia/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Cell Transplantation , Crosses, Genetic , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Phenotype , Proto-Oncogene Proteins c-kit/metabolism , Spermatogonia/metabolism
18.
Biol Reprod ; 81(1): 155-64, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19228594

ABSTRACT

Although spermatogonial stem cells (SSCs) are committed to spermatogenesis, they may also convert to an embryonic stem cell-like pluripotent state at a low frequency. Because changes in DNA methylation patterns are associated with this conversion, we examined the effect of manipulating DNA methyltransferase (Dnmt) expression on the fate of cultured SSCs, germline stem (GS) cells. Dnmt1 knockdown induced apoptosis in GS cells, which was attenuated by the loss of Trp53. In contrast, GS cells proliferated normally in vitro after Dnmt3a/Dnmt3b ablation or during Dnmt3l overexpression. However, Dnmt3a/Dnmt3b double-mutant cells showed hypomethylation in the SineB1 repetitive sequence, and Dnmt3l-overexpressing cells showed hypermethylation in major and minor satellite sequences; neither cell type formed teratomas and completed spermatogenesis following transplantation into the seminiferous tubules. Although genetic manipulation did not increase the conversion of GS cells to a pluripotent state, these results underscore the important role of DNMTs in survival and spermatogenic differentiation in SSCs.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Germ Cells/metabolism , Spermatogenesis/genetics , Stem Cells/metabolism , Animals , Apoptosis/genetics , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/metabolism , Gene Expression Regulation, Enzymologic , Germ Cells/enzymology , Infertility, Male/genetics , Male , Mice , Mice, Knockout , Spermatozoa/abnormalities , Spermatozoa/enzymology , Spermatozoa/metabolism , Stem Cells/enzymology , Stem Cells/physiology
19.
Biol Reprod ; 79(6): 1121-8, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18685123

ABSTRACT

Spermatogonial stem cells (SSCs) continue to proliferate in the testis to support spermatogenesis throughout life, which makes them ideal targets for germline modification. Although recent success in the production of transgenic and knockout animals using SSCs has opened up new experimental possibilities, several problems, including the low efficiency of germ cell transplantation and poor fertility rates, remain to be resolved. In the present study, we took advantage of the xenogeneic transplantation to resolve these problems. Rat SSCs were transduced in vitro with a lentiviral vector that expressed enhanced green fluorescent protein (EGFP), and then transplanted into the testes of immunodeficient mice. The transduced rat SSCs produced EGFP-expressing spermatogenic cells, and microinsemination using these cells was used to produce transgenic rats, which stably transmitted the transgene to the next generation. Thus, xenogeneic transplantation is a powerful strategy for transgenesis, and smaller xenogeneic surrogates can be used for male germline modification using SSCs.


Subject(s)
Lentivirus/genetics , Spermatogonia/transplantation , Stem Cell Transplantation , Transduction, Genetic , Transplantation, Heterologous/physiology , Animals , Animals, Genetically Modified , Blotting, Southern , DNA/biosynthesis , DNA/genetics , Genetic Vectors , Green Fluorescent Proteins , Infertility, Male/physiopathology , Insemination, Artificial , Male , Mice , Mice, Nude , Rats , Seminiferous Tubules/cytology , Seminiferous Tubules/physiology , Sertoli Cells/physiology , Sertoli Cells/transplantation
20.
Cell Stem Cell ; 3(5): 533-42, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18983968

ABSTRACT

Spermatogonial stem cells (SSCs) provide the foundation for spermatogenesis. In a manner comparable to hematopoietic stem cell transplantation, SSCs colonize the niche of recipient testes and reinitiate spermatogenesis following microinjection into the seminiferous tubules. However, little is known about the homing mechanism of SSCs. Here we examined the role of adhesion molecules in SSC homing. SSCs isolated from mice carrying loxP-tagged beta1-integrin alleles were ablated for beta1-integrin expression by in vitro adenoviral cre transduction. The beta1-integrin mutant SSCs showed significantly reduced ability to recolonize recipient testes in vivo and to attach to laminin molecules in vitro. In contrast, genetic ablation of E-cadherin did not impair homing, and E-cadherin mutant SSCs completed normal spermatogenesis. In addition, the deletion of beta1-integrin on Sertoli cells reduced SSC homing. These results identify beta1-integrin as an essential adhesion receptor for SSC homing and its association with laminin is critical in multiple steps of SSC homing.


Subject(s)
Integrin beta1/metabolism , Laminin/metabolism , Sertoli Cells/metabolism , Spermatogonia/cytology , Spermatogonia/metabolism , Adenoviridae , Animals , Cell Adhesion , Cell Movement , Integrases , Integrin beta1/genetics , Laminin/genetics , Male , Mice , Mice, Transgenic , Protein Binding , Sertoli Cells/cytology , Spermatogenesis , Stem Cell Niche/cytology , Stem Cell Transplantation , Testis/cytology , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL