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










Publication year range
1.
Sci Rep ; 14(1): 6777, 2024 03 21.
Article in English | MEDLINE | ID: mdl-38514682

ABSTRACT

Extracellular matrix (ECM) is an intricate structure providing the microenvironment niche that influences stem cell differentiation. This study aimed to investigate the efficacy of decellularized ECM derived from human dental pulp stem cells (dECM_DPSCs) and gingival-derived mesenchymal stem cells (dECM_GSCs) as an inductive scaffold for osteogenic differentiation of GSCs. The proteomic analysis demonstrated that common and signature matrisome proteins from dECM_DPSCs and dECM_GSCs were related to osteogenesis/osteogenic differentiation. RNA sequencing data from GSCs reseeded on dECM_DPSCs revealed that dECM_DPSCs upregulated genes related to the Hippo and Wnt signaling pathways in GSCs. In the inhibitor experiments, results revealed that dECM_DPSCs superiorly promoted GSCs osteogenic differentiation, mainly mediated through Hippo and Wnt signaling. The present study emphasizes the promising translational application of dECM_DPSCs as a bio-scaffold rich in favorable regenerative microenvironment for tissue engineering.


Subject(s)
Osteogenesis , Wnt Signaling Pathway , Humans , Osteogenesis/genetics , Proteomics , Dental Pulp , Extracellular Matrix/metabolism , Cell Differentiation , Stem Cells/metabolism , Cell Proliferation , Cells, Cultured
2.
Oral Dis ; 2023 Jul 19.
Article in English | MEDLINE | ID: mdl-37466141

ABSTRACT

OBJECTIVE: Periostin (PN), a major matricellular periodontal ligament (PDL) protein, modulates the remodeling of the PDL and bone, especially under mechanical stress. This study investigated the requirement of PN-integrin signaling in force-induced expression of transforming growth factor-beta 1 (TGF-ß1) and alpha-smooth muscle actin (α-SMA) in human PDL stem cells (hPDLSCs). METHODS: Cells were stimulated with intermittent compressive force (ICF) using computerized controlled apparatus. Cell migration was examined using in vitro scratch assay. The mRNA expression was examined using real-time polymerase chain reaction. The protein expression was determined using immunofluorescent staining and western blot analysis. RESULTS: Stimulation with ICF for 24 h increased the expression of PN, TGF-ß1, and α-SMA, along with increased SMAD2/3 phosphorylation. Knockdown of POSTN (PN gene) decreased the protein levels of TGF-ß1 and pSMAD2/3 upon force stimulation. POSTN knockdown of hPDLSCs resulted in delayed cell migration, as determined by a scratch assay. However, migration improved after seeding these knockdown cells on pre-PN-coated surfaces. Further, the knockdown of αVß5 significantly attenuated the force-induced TGF-ß1 expression. CONCLUSION: Our findings indicate the importance of PN-αVß5 interactions in ICF-induced TGF-ß1 signaling and the expression of α-SMA. Findings support the critical role of PN in maintaining the PDL's tissue integrity and homeostasis.

3.
BDJ Open ; 9(1): 31, 2023 Jul 18.
Article in English | MEDLINE | ID: mdl-37463885

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the effect of mechanical force on possible dynamic changes of the matrix proteins deposition in the PDL upon in vitro mechanical and in vivo occlusal forces in a rat model with hypofunctional conditions. MATERIALS AND METHODS: Intermittent compressive force (ICF) and shear force (SF) were applied to human periodontal ligament stem cells (PDLSCs). Protein expression of collagen I and POSTN was analyzed by western blot technique. To establish an in vivo model, rat maxillary molars were extracted to facilitate hypofunction of the periodontal ligament (PDL) tissue of the opposing mandibular molar. The mandibles were collected after 4-, 8-, and 12-weeks post-extraction and used for micro-CT and immunohistochemical analysis. RESULTS: ICF and SF increased the synthesis of POSTN by human PDLSCs. Histological changes in the hypofunctional teeth revealed a narrowing of the PDL space, along with a decreased amount of collagen I, POSTN, and laminin in perivascular structures compared to the functional contralateral molars. CONCLUSION: Our results revealed that loss of occlusal force disrupts deposition of some major matrix proteins in the PDL, underscoring the relevance of mechanical forces in maintaining periodontal tissue homeostasis by modulating ECM composition.

