Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Cells ; 13(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38727278

ABSTRACT

Spermatogenesis involves a complex process of cellular differentiation maintained by spermatogonial stem cells (SSCs). Being critical to male reproduction, it is generally assumed that spermatogenesis starts and ends in equivalent transcriptional states in related species. Based on single-cell gene expression profiling, it has been proposed that undifferentiated human spermatogonia can be subclassified into four heterogenous subtypes, termed states 0, 0A, 0B, and 1. To increase the resolution of the undifferentiated compartment and trace the origin of the spermatogenic trajectory, we re-analysed the single-cell (sc) RNA-sequencing libraries of 34 post-pubescent human testes to generate an integrated atlas of germ cell differentiation. We then used this atlas to perform comparative analyses of the putative SSC transcriptome both across human development (using 28 foetal and pre-pubertal scRNA-seq libraries) and across species (including data from sheep, pig, buffalo, rhesus and cynomolgus macaque, rat, and mouse). Alongside its detailed characterisation, we show that the transcriptional heterogeneity of the undifferentiated spermatogonial cell compartment varies not only between species but across development. Our findings associate 'state 0B' with a suppressive transcriptomic programme that, in adult humans, acts to functionally oppose proliferation and maintain cells in a ready-to-react state. Consistent with this conclusion, we show that human foetal germ cells-which are mitotically arrested-can be characterised solely as state 0B. While germ cells with a state 0B signature are also present in foetal mice (and are likely conserved at this stage throughout mammals), they are not maintained into adulthood. We conjecture that in rodents, the foetal-like state 0B differentiates at birth into the renewing SSC population, whereas in humans it is maintained as a reserve population, supporting testicular homeostasis over a longer reproductive lifespan while reducing mutagenic load. Together, these results suggest that SSCs adopt differing evolutionary strategies across species to ensure fertility and genome integrity over vastly differing life histories and reproductive timeframes.


Subject(s)
Spermatogonia , Humans , Animals , Male , Spermatogonia/cytology , Spermatogonia/metabolism , Adult Germline Stem Cells/metabolism , Adult Germline Stem Cells/cytology , Cell Differentiation/genetics , Spermatogenesis/genetics , Transcriptome/genetics , Adult , Mice , Fetus/cytology , Testis/cytology , Testis/metabolism , Rodentia , Rats , Single-Cell Analysis
2.
Procedia Comput Sci ; 192: 3722-3728, 2021.
Article in English | MEDLINE | ID: mdl-34691292

ABSTRACT

During the COVID 19 pandemic, the importance of global academia-industrial alliances has increased. It is hoped that the alliances will help us to solve the current problems caused by the pandemic. In this paper, we introduce the application of IT tools and communication skills utilized in a special educational project for an academia-industrial collaboration. Some concrete examples from 2020 are provided from the viewpoint of the national alliance project in Japan. A discussion is included that describes the plans available to increase and strengthen the national project in the future.

3.
Reproduction ; 161(6): 645-655, 2021 05 05.
Article in English | MEDLINE | ID: mdl-33835049

ABSTRACT

Initiation of spermatogonial differentiation in the mouse testis begins with the response to retinoic acid (RA) characterized by activation of KIT and STRA8 expression. In the adult, spermatogonial differentiation is spatiotemporally coordinated by a pulse of RA every 8.6 days that is localized to stages VII-VIII of the seminiferous epithelial cycle. Dogmatically, progenitor spermatogonia that express retinoic acid receptor gamma (RARG) at these stages will differentiate in response to RA, but this has yet to be tested functionally. Previous single-cell RNA-seq data identified phenotypically and functionally distinct subsets of spermatogonial stem cells (SSCs) and progenitor spermatogonia, where late progenitor spermatogonia were defined by expression of RARG and Dppa3. Here, we found late progenitor spermatogonia (RARGhigh KIT-) were further divisible into two subpopulations based on Dppa3 reporter expression (Dppa3-ECFP or Dppa3-EGFP) and were observed across all stages of the seminiferous epithelial cycle. However, nearly all Dppa3+ spermatogonia were differentiating (KIT+) late in the seminiferous epithelial cycle (stages X-XII), while Dppa3- late progenitors remained abundant, suggesting that Dppa3+ and Dppa3- late progenitors differentially responded to RA. Following acute RA treatment (2-4 h), significantly more Dppa3+ late progenitors induced KIT, including at the midpoint of the cycle (stages VI-IX), than Dppa3- late progenitors. Subsequently, single-cell analyses indicated a subset of Dppa3+ late progenitors expressed higher levels of Rxra, which we confirmed by RXRA whole-mount immunostaining. Together, these results indicate RARG alone is insufficient to initiate a spermatogonial response to RA in the adult mouse testis and suggest differential RXRA expression may discriminate responding cells.


