Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Dev Dyn ; 248(10): 931-941, 2019 10.
Article in English | MEDLINE | ID: mdl-31301196

ABSTRACT

BACKGROUND: The timing of developmental events is tightly regulated along a time axis for normal development. Although the RNA-binding protein Lin28a plays a crucial role in the regulation of developmental timing in Caenorhabditis elegans, how the timing of Lin28a expression affects the rate and/or duration of developmental events during mammalian development remains to be addressed. RESULTS: In this study, we discovered that the timing and the duration of Lin28a expression affect embryonic growth. During the neurulation stage of mouse development, endogenous Lin28a levels start to drop. When Lin28a expression was maintained transiently using the inducible tetracycline-regulated gene expression (Tet-ON) system [doxycycline (Dox)-inducible Lin28a transgenic (iLin28a Tg) mice] with Dox administration at E8.5 and E9.5, it resulted in neonatal lethality, increased body weight (organomegaly), and an increased number of caudal vertebrae at birth. On the other hand, Lin28a induction only at E8.5 caused neonatal lethality and organomegaly, but did not affect the caudal vertebra number. Of note, although Dox treatment before or after neurulation still caused neonatal lethality, it neither caused organomegaly nor the increased caudal vertebra number in iLin28a Tg neonates. CONCLUSIONS: Temporal regulation of Lin28a expression during neurulation affects developmental events such as cessation of axial elongation and embryonic growth in mice.


Subject(s)
Body Size , Neurulation/physiology , RNA-Binding Proteins/physiology , Animals , Animals, Newborn , Doxycycline/pharmacology , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , RNA-Binding Proteins/metabolism , Time Factors
2.
Placenta ; 84: 50-56, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31272680

ABSTRACT

The placenta is composed of the amnion, chorionic plate, villous and smooth chorion, decidua basalis, and umbilical cord. The amnion is a readily obtainable source of a large number of cells and cell types, including epithelium, mesenchyme, and endothelium, and is thus an allogeneic resource for regenerative medicine. Endothelial cells are obtained from large arteries and veins in the amniotic membrane as well as the umbilical cord. The amnion-derived cells exhibit transdifferentiation capabilities, including chondrogenesis and cardiomyogenesis, by introduction of transcription factors, in addition to their original and potential phenotypes. The amnion is also a source for production of induced pluripotent stem cells (AM-iPSCs). The AM-iPSCs exhibit stable phenotypes, such as multipotency and immortality, and a unique gene expression pattern. Through the use of amnion-derived cells, as well as other placenta-derived cells, preclinical proof of concept has been achieved in a mouse model of muscular dystrophy.


Subject(s)
Amnion/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Regenerative Medicine/methods , Regenerative Medicine/trends , Animals , Cell Differentiation , Cell Separation , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/physiology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cell Transplantation/trends , Mice , Pregnancy , Reproducibility of Results
4.
PLoS One ; 10(5): e0126562, 2015.
Article in English | MEDLINE | ID: mdl-25978455

ABSTRACT

Comprehensive analysis of alterations in gene expression along with neoplastic transformation in human cells provides valuable information about the molecular mechanisms underlying transformation. To further address these questions, we performed whole transcriptome analysis to the human mesenchymal stem cell line, UE6E7T-3, which was immortalized with hTERT and human papillomavirus type 16 E6/E7 genes, in association with progress of transformation in these cells. At early stages of culture, UE6E7T-3 cells preferentially lost one copy of chromosome 13, as previously described; in addition, tumor suppressor genes, DNA repair genes, and apoptosis-activating genes were overexpressed. After the loss of chromosome 13, additional aneuploidy and genetic alterations that drove progressive transformation, were observed. At this stage, the cell line expressed oncogenes as well as genes related to anti-apoptotic functions, cell-cycle progression, and chromosome instability (CIN); these pro-tumorigenic changes were concomitant with a decrease in tumor suppressor gene expression. At later stages after prolong culture, the cells exhibited chromosome translocations, acquired anchorage-independent growth and tumorigenicity in nude mice, (sarcoma) and exhibited increased expression of genes encoding growth factor and DNA repair genes, and decreased expression of adhesion genes. In particular, glypican-5 (GPC5), which encodes a cell-surface proteoglycan that might be a biomarker for sarcoma, was expressed at high levels in association with transformation. Patched (Ptc1), the cell surface receptor for hedgehog (Hh) signaling, was also significantly overexpressed and co-localized with GPC5. Knockdown of GPC5 expression decreased cell proliferation, suggesting that it plays a key role in growth in U3-DT cells (transformants derived from UE6E7T-3 cells) through the Hh signaling pathway. Thus, the UE6E7T-3 cell culture model is a useful tool for assessing the functional contribution of genes showed by expression profiling to the neoplastic transformation of human fibroblasts and human mesenchymal stem cells (hMSC).