4.
Heliyon ; 8(10): e10845, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36247165

ABSTRACT

Intermittent compressive force influences human periodontal ligament (PDL) cell behavior that facilitates periodontal tissue regeneration. In response to mechanical stimuli, Yes-associated protein (YAP) has been recognized as a mechanosensitive transcriptional activator that regulates cell proliferation and cell fate decisions. This study aimed to investigate whether compressive forces influence cell proliferation and cell fate decisions of human PDL cells via YAP signaling. YAP expression was silenced by shRNA. The effect of YAP on cell proliferation, adipogenesis and osteogenesis of PDL cells under ICF loading were determined. Adipogenic differentiation bias upon ICF loading was confirmed by fourier-transform infrared spectroscopy (FTIR). The results revealed that ICF-induced YAP promotes osteogenesis, but it inhibits adipogenesis in PDL cells. Depletion of YAP results in PDL cells that are irresponsive to ICF and, therefore, the failure of the PDL cells to undergo osteogenic differentiation. This was shown by a significant reduction in calcium deposited in the CF-derived osteoblasts of the YAP-knockdown (YAP-KD) PDL cells. As to control treatment, reduction of YAP promoted adipogenesis, whereas ICF-induced YAP inhibited this mechanism. However, the adipocyte differentiation in YAP-KD cells was not affected upon ICF treatment as the YAP-KD cells still exhibited a better adipogenic differentiation that was unrelated to the ICF. This study demonstrated that, in response to ICF treatment, YAP could be a crucial mechanosensitive transcriptional activator for the regulation of PDL cell behavior through a mechanobiological process. Our results may provide the possibility of facilitating PDL tissue regeneration by manipulation of the Hippo-YAP signaling pathway.

5.
Int J Mol Sci ; 23(13)2022 Jun 27.
Article in English | MEDLINE | ID: mdl-35806124

ABSTRACT

Relevant immunomodulatory effects have been proposed following allogeneic cell-based therapy with human periodontal ligament stem cells (hPDLSCs). This study aimed to examine the influence of shear stress on the immunosuppressive capacity of hPDLSCs. Cells were subjected to shear stress at different magnitudes (0.5, 5 and 10 dyn/cm2). The expression of immunosuppressive markers was evaluated in shear stress-induced hPDLSCs using qRT-PCR, western blot, enzyme activity and enzyme-linked immunosorbent assays. The effects of a shear stress-derived condition medium (SS-CM) on T cell proliferation were examined using a resazurin assay. Treg differentiation was investigated using qRT-PCR and flow cytometry analysis. Our results revealed that shear stress increased mRNA expression of IDO and COX2 but not TGF-ß1 and IFN-γ. IDO activity, kynurenine and active TGF-ß1 increased in SS-CM when compared to the non-shear stress-derived conditioned medium (CTL-CM). The amount of kynurenine in SS-CM was reduced in the presence of cycloheximide and ERK inhibitor. Subsequently, T cell proliferation decreased in SS-CM compared to CTL-CM. Treg differentiation was promoted in SS-CM, indicated by FOXP3, IL-10 expression and CD4+CD25hiCD127lo/- subpopulation. In conclusion, shear stress promotes kynurenine production through ERK signalling in hPDLSC, leading to the inhibition of T cell proliferation and the promotion of Treg cell differentiation.


Subject(s)
Kynurenine , Periodontal Ligament , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Kynurenine/metabolism , Osteogenesis , Stem Cells/metabolism
6.
Biomolecules ; 9(11)2019 10 25.
Article in English | MEDLINE | ID: mdl-31717706