Subject(s)
Adult Germline Stem Cells/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Receptors, Retinoic Acid/metabolism , Retinoid X Receptor alpha/metabolism , Spermatogenesis , Spermatogonia/metabolism , Tretinoin/pharmacology , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/drug effects , Animals , Antineoplastic Agents/pharmacology , Chromosomal Proteins, Non-Histone/genetics , Male , Mice , Receptors, Retinoic Acid/genetics , Retinoid X Receptor alpha/genetics , Spermatogonia/cytology , Spermatogonia/drug effects , Retinoic Acid Receptor gamma
4.
Cell Rep ; 34(7): 108752, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33596419

ABSTRACT

Spermatogonial stem cells (SSCs) sustain spermatogenesis by balancing self-renewal and initiation of differentiation to produce progenitor spermatogonia committed to forming sperm. To define the regulatory logic among SSCs and progenitors, we performed single-cell RNA velocity analyses and validated results in vivo. A predominant quiescent SSC population spawns a small subset of cell-cycle-activated SSCs via mitogen-activated protein kinase (MAPK)/AKT signaling. Activated SSCs form early progenitors and mTORC1 inhibition drives activated SSC accumulation consistent with blockade to progenitor formation. Mechanistically, mTORC1 inhibition suppresses transcription among spermatogonia and specifically alters expression of insulin growth factor (IGF) signaling in early progenitors. Tex14-/- testes lacking intercellular bridges do not accumulate activated SSCs following mTORC1 inhibition, indicating that steady-state mTORC1 signaling drives activated SSCs to produce progenitor clones. These results are consistent with a model of SSC self-renewal dependent on interconversion between activated and quiescent SSCs, and mTORC1-dependent initiation of differentiation from SSCs to progenitor clones.


Subject(s)
Adult Germline Stem Cells/physiology , Mechanistic Target of Rapamycin Complex 1/metabolism , Spermatogonia/physiology , Adult Germline Stem Cells/cytology , Adult Germline Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , Spermatogonia/metabolism
5.
Nat Commun ; 11(1): 5656, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33168808

ABSTRACT

Establishment of spermatogonia throughout the fetal and postnatal period is essential for production of spermatozoa and male fertility. Here, we establish a protocol for in vitro reconstitution of human prospermatogonial specification whereby human primordial germ cell (PGC)-like cells differentiated from human induced pluripotent stem cells are further induced into M-prospermatogonia-like cells and T1 prospermatogonia-like cells (T1LCs) using long-term cultured xenogeneic reconstituted testes. Single cell RNA-sequencing is used to delineate the lineage trajectory leading to T1LCs, which closely resemble human T1-prospermatogonia in vivo and exhibit gene expression related to spermatogenesis and diminished proliferation, a hallmark of quiescent T1 prospermatogonia. Notably, this system enables us to visualize the dynamic and stage-specific regulation of transposable elements during human prospermatogonial specification. Together, our findings pave the way for understanding and reconstructing human male germline development in vitro.