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Viral/genetics , Mesenchymal Stem Cells/metabolism , Transcription, Genetic/genetics , Aneuploidy , Animals , Cell Culture Techniques , Cell Cycle/genetics , Cell Proliferation/genetics , Chromosomal Instability/genetics , Chromosomes, Human, Pair 13/genetics , DNA Repair/genetics , Fibroblasts/metabolism , Glypicans/genetics , Hedgehogs/genetics , Human papillomavirus 6/genetics , Humans , Mice , Mice, Nude , Oncogenes/genetics , Signal Transduction/genetics , Telomerase/genetics , Transcriptional Activation/immunology
5.
Expert Opin Biol Ther ; 14(12): 1731-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25241883

ABSTRACT

BACKGROUND: Establishment of human osteoblast cultures that retain bone-forming capacity is one of the prerequisites for successful bone regeneration therapy. Because osteoblasts harvested from adults exhibit limited growth, the use of immature osteoblasts that can expand ex vivo should greatly facilitate bone regeneration therapy. In this study, we developed immature human osteoblasts isolated from aged alveolar bone (HAOBs). METHODS: HAOBs obtained after the collagenase digestion of alveolar bones from elderly donors. Then, we assessed osteogenic ability of HAOB after treatment with recombinant human bone morphogenic protein-2 or transplantation into immunodeficient mice. In addition, we performed global gene expression analysis to identify functional marker for HAOB. RESULTS: HAOBs, which can differentiate into osteoblasts and have a robust bone-forming ability, were successfully extracted from donors who were > 60 years of age. We found that the HAOBs exhibited a higher osteogenic ability compared with those of human mesenchymal stem cells and highly expressed NEBULETTE (NEBL) with osteogenic abilities. CONCLUSIONS: HAOBs have properties similar to those of human immature osteoblasts and appear to be a novel material for cell-based bone regeneration therapy. Additionally, the expression level of NEBL may serve as a marker for the osteogenic ability of these cells.


Subject(s)
Aging , Alveolar Process/cytology , Bone Regeneration , Guided Tissue Regeneration , Osteoblasts/cytology , Tissue Donors , Adult , Aging/physiology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Separation , Cells, Cultured , Guided Tissue Regeneration/methods , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Mice, SCID , Middle Aged , Osteoblasts/physiology , Osteogenesis/physiology
6.
Mol Biol Cell ; 23(18): 3511-21, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22833560

ABSTRACT

Cellular differentiation and lineage commitment are considered to be robust and irreversible processes during development. Recent work has shown that mouse and human fibroblasts can be reprogrammed to a pluripotent state with a combination of four transcription factors. We hypothesized that combinatorial expression of chondrocyte-specific transcription factors could directly convert human placental cells into chondrocytes. Starting from a pool of candidate genes, we identified a combination of only five genes (5F pool)-BCL6, T (also called BRACHYURY), c-MYC, MITF, and BAF60C (also called SMARCD3)-that rapidly and efficiently convert postnatal human chorion and decidual cells into chondrocytes. The cells generated expressed multiple cartilage-specific genes, such as Collagen type II α1, LINK PROTEIN-1, and AGGRECAN, and exhibited characteristics of cartilage both in vivo and in vitro. Expression of the endogenous genes for T and MITF was initiated, implying that the cell conversion is due to not only the forced expression of the transgenes, but also to cellular reprogramming by the transgenes. This direct conversion system from noncartilage tissue to cartilaginous tissue is a substantial advance toward understanding cartilage development, cell-based therapy, and oncogenesis of chondrocytes.