ABSTRACT

Research efforts have been made to develop human salivary gland (SG) secretory epithelia for transplantation in patients with SG hypofunction and dry mouth (xerostomia). However, the limited availability of human biopsies hinders the generation of sufficient cell numbers for epithelia formation and regeneration. Porcine SG have several similarities to their human counterparts, hence could replace human cells in SG modelling studies in vitro. Our study aims to establish porcine SG explant outgrowth models to generate functional secretory epithelia for regeneration purposes to rescue hyposalivation. Cells were isolated and expanded from porcine submandibular and parotid gland explants. Flow cytometry, immunocytochemistry, and gene arrays were performed to assess proliferation, standard mesenchymal stem cell, and putative SG epithelial stem/progenitor cell markers. Epithelial differentiation was induced and different SG-specific markers investigated. Functional assays upon neurostimulation determined α-amylase activity, trans-epithelial electrical resistance, and calcium influx. Primary cells exhibited SG epithelial progenitors and proliferation markers. After differentiation, SG markers were abundantly expressed resembling epithelial lineages (E-cadherin, Krt5, Krt14), and myoepithelial (α-smooth muscle actin) and neuronal (ß3-tubulin, Chrm3) compartments. Differentiated cells from submandibular gland explant models displayed significantly greater proliferation, number of epithelial progenitors, amylase activity, and epithelial barrier function when compared to parotid gland models. Intracellular calcium was mobilized upon cholinergic and adrenergic neurostimulation. In summary, this study highlights new strategies to develop secretory epithelia from porcine SG explants, suitable for future proof-of-concept SG regeneration studies, as well as for testing novel muscarinic agonists and other biomolecules for dry mouth.


Subject(s)
Epithelium , Salivary Glands , Tissue Engineering , Animals , Cell Differentiation , Swine
7.
Stem Cell Res Ther ; 9(1): 46, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29482624

ABSTRACT

BACKGROUND: Thalassemia is the most common genetic disease worldwide; those with severe disease require lifelong blood transfusion and iron chelation therapy. The definitive cure for thalassemia is allogeneic hematopoietic stem cell transplantation, which is limited due to lack of HLA-matched donors and the risk of post-transplant complications. Induced pluripotent stem cell (iPSC) technology offers prospects for autologous cell-based therapy which could avoid the immunological problems. We now report genetic correction of the beta hemoglobin (HBB) gene in iPSCs derived from a patient with a double heterozygote for hemoglobin E and ß-thalassemia (HbE/ß-thalassemia), the most common thalassemia syndrome in Thailand and Southeast Asia. METHODS: We used the CRISPR/Cas9 system to target the hemoglobin E mutation from one allele of the HBB gene by homology-directed repair with a single-stranded DNA oligonucleotide template. DNA sequences of the corrected iPSCs were validated by Sanger sequencing. The corrected clones were differentiated into hematopoietic progenitor and erythroid cells to confirm their multilineage differentiation potential and hemoglobin expression. RESULTS: The hemoglobin E mutation of HbE/ß-thalassemia iPSCs was seamlessly corrected by the CRISPR/Cas9 system. The corrected clones were differentiated into hematopoietic progenitor cells under feeder-free and OP9 coculture systems. These progenitor cells were further expanded in erythroid liquid culture system and developed into erythroid cells that expressed mature HBB gene and HBB protein. CONCLUSIONS: Our study provides a strategy to correct hemoglobin E mutation in one step and these corrected iPSCs can be differentiated into hematopoietic stem cells to be used for autologous transplantation in patients with HbE/ß-thalassemia in the future.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Hemoglobin E , Induced Pluripotent Stem Cells/metabolism , beta-Thalassemia , Autografts , Female , Hemoglobin E/genetics , Hemoglobin E/metabolism , Humans , Male , Mutation , Stem Cell Transplantation , beta-Thalassemia/genetics , beta-Thalassemia/metabolism , beta-Thalassemia/therapy
8.
Ann Hematol ; 95(10): 1617-25, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27465155

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is an acquired hemolytic anemia caused by lack of CD55 and CD59 on blood cell membrane leading to increased sensitivity of blood cells to complement. Hematopoietic stem cell transplantation (HSCT) is the only curative therapy for PNH, however, lack of HLA-matched donors and post-transplant complications are major concerns. Induced pluripotent stem cells (iPSCs) derived from patients are an attractive source for generating autologous HSCs to avoid adverse effects resulting from allogeneic HSCT. The disease involves only HSCs and their progeny; therefore, other tissues are not affected by the mutation and may be used to produce disease-free autologous HSCs. This study aimed to derive PNH patient-specific iPSCs from human dermal fibroblasts (HDFs), characterize and differentiate to hematopoietic cells using a feeder-free protocol. Analysis of CD55 and CD59 expression was performed before and after reprogramming, and hematopoietic differentiation. Patients' dermal fibroblasts expressed CD55 and CD59 at normal levels and the normal expression remained after reprogramming. The iPSCs derived from PNH patients had typical pluripotent properties and differentiation capacities with normal karyotype. After hematopoietic differentiation, the differentiated cells expressed early hematopoietic markers (CD34 and CD43) with normal CD59 expression. The iPSCs derived from HDFs of PNH patients have normal levels of CD55 and CD59 expression and hold promise as a potential source of HSCs for autologous transplantation to cure PNH patients.