Subject(s)
Embryonic Germ Cells/physiology , Induced Pluripotent Stem Cells/physiology , Spermatogenesis/genetics , Spermatogenesis/physiology , Animals , Cell Differentiation , Epigenomics , Female , Gene Expression Regulation, Developmental , Humans , Male , Mice , Mice, Inbred ICR , Regulatory Elements, Transcriptional , Sequence Analysis, RNA , Spermatogonia/cytology , Spermatozoa , Testis/cytology , Transcriptome
6.
Biol Reprod ; 101(3): 617-634, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31077285

ABSTRACT

Mammalian spermatogenesis is a complex developmental program that transforms mitotic testicular germ cells (spermatogonia) into mature male gametes (sperm) for production of offspring. For decades, it has been known that this several-weeks-long process involves a series of highly ordered and morphologically recognizable cellular changes as spermatogonia proliferate, spermatocytes undertake meiosis, and spermatids develop condensed nuclei, acrosomes, and flagella. Yet, much of the underlying molecular logic driving these processes has remained opaque because conventional characterization strategies often aggregated groups of cells to meet technical requirements or due to limited capability for cell selection. Recently, a cornucopia of single-cell transcriptome studies has begun to lift the veil on the full compendium of gene expression phenotypes and changes underlying spermatogenic development. These datasets have revealed the previously obscured molecular heterogeneity among and between varied spermatogenic cell types and are reinvigorating investigation of testicular biology. This review describes the extent of available single-cell RNA-seq profiles of spermatogenic and testicular somatic cells, how those data were produced and evaluated, their present value for advancing knowledge of spermatogenesis, and their potential future utility at both the benchtop and bedside.


Subject(s)
Mammals/genetics , RNA-Seq/methods , Single-Cell Analysis/methods , Spermatogenesis/genetics , Animals , Humans , Male , Transcriptome/physiology , Translational Research, Biomedical/methods , Translational Research, Biomedical/trends
7.
Adv Anat Embryol Cell Biol ; 229: 15-30, 2018.
Article in English | MEDLINE | ID: mdl-29177762

ABSTRACT

In mammals, the processes spanning from fertilization to the generation of a new organism are very complex and are controlled by multiple genes. Life begins with the encounter of eggs and spermatozoa, in which gene expression is inactive prior to fertilization. After several cell divisions, cells arise that are specialized in implantation, a developmental process unique to mammals. Cells involved in the establishment and maintenance of implantation differentiate from totipotent embryos, and the remaining cells generate the embryo proper. Although this process of differentiation, termed cell lineage specification, is supported by various gene expression networks, many components have yet to be identified. Moreover, despite extensive research it remains unclear which genes are controlled by each of the factors involved. Although it has become clear that epigenetic factors regulate gene expression, elucidation of the underlying mechanisms remains challenging. In this chapter, we propose that the chromatin remodeling factor CHD1, together with epigenetic factors, is involved in a subset of gene expression networks involved in processes spanning from zygotic genome activation to cell lineage specification.


Subject(s)
Cell Differentiation , Embryo Implantation , Gene Expression Regulation, Developmental , Genome , Zygote , Animals , Cell Division , Chromatin , Embryo, Mammalian , Zygote/metabolism
8.
Development ; 142(13): 2375-84, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26092847

ABSTRACT

The protein CHD1 is a member of the family of ATPase-dependent chromatin remodeling factors. CHD1, which recognizes trimethylated histone H3 lysine 4, has been implicated in transcriptional activation in organisms ranging from yeast to humans. It is required for pre-mRNA maturation, maintenance of mouse embryonic stem cell pluripotency and rapid growth of the mouse epiblast. However, the function(s) of CHD1 in mouse preimplantation embryos has not yet been examined. Here, we show that loss of CHD1 function led to embryonic lethality after implantation. In mouse embryos in which Chd1 was targeted by siRNA microinjection, the expression of the key regulators of cell fate specification Pou5f1 (also known as Oct4), Nanog and Cdx2 was dramatically decreased, starting at mid-preimplantation gene activation (MGA). Moreover, expression of Hmgpi and Klf5, which regulate Pou5f1, Nanog and Cdx2, was also significantly suppressed at zygotic gene activation (ZGA). Suppression of Hmgpi expression in Chd1-knockdown embryos continued until the blastocyst stage, whereas suppression of Klf5 expression was relieved by the morula stage. Next, we rescued HMGPI expression via Hmgpi mRNA microinjection in Chd1-knockdown embryos. Consequently, Pou5f1, Nanog and Cdx2 expression was restored at MGA and live offspring were recovered. These findings indicate that CHD1 plays important roles in mouse early embryogenesis via activation of Hmgpi at ZGA.