Subject(s)
Cartilage/cytology , Cell Dedifferentiation , Chondrocytes/cytology , Placenta/cytology , Adult , Animals , Blotting, Western , Cartilage/metabolism , Cells, Cultured , Chondrocytes/metabolism , Chondrogenesis/genetics , Chorion/cytology , Chorion/metabolism , Chromosomal Proteins, Non-Histone , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Decidua/cytology , Decidua/metabolism , Female , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Mice , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Placenta/metabolism , Pregnancy , Proto-Oncogene Proteins c-bcl-6 , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transfection
7.
Arthritis Res Ther ; 14(3): R136, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22676383

ABSTRACT

INTRODUCTION: Transplantation of mesenchymal stem cells (MSCs) derived from synovium is a promising therapy for cartilage regeneration. For clinical application, improvement of handling operation, enhancement of chondrogenic potential, and increase of MSCs adhesion efficiency are needed to achieve a more successful cartilage regeneration with a limited number of MSCs without scaffold. The use of aggregated MSCs may be one of the solutions. Here, we investigated the handling, properties and effectiveness of aggregated MSCs for cartilage regeneration. METHODS: Human and rabbit synovial MSCs were aggregated using the hanging drop technique. The gene expression changes after aggregation of synovial MSCs were analyzed by microarray and real time RT-PCR analyses. In vitro and in vivo chondrogenic potential of aggregates of synovial MSCs was examined. RESULTS: Aggregates of MSCs cultured for three days became visible, approximately 1 mm in diameter and solid and durable by manipulation; most of the cells were viable. Microarray analysis revealed up-regulation of chondrogenesis-related, anti-inflammatory and anti-apoptotic genes in aggregates of MSCs. In vitro studies showed higher amounts of cartilage matrix synthesis in pellets derived from aggregates of MSCs compared to pellets derived from MSCs cultured in a monolayer. In in vivo studies in rabbits, aggregates of MSCs could adhere promptly on the osteochondral defects by surface tension, and stay without any loss. Transplantation of aggregates of MSCs at relatively low density achieved successful cartilage regeneration. Contrary to our expectation, transplantation of aggregates of MSCs at high density failed to regenerate cartilage due to cell death and nutrient deprivation of aggregates of MSCs. CONCLUSIONS: Aggregated synovial MSCs were a useful source for cartilage regeneration considering such factors as easy preparation, higher chondrogenic potential and efficient attachment.


Subject(s)
Cartilage/cytology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Tissue Engineering/methods , Animals , Cartilage/physiology , Cell Differentiation/physiology , Chondrogenesis/physiology , Humans , In Situ Nick-End Labeling , Oligonucleotide Array Sequence Analysis , Rabbits , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Synovial Membrane , Transcriptome
8.
PLoS One ; 7(1): e29677, 2012.
Article in English | MEDLINE | ID: mdl-22276123

ABSTRACT

Hexagonal-shaped human corneal endothelial cells (HCEC) form a monolayer by adhering tightly through their intercellular adhesion molecules. Located at the posterior corneal surface, they maintain corneal translucency by dehydrating the corneal stroma, mainly through the Na(+)- and K(+)-dependent ATPase (Na(+)/K(+)-ATPase). Because HCEC proliferative activity is low in vivo, once HCEC are damaged and their numbers decrease, the cornea begins to show opacity due to overhydration, resulting in loss of vision. HCEC cell cycle arrest occurs at the G1 phase and is partly regulated by cyclin-dependent kinase inhibitors (CKIs) in the Rb pathway (p16-CDK4/CyclinD1-pRb). In this study, we tried to activate proliferation of HCEC by inhibiting CKIs. Retroviral transduction was used to generate two new HCEC lines: transduced human corneal endothelial cell by human papillomavirus type E6/E7 (THCEC (E6/E7)) and transduced human corneal endothelial cell by Cdk4R24C/CyclinD1 (THCEH (Cyclin)). Reverse transcriptase polymerase chain reaction analysis of gene expression revealed little difference between THCEC (E6/E7), THCEH (Cyclin) and non-transduced HCEC, but cell cycle-related genes were up-regulated in THCEC (E6/E7) and THCEH (Cyclin). THCEH (Cyclin) expressed intercellular molecules including ZO-1 and N-cadherin and showed similar Na(+)/K(+)-ATPase pump function to HCEC, which was not demonstrated in THCEC (E6/E7). This study shows that HCEC cell cycle activation can be achieved by inhibiting CKIs even while maintaining critical pump function and morphology.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Corneal/cytology , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cyclin D1/genetics , Cyclin D1/metabolism , Human papillomavirus 6/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium-Potassium-Exchanging ATPase/genetics , Sodium-Potassium-Exchanging ATPase/metabolism , Transduction, Genetic , Zonula Occludens-1 Protein
9.
J Artif Organs ; 14(3): 215-22, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21534010