Subject(s)
Cellular Reprogramming Techniques , Hematopoietic Stem Cell Transplantation , Hemoglobinuria, Paroxysmal/therapy , Induced Pluripotent Stem Cells/cytology , Animals , Antigens, CD34/biosynthesis , CD55 Antigens/biosynthesis , CD59 Antigens/biosynthesis , Embryoid Bodies , Female , Fibroblasts/cytology , History, 16th Century , History, 17th Century , Humans , Induced Pluripotent Stem Cells/transplantation , Leukosialin/biosynthesis , Mice, Inbred BALB C , Mice, Nude , Skin/cytology , Teratoma/pathology , Transplantation, Autologous
9.
Thromb Haemost ; 114(3): 593-602, 2015 Aug 31.
Article in English | MEDLINE | ID: mdl-26063314

ABSTRACT

Platelet transfusion has been widely used to prevent and treat life-threatening thrombocytopenia; however, preparation of a unit of concentrated platelet for transfusion requires at least 4-6 units of whole blood. At present, a platelet unit from a single donor can be prepared using apheresis, but lack of donors is still a major problem. Several approaches to produce platelets from other sources, such as haematopoietic stem cells and pluripotent stem cells, have been attempted but the system is extremely complicated, time-consuming and expensive. We now report a novel and simpler technology to obtain platelets using transdifferentiation of human bone marrow erythroblasts to megakaryocytes with overexpression of the FLI1 and ERG genes. The obtained transdifferentiated erythroblasts (both from CD71+ and GPA+ erythroblast subpopulations) exhibit typical features of megakaryocytes including morphology, expression of specific genes (cMPL and TUBB1) and a marker protein (CD41). They also have the ability to generate megakaryocytic CFU in culture and produce functional platelets, which aggregate with normal human platelets to form a normal-looking clot. Overexpression of FLI1 and ERG genes is sufficient to transdifferentiate erythroblasts to megakaryocytes that can produce functional platelets.


Subject(s)
Cell Transdifferentiation , Erythroblasts/metabolism , Megakaryocytes/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Trans-Activators/metabolism , Biomarkers/metabolism , Blood Coagulation , Cell Proliferation , Cell Shape , Cells, Cultured , Genotype , Humans , Phenotype , Platelet Aggregation , Platelet Membrane Glycoprotein IIb/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Receptors, Thrombopoietin/metabolism , Signal Transduction , Time Factors , Trans-Activators/genetics , Transcriptional Regulator ERG , Transfection , Tubulin/metabolism , Up-Regulation
10.
Ann Hematol ; 94(2): 319-27, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25245632

ABSTRACT

Human mesenchymal stem cells (hMSCs) have been used to improve engraftment and to treat graft versus host disease following allogeneic hematopoietic stem cell transplantation. However, oxidative stress presented in the microenvironment can damage the transplanted hMSCs and therefore reduce their survival in target organs. We investigated how to enhance the survival of hMSCs under oxidative stress by overexpressing secreted frizzled-related protein 2 (sFRP2) gene in bone marrow-derived hMSCs and umbilical cord-derived hMSCs. The survival and characteristics of those sFRP2-overexpressing hMSCs (sFRP2-BM-hMSCs and sFRP2-UC-hMSCs) were studied compared with non-transduced hMSCs. We found that the percentages of viable cells in culture of sFRP2-BM-hMSCs and sFRP2-UC-hMSCs in the absence or presence of 0.75 mM H2O2 were significantly higher than those of their non-transduced counterparts. The overexpression of sFRP2 gene did not affect the characteristics of hMSCs regarding their morphology, surface marker expression, and differentiation potential. Our study suggests that overexpression of sFRP2 gene in hMSCs might improve the therapeutic effectiveness of hMSC transplantation.