Subject(s)
DNA-Binding Proteins/metabolism , Embryonic Development , HMGB Proteins/metabolism , Signal Transduction , Animals , DNA-Binding Proteins/genetics , Embryo Implantation/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , HMGB Proteins/genetics , Humans , Litter Size , Mice, Inbred ICR , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/genetics , Zygote/metabolism
9.
Reproduction ; 150(1): 21-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25918436

ABSTRACT

SET and MYND domain-containing protein 3 (Smyd3) is a histone H3 lysine 4 (H3K4) di- and tri-methyltransferase that forms a transcriptional complex with RNA polymerase II and activates the transcription of oncogenes and cell cycle genes in human cancer cells. However, the study of Smyd3 in mammalian early embryonic development has not yet been addressed. In the present study, we investigated the expression pattern of Smyd3 in mouse preimplantation embryos and the effects of RNA interference (RNAi)-mediated Smyd3 repression on the development of mouse embryos. We showed that Smyd3 mRNA levels increased after the two-cell stage, peaked at the four-cell stage, and gradually decreased thereafter. Moreover, in two-cell to eight-cell embryos, SMYD3 staining was more intense in the nuclei than it was in the cytoplasm. In Smyd3-knockdown embryos, the percentage of inner cell mass (ICM)-derived colony formation and trophectoderm (TE)-derived cell attachment were significantly decreased, which resulted in a reduction in the number of viable offspring. Furthermore, the expression of Oct4 and Cdx2 during mid-preimplantation gene activation was significantly decreased in Smyd3-knockdown embryos. In addition, the transcription levels of ICM and epiblast markers, such as Oct4, Nanog, and Sox2, the transcription levels of primitive endoderm markers, such as Gata6, and the transcription levels of TE markers, such as Cdx2 and Eomes, were significantly decreased in Smyd3-knockdown blastocysts. These findings indicate that SMYD3 plays an important role in early embryonic lineage commitment and peri-implantation development through the activation of lineage-specific genes.


Subject(s)
Embryo Implantation/genetics , Embryonic Development/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , Blastocyst/metabolism , CDX2 Transcription Factor , Female , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy , RNA Interference , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Reprod Dev ; 61(3): 241-4, 2015.
Article in English | MEDLINE | ID: mdl-25740401

ABSTRACT

Transgenic mice are important tools for genetic analysis. A current prominent method for producing transgenic mice involves pronuclear microinjection into 1-cell embryos. However, the total transgenic efficiency obtained using this method is less than 10%. Here, we demonstrate that highly efficient transgenesis in mice can be achieved by cytoplasmic microinjection using a hyperactive piggyBac system. In embryos in which hyPBase mRNA and pPB-CAG-TagRFP DNA were co-injected into the cytoplasm, TagRFP fluorescence was observed after the 2-cell stage; when 30 ng/µl pPB-CAG-TagRFP DNA and 30 ng/µl hyPBase mRNA were co-injected, 94.4% of blastocysts were TagRFP positive. Furthermore, a high concentration of hyPBase mRNA resulted in creation of mosaic embryos in which the TagRFP signals partially disappeared. However, suitable concentrations of injected DNA and hyPBase mRNA produced embryos in which almost all blastomeres were TagRFP positive. Thus, the hyperactive piggyBac transposon system is an easy-to-implement and highly effective method that can contribute to production of transgenic mice.