ABSTRACT

Low efficiencies of nonviral gene vectors, such as transfection reagent, limit their utility in gene therapy. To overcome this disadvantage, we report on the preparation and properties of magnetic nanoparticles [diameter (d) = 121.32 ± 27.36 nm] positively charged by cationic polymer deacylated polyethylenimine (PEI max), which boosts gene delivery efficiency compare with polyethylenimine (PEI), and their use for the forced expression of plasmid delivery by application of a magnetic field. Magnetic nanoparticles were coated with PEI max, which enabled their electrostatic interaction with negatively charged molecules such as plasmid. We successfully transfected 81.1 ± 4.0% of the cells using PEI max-coated magnetic nanoparticles (PEI max-nanoparticles). Along with their superior properties as a DNA delivery vehicle, PEI max-nanoparticles offer to deliver various DNA formulations in addition to traditional methods. Furthermore, efficiency of the gene transfer was not inhibited in the presence of serum in the cells. PEI max-nanoparticles may be a promising gene carrier that has high transfection efficiency as well as low cytotoxicity.


Subject(s)
Nanoparticles , Polyethyleneimine , Transfection/methods , Animals , Cell Line , Cells, Cultured , Drug Carriers , Mice
10.
Hum Mol Genet ; 20(2): 235-44, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-20947660

ABSTRACT

Human leukocyte antigen (HLA)-E is a non-classical major histocompatibility complex class I (Ib) molecule, which plays an important role in immunosuppression. In this study, we investigated the immunomodulating effect of HLA-E in a xenogeneic system, using human placental artery-derived endothelial (hPAE) cells expressing HLA-E in a mouse model. In vitro cell lysis analysis by primed lymphocytes in combination with siRNA transfection showed that HLA-E is necessary for inhibition of the immune response. Similarly, in vivo cell implantation analysis with siRNA-mediated down-regulation of HLA-E demonstrates that HLA-E is involved in immunosuppression. As hPAE cells efficiently transdifferentiate into myoblasts/myocytes in vitro, we transplanted the cells into mdx mice, a model of Duchenne muscular dystrophy. hPAE cells conferred dystrophin to myocytes of the 'immunocompetent' mdx mice with extremely high efficiency. These findings suggest that HLA-E-expressing cells with a myogenic potential represent a promising source for cell-based therapy of patients with muscular dystrophy.


Subject(s)
Dystrophin/genetics , Dystrophin/metabolism , Endothelial Cells , Histocompatibility Antigens Class I/genetics , Muscular Dystrophy, Duchenne/genetics , Animals , Arteries/cytology , Cell Culture Techniques , Disease Models, Animal , Endothelial Cells/metabolism , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Immunocompetence/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred mdx , Muscle Development/genetics , Muscular Dystrophy, Duchenne/immunology , Muscular Dystrophy, Duchenne/therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , HLA-E Antigens
11.
PLoS One ; 5(9): e13017, 2010 Sep 27.
Article in English | MEDLINE | ID: mdl-20885964

ABSTRACT

BACKGROUND: Human induced pluripotent stem (iPS) cells are currently used as powerful resources in regenerative medicine. During very early developmental stages, DNA methylation decreases to an overall low level at the blastocyst stage, from which embryonic stem cells are derived. Therefore, pluripotent stem cells, such as ES and iPS cells, are considered to have hypo-methylated status compared to differentiated cells. However, epigenetic mechanisms of "stemness" remain unknown in iPS cells derived from extra-embryonic and embryonic cells. METHODOLOGY/PRINCIPAL FINDINGS: We examined genome-wide DNA methylation (24,949 CpG sites covering 1,3862 genes, mostly selected from promoter regions) with six human iPS cell lines derived from human amniotic cells and fetal lung fibroblasts as well as two human ES cell lines, and eight human differentiated cell lines using Illumina's Infinium HumanMethylation27. A considerable fraction (807 sites) exhibited a distinct difference in the methylation level between the iPS/ES cells and differentiated cells, with 87.6% hyper-methylation seen in iPS/ES cells. However, a limited fraction of CpG sites with hypo-methylation was found in promoters of genes encoding transcription factors. Thus, a group of genes becomes active through a decrease of methylation in their promoters. Twenty-three genes including SOX15, SALL4, TDGF1, PPP1R16B and SOX10 as well as POU5F1 were defined as genes with hypo-methylated SS-DMR (Stem cell-Specific Differentially Methylated Region) and highly expression in iPS/ES cells. CONCLUSIONS/SIGNIFICANCE: We show that DNA methylation profile of human amniotic iPS cells as well as fibroblast iPS cells, and defined the SS-DMRs. Knowledge of epigenetic information across iPS cells derived from different cell types can be used as a signature for "stemness" and may allow us to screen for optimum iPS/ES cells and to validate and monitor iPS/ES cell derivatives for human therapeutic applications.