Subject(s)
Gene Expression , Membrane Proteins/genetics , Mesenchymal Stem Cells/metabolism , Oxidative Stress , Amnion/cytology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Female , Fetal Blood/cytology , Flow Cytometry , Humans , Hydrogen Peroxide/pharmacology , Membrane Proteins/metabolism , Oxidants/pharmacology , Placenta/cytology , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Transfection
11.
PLoS One ; 9(9): e106952, 2014.
Article in English | MEDLINE | ID: mdl-25207966

ABSTRACT

Incurable neurological disorders such as Parkinson's disease (PD), Huntington's disease (HD), and Alzheimer's disease (AD) are very common and can be life-threatening because of their progressive disease symptoms with limited treatment options. To provide an alternative renewable cell source for cell-based transplantation and as study models for neurological diseases, we generated induced pluripotent stem cells (iPSCs) from human dermal fibroblasts (HDFs) and then differentiated them into neural progenitor cells (NPCs) and mature neurons by dual SMAD signaling inhibitors. Reprogramming efficiency was improved by supplementing the histone deacethylase inhibitor, valproic acid (VPA), and inhibitor of p160-Rho associated coiled-coil kinase (ROCK), Y-27632, after retroviral transduction. We obtained a number of iPS colonies that shared similar characteristics with human embryonic stem cells in terms of their morphology, cell surface antigens, pluripotency-associated gene and protein expressions as well as their in vitro and in vivo differentiation potentials. After treatment with Noggin and SB431542, inhibitors of the SMAD signaling pathway, HDF-iPSCs demonstrated rapid and efficient differentiation into neural lineages. Six days after neural induction, neuroepithelial cells (NEPCs) were observed in the adherent monolayer culture, which had the ability to differentiate further into NPCs and neurons, as characterized by their morphology and the expression of neuron-specific transcripts and proteins. We propose that our study may be applied to generate neurological disease patient-specific iPSCs allowing better understanding of disease pathogenesis and drug sensitivity assays.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/cytology , Smad Proteins/metabolism , Amides/pharmacology , Benzamides/pharmacology , Biomarkers/metabolism , Carrier Proteins/pharmacology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/drug effects , Dermis/cytology , Dermis/drug effects , Dermis/metabolism , Dioxoles/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Karyotyping , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Pyridines/pharmacology , Signal Transduction , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics , Valproic Acid/pharmacology , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
12.
Neurosci Lett ; 578: 1-6, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24923762

ABSTRACT

The transforming growth factor-ß1 (TGF-ß1), a polypeptide member of the TGF-ß superfamily, has myriad cellular functions, including cell fate differentiation. We hypothesized that suppression of TGF-ß1 signaling would improve the efficacy of neuronal differentiation during embryoid body (EB) development. In this study, mouse embryonic stem cells (ESCs) were allowed to differentiate into their neuronal lineage, both with, and without the TGF-ß1 inhibitor (A83-01). After 8 days of EB suspension culture, the samples were examined by morphological analysis, immunocytochemistry and immunohistochemistry with pluripotent (Oct4, Sox2) and neuronal specific markers (Pax6, NeuN). The alteration of gene expressions during EB development was determined by quantitative RT-PCR. Our results revealed that the TGF-ß1/ALK inhibitor potentially suppressed pluripotent gene (Oct4) during a rapidly up-regulation of neuronal associated genes including Sox1 and MAP2. Strikingly, during EB development, the expression of GFAP, the astrocyte specific gene, remarkably decreased compared to the non-treated control. This strategy demonstrated the beneficial function of TGF-ß1/ALK inhibitor that rapidly and uniformly drives cell fate alteration from pluripotent state toward neuronal lineages.


Subject(s)
Embryonic Stem Cells/drug effects , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Pyrazoles/pharmacology , Thiocarbamates/pharmacology , Transforming Growth Factor beta1/antagonists & inhibitors , Activin Receptors, Type I/antagonists & inhibitors , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Embryoid Bodies , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/physiology , Mice , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Neuroglia/metabolism , Neuroglia/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction , Thiosemicarbazones , Transforming Growth Factor beta1/metabolism
13.
Biochem Biophys Res Commun ; 447(4): 580-5, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24747566