Subject(s)
Blastocyst/metabolism , DNA Transposable Elements , Gene Transfer Techniques , Animals , Blastocyst/cytology , Cytoplasm/metabolism , Embryo Culture Techniques/methods , Female , Luminescent Proteins/metabolism , Mice , Mice, Inbred ICR , Mice, Transgenic , Microinjections , RNA, Messenger/metabolism , Transgenes , Red Fluorescent Protein
11.
PLoS One ; 8(9): e74749, 2013.
Article in English | MEDLINE | ID: mdl-24066152

ABSTRACT

ING3 (inhibitor of growth family, member 3) is a subunit of the nucleosome acetyltransferase of histone 4 (NuA4) complex, which activates gene expression. ING3, which contains a plant homeodomain (PHD) motif that can bind to trimethylated lysine 4 on histone H3 (H3K4me3), is ubiquitously expressed in mammalian tissues and governs transcriptional regulation, cell cycle control, and apoptosis via p53-mediated transcription or the Fas/caspase-8 pathway. Thus, ING3 plays a number of important roles in various somatic cells. However, the role(s) of ING3 in germ cells remains unknown. Here, we show that loss of ING3 function led to the failure of asymmetric cell division and cortical reorganization in the mouse oocyte. Immunostaining showed that in fully grown germinal vesicle (GV) oocytes, ING3 localized predominantly in the GV. After germinal vesicle breakdown (GVBD), ING3 homogeneously localized in the cytoplasm. In oocytes where Ing3 was targeted by siRNA microinjection, we observed symmetric cell division during mouse oocyte maturation. In those oocytes, oocyte polarization was not established due to the failure to form an actin cap or a cortical granule-free domain (CGFD), the lack of which inhibited spindle migration. These features were among the main causes of abnormal symmetric cell division. Interestingly, an analysis of the mRNA expression levels of genes related to asymmetric cell division revealed that only mTOR was downregulated, and, furthermore, that genes downstream of mTOR (e.g., Cdc42, Rac1, and RhoA) were also downregulated in siIng3-injected oocytes. Therefore, ING3 may regulate asymmetric cell division through the mTOR pathway during mouse oocyte maturation.


Subject(s)
Asymmetric Cell Division/physiology , Homeodomain Proteins/metabolism , Oocytes/cytology , Oocytes/metabolism , Animals , Asymmetric Cell Division/genetics , Cells, Cultured , Female , Homeodomain Proteins/genetics , Mice , Mice, Inbred ICR , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
12.
Cell Metab ; 16(3): 394-406, 2012 Sep 05.
Article in English | MEDLINE | ID: mdl-22958922

ABSTRACT

Brown adipose tissue is attracting much attention due to its antiobestic effects; however, its development and involvement in metabolic improvement remain elusive. Here we established a method for a high-efficiency (>90%) differentiation of human pluripotent stem cells (hPSCs) into functional classical brown adipocytes (BAs) using specific hemopoietin cocktail (HC) without exogenous gene transfer. BAs were not generated without HC, and lack of a component of HC induced white adipocyte (WA) marker expressions. hPSC-derived BA (hPSCdBA) showed respiratory and thermogenic activation by ß-adrenergic receptor (AdrRß) stimuli and augmented lipid and glucose tolerance, whereas human multipotent stromal cell-derived WA (hMSCdWA) improved lipid but inhibited glucose metabolism. Cotransplantation of hPSCdBA normalized hMSCdWA-induced glucose intolerance. Surprisingly, hPSCdBAs expressed various hemopoietin genes, serving as stroma for myeloid progenitors. Moreover, AdrRß stimuli enhanced recovery from chemotherapy-induced myelosuppression. Our study enhances our understanding of BA, identifying roles in metabolic and hemogenic regulation.


Subject(s)
Adipocytes, Brown/cytology , Cell Differentiation/physiology , Hematopoietic Cell Growth Factors/pharmacology , Pluripotent Stem Cells/cytology , Receptors, Adrenergic, beta/metabolism , Adipocytes, Brown/metabolism , Blotting, Western , Cell Differentiation/drug effects , Glucose Tolerance Test , Hematopoietic Cell Growth Factors/metabolism , Humans , Microscopy, Electron , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thermogenesis/physiology
13.
Differentiation ; 84(5): 392-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22906707