Subject(s)
Amnion/cytology , DNA Methylation , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Lung/cytology , Promoter Regions, Genetic , Amnion/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Embryonic Stem Cells/cytology , Epigenesis, Genetic , Fibroblasts/cytology , Humans , Induced Pluripotent Stem Cells/cytology , Lung/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Circ Res ; 106(10): 1613-23, 2010 May 28.
Article in English | MEDLINE | ID: mdl-20508201

ABSTRACT

RATIONALE: Amniotic membrane is known to have the ability to transdifferentiate into multiple organs and is expected to stimulate a reduced immunologic reaction. OBJECTIVE: Determine whether human amniotic membrane-derived mesenchymal cells (hAMCs) can be an ideal allograftable stem cell source for cardiac regenerative medicine. METHODS AND RESULTS: We established hAMCs. After cardiomyogenic induction in vitro, hAMCs beat spontaneously, and the calculated cardiomyogenic transdifferentiation efficiency was 33%. Transplantation of hAMCs 2 weeks after myocardial infarction improved impaired left ventricular fractional shortening measured by echocardiogram (34+/-2% [n=8] to 39+/-2% [n=11]; P<0.05) and decreased myocardial fibrosis area (18+/-1% [n=9] to 13+/-1% [n=10]; P<0.05), significantly. Furthermore hAMCs transplanted into the infarcted myocardium of Wistar rats were transdifferentiated into cardiomyocytes in situ and survived for more than 4 weeks after the transplantation without using any immunosuppressant. Immunologic tolerance was caused by the hAMC-derived HLA-G expression, lack of MHC expression of hAMCs, and activation of FOXP3-positive regulatory T cells. Administration of IL-10 or progesterone, which is known to play an important role in feto-maternal tolerance during pregnancy, markedly increased HLA-G expression in hAMCs in vitro and, surprisingly, also increased cardiomyogenic transdifferentiation efficiency in vitro and in vivo. CONCLUSIONS: Because hAMCs have a high ability to transdifferentiate into cardiomyocytes and to acquire immunologic tolerance in vivo, they can be a promising cellular source for allograftable stem cells for cardiac regenerative medicine.


Subject(s)
Amnion/cytology , Amnion/physiology , Mesenchymal Stem Cell Transplantation/methods , Myocytes, Cardiac/cytology , Transplantation, Heterologous/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Delivery, Obstetric , Echocardiography , Female , Graft Rejection/prevention & control , Heart/physiology , Humans , Infant, Newborn , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Myocytes, Cardiac/physiology , Pregnancy , Rats , Rats, Wistar , Transplantation Tolerance , Ventricular Function, Left/physiology
13.
Am J Pathol ; 176(4): 1973-82, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20203285

ABSTRACT

POU5F1 is a transcription factor essential for the self-renewal activity and pluripotency of embryonic stem cells and germ cells. We have previously reported that POU5F1 is fused to EWSR1 in a case of undifferentiated sarcoma with chromosomal translocation t(6;22)(p21;q12). In addition, the EWS-POU5F1 chimeras have been recently identified in human neoplasms of the skin and salivary glands. To clarify the roles of the EWS-POU5F1 chimera in tumorigenesis and tumor cell maintenance, we used small-interfering RNA-mediated gene silencing. Knockdown of EWS-POU5F1 in the t(6;22) sarcoma-derived GBS6 cell line resulted in a significant decrease of cell proliferation because of G1 cell cycle arrest associated with p27(Kip1) up-regulation. Moreover, senescence-like morphological changes accompanied by actin polymerization were observed. In contrast, EWS-POU5F1 down-regulation markedly increased the cell migration and invasion as well as activation of metalloproteinase 2 and metalloproteinase 14. The results indicate that the proliferative activity of cancer cells and cell motility are discrete processes in multistep carcinogenesis. These findings reveal the functional role of the sarcoma-related chimeric protein as well as POU5F1 in the development and progression of human neoplasms.