ABSTRACT

Bortezomib (BZB) is a chemotherapeutic agent approved for treating multiple myeloma (MM) patients. In addition, there are several reports showing that bortezomib can induce murine mesenchymal stem cells (MSCs) to undergo osteogenic differentiation and increase bone formation in vivo. MSCs are the multipotent stem cells that have capacity to differentiate into several mesodermal derivatives including osteoblasts. Nowadays, MSCs mostly bone marrow derived have been considered as a valuable source of cell for tissue replacement therapy. In this study, the effect of bortezomib on the osteogenic differentiation of human MSCs derived from both bone marrow (BM-MSCs) and postnatal sources such as placenta (PL-MSCs) were investigated. The degree of osteogenic differentiation of BM-MSCs and PL-MSCs after bortezomib treatment was assessed by alkaline phosphatase (ALP) activity, matrix mineralization by Alizarin Red S staining and the expression profiles of osteogenic differentiation marker genes, Osterix, RUNX2 and BSP. The results showed that 1 nM and 2 nM BZB can induce osteogenic differentiation of BM-MSCs and PL-MSCs as demonstrated by increased ALP activity, increased matrix mineralization and up-regulation of osteogenic differentiation marker genes, Osterix, RUNX2 and BSP as compared to controls. The enhancement of osteogenic differentiation of MSCs by bortezomib may lead to the potential therapeutic applications in human diseases especially patients with osteopenia.


Subject(s)
Boronic Acids/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Placenta/cytology , Pyrazines/pharmacology , Adipogenesis/drug effects , Alkaline Phosphatase/metabolism , Bone Diseases, Metabolic/drug therapy , Bortezomib , Cell Differentiation/drug effects , Cell Differentiation/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Female , Humans , Integrin-Binding Sialoprotein/genetics , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Osteoporosis/drug therapy , Pregnancy , Sp7 Transcription Factor , Transcription Factors/genetics , Up-Regulation/drug effects
14.
Tissue Eng Part C Methods ; 20(5): 383-92, 2014 May.
Article in English | MEDLINE | ID: mdl-24006854

ABSTRACT

Somatic cell reprogramming has generated enormous interest after the first report by Yamanaka and his coworkers in 2006 on the generation of induced pluripotent stem cells (iPSCs) from mouse fibroblasts. Here we report the generation of stable iPSCs from mouse fibroblasts by recombinant protein transduction (Klf4, Oct4, Sox2, and c-Myc), a procedure designed to circumvent the risks caused by integration of exogenous sequences in the target cell genome associated with gene delivery systems. The recombinant proteins were fused in the frame to the glutathione-S-transferase tag for affinity purification and to the transactivator transcription-nuclear localization signal polypeptide to facilitate membrane penetration and nuclear localization. We performed the reprogramming procedure on embryonic fibroblasts from inbred (C57BL6) and outbred (ICR) mouse strains. The cells were treated with purified proteins four times, at 48-h intervals, and cultured on mitomycin C treated mouse embryonic fibroblast (MEF) cells in complete embryonic stem cell (ESC) medium until colonies formed. The iPSCs generated from the outbred fibroblasts exhibited similar morphology and growth properties to ESCs and were sustained in an undifferentiated state for more than 20 passages. The cells were checked for pluripotency-related markers (Oct4, Sox2, Klf4, cMyc, Nanog) by immunocytochemistry and by reverse transcription-polymerase chain reaction. The protein iPSCs (piPSCs) formed embryoid bodies and subsequently differentiated towards all three germ layer lineages. Importantly, the piPSCs could incorporate into the blastocyst and led to variable degrees of chimerism in newborn mice. These data show that recombinant purified cell-penetrating proteins are capable of reprogramming MEFs to iPSCs. We also demonstrated that the cells of the generated cell line satisfied all the requirements of bona fide mouse ESCs: form round colonies with defined boundaries; have a tendency to attach together with high nuclear/cytoplasmic ratio; express key pluripotency markers; and are capable of in vitro differentiation into ecto-, endo-, and mesoderm, and in vivo chimera formation.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Recombinant Proteins/metabolism , Transduction, Genetic/methods , Animals , Cell Differentiation/genetics , Chimera/metabolism , Gene Expression Regulation , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Mice , Recombinant Proteins/isolation & purification , Reproducibility of Results , Transcription Factors/metabolism
15.
Cell Reprogram ; 15(5): 443-58, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24020697