ABSTRACT

While the role of p75(NTR) signaling in the regulation of nerve-related cell growth and survival has been well documented, its actions in osteoblasts are poorly understood. In this study, we examined the effects of p75(NTR) on osteoblast proliferation and differentiation using the MC3T3-E1 pre-osteoblast cell line. Proliferation and osteogenic differentiation were significantly enhanced in p75(NTR)-overexpressing MC3T3-E1 cells (p75GFP-E1). In addition, expression of osteoblast-specific osteocalcin (OCN), bone sialoprotein (BSP), and osterix mRNA, ALP activity, and mineralization capacity were dramatically enhanced in p75GFP-E1 cells, compared to wild MC3T3-E1 cells (GFP-E1). To determine the binding partner of p75(NTR) in p75GFP-E1 cells during osteogenic differentiation, we examined the expression of trkA, trkB, and trkC that are known binding partners of p75(NTR), as well as NgR. Pharmacological inhibition of trk tyrosine kinase with the K252a inhibitor resulted in marked reduction in the level of ALPase under osteogenic conditions. The deletion of the GDI binding domain in the p75(NTR)-GFP construct had no effect on mineralization. Taken together, our studies demonstrated that p75(NTR) signaling through the trk tyrosine kinase pathway affects osteoblast functions by targeting osteoblast proliferation and differentiation.


Subject(s)
Cell Differentiation , Osteoblasts/cytology , Receptors, Nerve Growth Factor/metabolism , Animals , Calcification, Physiologic , Carbazoles , Cell Culture Techniques , Cell Proliferation , Gene Expression Regulation , Indole Alkaloids , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Mice , Osteoblasts/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Protein Binding , Protein Kinase C/antagonists & inhibitors , RNA, Messenger/biosynthesis , Receptor, trkC/genetics , Receptor, trkC/metabolism , Receptors, Nerve Growth Factor/genetics , Signal Transduction , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism
14.
Cell Reprogram ; 13(4): 361-70, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21718107

ABSTRACT

Human-induced pluripotent stem cells (hiPSCs) are expected to become a powerful tool for regenerative medicine. Their efficacy in the use of clinical purposes is currently under intensive verification. It was reported that hiPSC-derived hemangioblasts had severely limited expansion capability due to an induction of early senescence: hiPSC-derived vascular endothelial cells (VECs) senesced after one passage and hiPSC-derived hematopoietic progenitor cells (HPCs) showed substantially decreased colony-forming activities. Here we show that early senescence is not an inevitable fate of hiPSC-derived cells. Applying our unique feeder-free culture methods for the differentiations of human embryonic stem cells (hESCs), we successfully generated VECs and HPCs from three lines of hiPSCs that were established by using a retrovirus vector system. All hiPS-derived VECs could be subcultured by 2:1∼3:1 dilutions up to 10∼20 passages, after which the cells underwent senescence. Among the three lines of hiPSCs, two lines generated HPCs that bore comparable granulocyte colony-forming units to those of hESCs. Moreover, one line effectively reproduced HPCs within the sac-like structures, the fields of in vitro hematopoiesis, as in the case of hESCs. Surprisingly, release of neutrophils into culture supernatant persisted even longer (∼60 days) than the case of hESCs (∼40 days). Thus, the problem of early senescence can be overcome by selecting appropriate lines of hiPSCs and applying proper differentiation methods to them.


Subject(s)
Cell Culture Techniques/methods , Cellular Senescence/physiology , Induced Pluripotent Stem Cells/physiology , Cell Differentiation/physiology , Cells, Cultured , Coculture Techniques/methods , Endothelial Cells/cytology , Feeder Cells/cytology , Hematopoietic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology
15.
Stem Cells Dev ; 20(5): 901-13, 2011 May.
Article in English | MEDLINE | ID: mdl-21142793