Subject(s)
Octamer Transcription Factor-3/biosynthesis , Octamer Transcription Factor-3/genetics , RNA-Binding Protein EWS/biosynthesis , RNA-Binding Protein EWS/genetics , Sarcoma/pathology , Cell Line, Tumor , Cell Movement , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p27 , Flow Cytometry/methods , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Invasiveness , Oligonucleotide Array Sequence Analysis , RNA Interference , RNA, Small Interfering/metabolism , Sarcoma/metabolism , Skin Neoplasms/pathology
14.
J Cell Physiol ; 223(3): 695-702, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20162617

ABSTRACT

Duchenne muscular dystrophy is an X-linked recessive genetic disease characterized by severe skeletal muscular degeneration. The placenta is considered to be a promising candidate cell source for cellular therapeutics because it contains a large number of cells and heterogenous cell populations with myogenic potentials. We analyzed the myogenic potential of cells obtained from six parts of the placenta, that is, umbilical cord, amniotic epithelium, amniotic mesoderm, chorionic plate, villous chorion, and decidua basalis. In vitro cells derived from amniotic mesoderm, chorionic plate, and villous chorion efficiently transdifferentiate into myotubes. In addition, in vivo implantation of placenta-derived cells into dystrophic muscles of immunodeficient mdx mice restored sarcolemmal expression of human dystrophin. Differential contribution to myogenesis in this study may be attributed to placental portion-dependent default cell state. Molecular taxonomic characterization of placenta-derived maternal and fetal cells in vitro will help determine the feasibility of cell-based therapy.


Subject(s)
Dystrophin/metabolism , Extraembryonic Membranes/cytology , Extraembryonic Membranes/transplantation , Mesoderm/cytology , Mesoderm/transplantation , Muscular Dystrophy, Duchenne/therapy , Animals , Biomarkers/metabolism , Cell Membrane/metabolism , Cell Shape , Cell Transplantation , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred mdx , Mice, SCID , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Oligonucleotide Array Sequence Analysis , Placenta/cytology , Pregnancy
15.
Genes Cells ; 14(12): 1395-404, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19912344

ABSTRACT

Practical clinical applications for current induced pluripotent stem cell (iPSC) technologies are hindered by very low generation efficiencies. Here, we demonstrate that newborn human (h) and mouse (m) extra-embryonic amnion (AM) and yolk-sac (YS) cells, in which endogenous KLF4/Klf4, c-MYC/c-Myc and RONIN/Ronin are expressed, can be reprogrammed to hiPSCs and miPSCs with efficiencies for AM cells of 0.02% and 0.1%, respectively. Both hiPSC and miPSCs are indistinguishable from embryonic stem cells in colony morphology, expression of pluripotency markers, global gene expression profile, DNA methylation status of OCT4 and NANOG, teratoma formation and, in the case of miPSCs, generation of germline transmissible chimeric mice. As copious amounts of human AM cells can be collected without invasion, and stored long term by conventional means without requirement for in vitro culture, they represent an ideal source for cell banking and subsequent 'on demand' generation of hiPSCs for personal regenerative and pharmaceutical applications.


Subject(s)
Amnion/cytology , Cellular Reprogramming/physiology , Embryonic Stem Cells/physiology , Pluripotent Stem Cells/physiology , Yolk Sac/cytology , Amnion/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation , Chimera , DNA Methylation , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Immunoenzyme Techniques , Infant, Newborn , Kruppel-Like Factor 4 , Male , Mice , Mice, Transgenic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Teratoma/metabolism , Teratoma/pathology , Yolk Sac/metabolism
16.
Exp Cell Res ; 315(16): 2727-40, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19559696