ABSTRACT

Embryonic stem cells (ESCs) have the ability to form aggregates, which are called embryoid bodies (EBs). EBs mimic early embryonic development and are commonly produced for cardiomyogenesis. Here, we describe a method of EB formation in hydrodynamic conditions using a slow-turning lateral vessel (STLV) bioreactor and the subsequent differentiation of EBs into cardiomyocytes. EBs formed in the STLV were compared with conventional techniques, such as hanging drop (HD) or static suspension cell culture (SSC), for homogeneity of EB size, shape, proliferation, apoptosis, and in vitro cardiac differentiation. After 3 days of culture, a four-fold improvement in the yield of EB formation/mL, a six-fold enhancement in total yield of EB/mL, and a nearly 10-fold reduction of cells that failed to incorporate into EBs were achieved in STLV versus SSC. During cardiac differentiation, a 1.5- to 4.2-fold increase in the area of cardiac troponin T (cTnT) per single EB in STLV versus SSC and HD was achieved. These results demonstrate that the STLV method improves the quality and quantity of ES cells to form EBs and enhances the efficiency of cardiac differentiation. We have demonstrated that the mechanical method of cell differentiation creates different microenvironments for the cells and thus influences their lineage commitments, even when genetic origin and the culture medium are the same. Ascorbic acid (ASC) improved further cardiac commitment in differentiation assays. Hence, this culture system is suitable for the production of large numbers of cells for clinical cell replacement therapies and industrial drug testing applications.


Subject(s)
Bioreactors , Cell Differentiation , Embryoid Bodies/cytology , Embryonic Stem Cells/cytology , Heart/embryology , Animals , Base Sequence , Cells, Cultured , DNA Primers , Gene Expression Regulation, Developmental , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
16.
Cell Reprogram ; 14(5): 390-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22917491

ABSTRACT

Mouse embryonic stem cells (ESCs) and induced pluripotent stem (iPS) cells can be used as models of neuronal differentiation for the investigation of mammalian neurogenesis, pharmacological testing, and development of cell-based therapies. Recently, mouse iPS cell lines have been generated by Sleeping Beauty (SB) transposon-mediated transgenesis (SB-iPS). In this study, we determined for the first time the differentiation potential of mouse SB-iPS cells to form neuronal progenitor cells (NPCs) and neurons. Undifferentiated SB-iPS and ES cells were aggregated into embryoid bodies (EBs) and cultured in neuronal differentiation medium supplemented with 5 µM all-trans retinoic acid. Thereafter, EBs were dissociated and plated to observe further neuronal differentiation. Samples were fixed on days 10 and 14 for immunocytochemistry staining using the NPC markers Pax6 and Nestin and the neuron marker ßIII-tubulin/Tuj1. Nestin-labeled cells were analyzed further by flow cytometry. Our results demonstrated that SB-iPS cells can generate NPCs and differentiate further into neurons in culture, although SB-iPS cells produced less nestin-positive cells than ESCs (6.12 ± 1.61 vs. 74.36 ± 1.65, respectively). In conclusion, the efficiency of generating SB-iPS cells-derived NPCs needs to be improved. However, given the considerable potential of SB-iPS cells for drug testing and as therapeutic models in neurological disorders, continuing investigation of their neuronal differentiation ability is required.


Subject(s)
DNA Transposable Elements , Neurons/cytology , Pluripotent Stem Cells/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Flow Cytometry , Immunohistochemistry , Mice , Neurogenesis
17.
Biotechnol Lett ; 33(8): 1565-73, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21476093

ABSTRACT

Embryoid body (EB) formation is a common intermediate during in vitro differentiation of pluripotent stem cells into specialized cell types. We have optimized the slow-turning, lateral vessel (STLV) for large scale and homogenous EB production from mouse embryonic stem cells. The effects of inoculating different cell numbers, time of EB adherence to gelatin-coated dishes, and rotation speed for optimal EB formation and cardiac differentiation were investigated. Using 3 × 10(5) cells/ml, 10 rpm rotary speed and plating of EBs onto gelatin-coated surfaces three days after culture, were the best parameters for optimal size and EB quality on consequent cardiac differentiation. These optimized parameters enrich cardiac differentiation in ES cells when using the STLV method.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Cell Adhesion/physiology , Cell Culture Techniques/methods , Cell Differentiation/genetics , Cell Size , Embryoid Bodies/cytology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Genetic Markers , Mice , Myocytes, Cardiac/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...