ABSTRACT

We describe a novel role for CD271 in the differentiation of mesenchymal stem cells (MSCs), including deciduous dental pulp stem cells (DDPSCs) and murine multipotent MSCs (C3H10T1/2 cells). The CD271(+) subpopulation of deciduous dental pulp cells (CD271(+)/DDPSCs) and the forced expression of CD271 in C3H10T1/2 (10T271) were analyzed by fluorescence-activated cell sorting. CD271 expression was detected in DDPSCs that expressed both CD44 and CD90, simultaneously, and the clonogenic capacity of the CD271(+)/DDPSCs was higher than that of the CD271(-)/DDPSCs that expressed both CD44 and CD90. Further, the differentiation of CD271(+)/DDPSCs into osteoblasts and adipocytes was inhibited although CD271(-)/DDPSCs were capable of differentiating into osteoblasts and adipocytes. CD271 was overexpressed in C3H10T1/2 cells, which have the potential to differentiate into osteoblasts, adipocytes, chondrocytes, and myocytes. CD271 inhibited the differentiation of C3H10T1/2 cells into any of these lineages. These results indicate a role for CD271 in inhibiting the differentiation of MSCs.


Subject(s)
Dental Pulp/cytology , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Differentiation , Cell Lineage/genetics , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Dental Pulp/metabolism , Flow Cytometry , Gene Expression Regulation, Developmental , Humans , Mesenchymal Stem Cells/metabolism , Mice , Multipotent Stem Cells/metabolism , Muscle Cells/cytology , Muscle Cells/metabolism , Nerve Tissue Proteins/genetics , Osteoblasts/cytology , Osteoblasts/metabolism , Plasmids , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Nerve Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tooth, Deciduous/cytology , Tooth, Deciduous/metabolism , Transfection
16.
Article in English | MEDLINE | ID: mdl-20123389

ABSTRACT

OBJECTIVES: Bacterial metabolites demineralize dental hard tissues, and soluble factors lead to tertiary dentinogenesis in the area of the dentin-pulp complex. However, it is unclear whether the oral bacteria are directly involved in the differentiation of dental pulp cells. In this study, we evaluated the effect of oral bacterial extracts on cellular differentiation in human dental pulp-derived cells (hDPC). STUDY DESIGN: The hDPC were obtained from third molar teeth, and the cells were subcultured. The sonicated extracts were obtained from Porphyromonas gingivalis (gram-negative) and Streptococcus mutans (gram-positive). The effect of bacterial extracts on cellular growth and differentiation in hDPC were tested. RESULTS: Alkaline phosphatase activity and bone sialoprotein (BSP) gene expression were increased in hDPC exposed to low concentrations of both sonicated extracts, whereas the activity was decreased upon exposure to high concentrations of sonicated extracts from P. gingivalis. CONCLUSION: This is the first evidence that oral bacteria have a positive effect on cellular differentiation in hPDC.


Subject(s)
Culture Media, Conditioned/pharmacology , Dental Pulp/drug effects , Dentinogenesis/drug effects , Osteogenesis/drug effects , Porphyromonas gingivalis , Streptococcus mutans , Analysis of Variance , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dental Pulp/cytology , Dental Pulp/metabolism , Endotoxins , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Gene Expression , Humans , Integrin-Binding Sialoprotein , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Sialoglycoproteins/biosynthesis , Sialoglycoproteins/genetics , Sonication , Statistics, Nonparametric
17.
J Artif Organs ; 6(2): 145-8, 2003.
Article in English | MEDLINE | ID: mdl-14598116

ABSTRACT

In this paper, the authors propose a novel electric power supply system for implanted medical devices. The system is noninvasive and uses two kinds of energy, magnetic and ultrasonic. The system can provide high power levels harmlessly. The energies are obtained by two types of vibrator, i.e., piezo and magnetostriction devices. A prototype was built and it was verified experimentally that the system is basically able to provide power. At high frequencies, such as 100 kHz, the output power was higher than the conventional system using a transformer. The normalized output power per unit volume also exceeded the transformer system.


Subject(s)
Electric Conductivity , Electric Power Supplies , Electronics, Medical/instrumentation , Magnetics , Prostheses and Implants , Ultrasonics , Equipment Design , Humans , Materials Testing
SELECTION OF CITATIONS
SEARCH DETAIL
...