ABSTRACT

POU5F1 (more commonly known as OCT4/3) is one of the stem cell markers, and affects direction of differentiation in embryonic stem cells. To investigate whether cells of mesenchymal origin acquire embryonic phenotypes, we generated human cells of mesodermal origin with overexpression of the chimeric OCT4/3 gene with physiological co-activator EWS (product of the EWSR1 gene), which is driven by the potent EWS promoter by translocation. The cells expressed embryonic stem cell genes such as NANOG, lost mesenchymal phenotypes, and exhibited embryonal stem cell-like alveolar structures when implanted into the subcutaneous tissue of immunodeficient mice. Hierarchical analysis by microchip analysis and cell surface analysis revealed that the cells are subcategorized into the group of human embryonic stem cells and embryonal carcinoma cells. These results imply that cells of mesenchymal origin can be traced back to cells of embryonic phenotype by the OCT4/3 gene in collaboration with the potent cis-regulatory element and the fused co-activator. The cells generated in this study with overexpression of chimeric OCT4/3 provide us with insight into cell plasticity involving OCT4/3 that is essential for embryonic cell maintenance, and the complexity required for changing cellular identity.


Subject(s)
Embryo, Mammalian , Mesoderm/physiology , Morphogenesis/physiology , Octamer Transcription Factor-3 , RNA-Binding Protein EWS/metabolism , Recombinant Fusion Proteins , Animals , Biomarkers/metabolism , Cell Lineage , Cells, Cultured , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Karyotyping , Mice , Mice, Inbred NOD , Mice, SCID , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Principal Component Analysis , RNA-Binding Protein EWS/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Teratoma/metabolism , Teratoma/pathology
17.
Biomed Chromatogr ; 23(12): 1245-50, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19488985

ABSTRACT

A new method for the rapid and simultaneous detection of ketamine and its major metabolite, norketamine, in rat hair has been developed by combining micropulverized extraction and ultraperformance liquid chromatography-electrospray ionization mass spectrometry. By using reversed-phase UPLC, ketamine and norketamine were well separated within 2 min. Using ketamine-dosed rat hair, the conditions for micropulverized extraction were optimized, and the limits of detection and quantification of the developed method were found to be 1.7 and 5.7 pg/mg hair for ketamine, respectively. The precisions achieved with this method were slightly better than that obtained with conventional acidic methanol extraction method. Using this proposed method, analysis of the washed rat hair could be completed within 16-17 min. This method is expected to be applied for the analysis of the hair samples of not only rats but also ketamine abusers.


Subject(s)
Anesthetics, Dissociative/analysis , Chromatography, Liquid/methods , Hair/chemistry , Ketamine/analogs & derivatives , Ketamine/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Rats
18.
J Orthop Res ; 27(4): 435-41, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18973232

ABSTRACT

Mesenchymal stem cells (MSCs) can be obtained from various tissues, and contain common features. However, an increasing number of reports have described variant properties dependent of cell sources. We examined (1) whether MSCs existed in several intraarticular tissues, (2) whether gene expression profiles in intraarticular tissue MSCs closely resembled each other, and (3) whether identified genes were specific to intraarticular tissue MSCs. Human synovium, meniscus, intraarticular ligament, muscle, adipose tissue, and bone marrow were harvested, and colony-forming cells were analyzed. All these cells showed multipotentiality and surface markers typical of MSCs. Gene profiles of intraarticular tissue MSCs and chondrocytes were closer to each other than those of extraarticular tissues MSCs. Among three characteristic genes specific for intraarticular tissue MSCs, we focused on proline arginine-rich end leucine-rich repeat protein (PRELP). Higher expression of PRELP was confirmed in chondrocytes and intraarticular tissue MSCs among three elderly and three young donors. Synovium MSCs stably expressed PRELP, contrarily, bone marrow MSCs increased PRELP expression during in vitro chondrogenesis. In conclusion, MSCs could be isolated from various intraarticular tissues including meniscus and ligament, gene expression profiles of intraarticular tissue MSCs closely resembled each other, and the higher expression of PRELP was characteristic of intraarticular tissue MSCs.


Subject(s)
Anterior Cruciate Ligament/cytology , Chondrocytes/cytology , Gene Expression Profiling , Menisci, Tibial/cytology , Mesenchymal Stem Cells/metabolism , Synovial Membrane/cytology , Aged , Cell Differentiation , Extracellular Matrix Proteins/genetics , Female , Glycoproteins/genetics , Humans
19.
PLoS One ; 3(12): e3945, 2008.
Article in English | MEDLINE | ID: mdl-19079602

ABSTRACT

BACKGROUND: Cigarette smoking adversely affects endochondral ossification during the course of skeletal growth. Among a plethora of cigarette chemicals, nicotine is one of the primary candidate compounds responsible for the cause of smoking-induced delayed skeletal growth. However, the possible mechanism of delayed skeletal growth caused by nicotine remains unclarified. In the last decade, localization of neuronal nicotinic acetylcholine receptor (nAChR), a specific receptor of nicotine, has been widely detected in non-excitable cells. Therefore, we hypothesized that nicotine affect growth plate chondrocytes directly and specifically through nAChR to delay skeletal growth. METHODOLOGY/PRINCIPAL FINDINGS: We investigated the effect of nicotine on human growth plate chondrocytes, a major component of endochondral ossification. The chondrocytes were derived from extra human fingers. Nicotine inhibited matrix synthesis and hypertrophic differentiation in human growth plate chondrocytes in suspension culture in a concentration-dependent manner. Both human and murine growth plate chondrocytes expressed alpha7 nAChR, which constitutes functional homopentameric receptors. Methyllycaconitine (MLA), a specific antagonist of alpha7 nAChR, reversed the inhibition of matrix synthesis and functional calcium signal by nicotine in human growth plate chondrocytes in vitro. To study the effect of nicotine on growth plate in vivo, ovulation-controlled pregnant alpha7 nAChR +/- mice were given drinking water with or without nicotine during pregnancy, and skeletal growth of their fetuses was observed. Maternal nicotine exposure resulted in delayed skeletal growth of alpha7 nAChR +/+ fetuses but not in alpha7 nAChR -/- fetuses, implying that skeletal growth retardation by nicotine is specifically mediated via fetal alpha7 nAChR. CONCLUSIONS/SIGNIFICANCE: These results suggest that nicotine, from cigarette smoking, acts directly on growth plate chondrocytes to decrease matrix synthesis, suppress hypertrophic differentiation via alpha7 nAChR, leading to delayed skeletal growth.


Subject(s)
Bone Development/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Growth Plate/cytology , Nicotine/pharmacology , Receptors, Nicotinic/metabolism , Alginates/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cells, Cultured , Chondrocytes/cytology , Female , Gels , Glucuronic Acid/metabolism , Hexuronic Acids/metabolism , Humans , Maternal Exposure , Mice , Microspheres , Mutation/genetics , Protein Subunits/metabolism , Protein Transport/drug effects , Sepharose , Time Factors , alpha7 Nicotinic Acetylcholine Receptor
20.
PLoS One ; 3(11): e3709, 2008.
Article in English | MEDLINE | ID: mdl-19002264

ABSTRACT

BACKGROUND: The sclera maintains and protects the eye ball, which receives visual inputs. Although the sclera does not contribute significantly to visual perception, scleral diseases such as refractory scleritis, scleral perforation and pathological myopia are considered incurable or difficult to cure. The aim of this study is to identify characteristics of the human sclera as one of the connective tissues derived from the neural crest and mesoderm. METHODOLOGY/PRINCIPAL FINDINGS: We have demonstrated microarray data of cultured human infant scleral cells. Hierarchical clustering was performed to group scleral cells and other mesenchymal cells into subcategories. Hierarchical clustering analysis showed similarity between scleral cells and auricular cartilage-derived cells. Cultured micromasses of scleral cells exposed to TGF-betas and BMP2 produced an abundant matrix. The expression of cartilage-associated genes, such as Indian hedge hog, type X collagen, and MMP13, was up-regulated within 3 weeks in vitro. These results suggest that human 'sclera'-derived cells can be considered chondrocytes when cultured ex vivo. CONCLUSIONS/SIGNIFICANCE: Our present study shows a chondrogenic potential of human sclera. Interestingly, the sclera of certain vertebrates, such as birds and fish, is composed of hyaline cartilage. Although the human sclera is not a cartilaginous tissue, the human sclera maintains chondrogenic potential throughout evolution. In addition, our findings directly explain an enigma that the sclera and the joint cartilage are common targets of inflammatory cells in rheumatic arthritis. The present global gene expression database will contribute to the clarification of the pathogenesis of developmental diseases such as high myopia.


Subject(s)
Cartilage/metabolism , Evolution, Molecular , Sclera/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cartilage/cytology , Cells, Cultured , Chondrogenesis/genetics , Gene Expression , Humans , Sclera/cytology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...