Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nanomaterials (Basel) ; 14(3)2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38334522

ABSTRACT

The progress in artificial bone research is crucial for addressing fractures and bone defects in the aging population. However, challenges persist in terms of biocompatibility and structural complexity. Nanotechnology provides a promising avenue by which to overcome these challenges, with nano-ferrite particles (NFPs) exhibiting superparamagnetic properties. The ability to control cell positioning using a magnetic field opens up new possibilities for customizing artificial bones with specific shapes. This study explores the biological effects of NFPs on osteoblast-like cell lines (MC3T3-E1), including key analyses, such as cell viability, cellular uptake of NFPs, calcification processes, cell migration under external magnetic field conditions, and three-dimensional modeling. The results indicate that the impact of NFPs on cell proliferation is negligible. Fluorescence and transmission electron microscopy validated the cellular uptake of NFPs, demonstrating the potential for precise cell positioning through an external magnetic field. Under calcification-inducing conditions, the cells exhibited sustained calcification ability even in the presence of NFPs. The cell movement analysis observed the controlled movement of NFP-absorbing cells under an external magnetic field. Applying a magnetic field along the z-axis induced the three-dimensional shaping of cells incorporating NFPs, resulting in well-arranged z-axis directional patterns. In this study, NFPs demonstrated excellent biocompatibility and controllability under an external magnetic field, laying the foundation for innovative treatment strategies for customizing artificial bones.

2.
Genes (Basel) ; 14(3)2023 03 16.
Article in English | MEDLINE | ID: mdl-36981001

ABSTRACT

Musculocontractural Ehlers-Danlos syndrome caused by mutations in the carbohydrate sulfotransferase 14 gene (mcEDS-CHST14) is a heritable connective tissue disorder characterized by multiple congenital malformations and progressive connective tissue fragility-related manifestations in the cutaneous, skeletal, cardiovascular, visceral, and ocular systems. Progressive skeletal deformities are among the most frequent and serious complications affecting the quality of life and activities of daily living in patients. After establishing induced pluripotent stem cells (iPSCs) from cultured skin fibroblasts of three patients with mcEDS-CHST14, we generated a patient iPSC-based human osteogenesis model and performed an in vitro assessment of the phenotype and pathophysiology of skeletal deformities. Patient-derived iPSCs presented with remarkable downregulation of osteogenic-specific gene expression, less alizarin red staining, and reduced calcium deposition compared with wild-type iPSCs at each stage of osteogenic differentiation, including osteoprogenitor cells, osteoblasts, and osteocytes. These findings indicated that osteogenesis was impaired in mcEDS-CHST14 iPSCs. Moreover, the decrease in decorin (DCN) expression and increase in collagen (COL12A1) expression in patient-derived iPSCs elucidated the contribution of CHST14 dysfunction to skeletal deformities in mcEDS-CHST14. In conclusion, this disease-in-a-dish model provides new insight into the pathophysiology of EDS and may have the potential for personalized gene or drug therapy.


Subject(s)
Ehlers-Danlos Syndrome , Induced Pluripotent Stem Cells , Humans , Induced Pluripotent Stem Cells/metabolism , Activities of Daily Living , Osteogenesis/genetics , Quality of Life , Sulfotransferases/genetics , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/metabolism
3.
Stem Cell Res ; 64: 102907, 2022 10.
Article in English | MEDLINE | ID: mdl-36099765

ABSTRACT

Peripheral blood mononuclear cells (PBMCs) of a 38-year-old healthy female were isolated and reprogrammed into the induced pluripotent stem cells (iPSCs). The established iPSC line expressed various pluripotency stem cell markers and potential of differentiating into three germ layers, meanwhile maintained normal karyotype.


Subject(s)
Induced Pluripotent Stem Cells , Female , Humans , Adult , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear/metabolism , Germ Layers/metabolism , Biomarkers/metabolism , Cell Differentiation
4.
Stem Cell Res ; 64: 102908, 2022 10.
Article in English | MEDLINE | ID: mdl-36113356

ABSTRACT

Peripheral blood mononuclear cells derived from a 35-year-old healthy male were reprogrammed into induced pluripotent stem cells (iPSCs). The iPSCs maintained a normal karyotype, expressed various pluripotency stem cell markers, and showed potential of differentiating into three germ layers. This iPSCs could be differentiated into multiple cell subtypes for drug discovery and investigation of mechanisms.


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Male , Humans , Adult , Induced Pluripotent Stem Cells/metabolism , Leukocytes, Mononuclear , Cell Differentiation , Germ Layers
5.
Hum Cell ; 35(5): 1475-1486, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35792978

ABSTRACT

Cancer stem cells (CSCs) are involved in cancer recurrence and metastasis owing to their self-renewal properties and drug-resistance capacity. Lipocalin-2 (Lcn2) of the lipocalin superfamily is highly expressed in pancreatic cancer. Nevertheless, reports on the involvement of Lcn2 in the regulation of pancreatic CSC properties are scant. This study is purposed to investigate whether Lcn2 plays a crucial role in CSC renewal and stemness maintenance in pancreatic carcinoma. Immunohistochemistry results of tumor tissue chips together with Gene Expression Omnibus sequencing files confirmed that Lcn2 is highly expressed in pancreatic carcinoma compared with that in normal tissues. The exogenous expression of Lcn2 attenuated CSC-associated SOX2, CD44, and EpCAM expression and suppressed sarcosphere formation and tumorigenesis in the pancreatic carcinoma cell line PANC-1, which showed low expression of Lcn2. However, Lcn2 knockout in BxPC-3 cell line, which presented high Lcn2 expression, promoted CSC stemness, further enhancing sarcosphere formation and tumorigenesis. Moreover, Lcn2 was found to regulate stemness in pancreatic cancer depending on the activation of AKT and c-Jun. Lcn2 suppresses stemness properties in pancreatic carcinoma by activating the AKT-c-Jun pathway, and thus, it may be a novel candidate to suppress the stemness of pancreatic cancer. This study provides a new insight into disease progression.


Subject(s)
Pancreatic Neoplasms , Proto-Oncogene Proteins c-akt , Carcinogenesis/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Lipocalin-2/genetics , Lipocalin-2/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun , Pancreatic Neoplasms
6.
Life Sci ; 262: 118492, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32980390

ABSTRACT

Lipocalin2 (Lcn2) has been shown to be a vital regulator of tumorigenesis in a variety of different cancers. However, its expression patterns and possible roles in ovarian cancer remain obscure. The aim of this study was to investigate the expression of Lcn2 in ovarian cancer cells and to determine any potential association between Lcn2 and ovarian tumor development and cancer progression. Our results indicated that Lcn2 was upregulated in tumor tissue from ovarian cancer patients as well as in three ovarian cancer cell lines compared to normal tissues and cells. Overexpression of Lcn2 promoted both cell proliferation and migration in ovarian cancer cells. Conversely, knockdown of Lcn2 in cell lines suppressed both migration and proliferation. Moreover, upregulation of Lcn2 contributed to tumor growth in nude mice in vivo. Mechanistically, Lcn2 was found to lead to tumor progression in ovarian cancer cells through activation of the ERK/GSK3ß/ß-catenin signaling pathway. In summary, Lcn2 promotes cell proliferation and migration in ovarian cancer through activation of the ERK/GSK3ß/ß-catenin signaling pathway, suggesting that Lcn2 might be a novel therapeutic target for ovarian cancer.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Lipocalin-2/genetics , Ovarian Neoplasms/pathology , Animals , Cell Line, Tumor , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glycogen Synthase Kinase 3 beta/metabolism , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Nude , Ovarian Neoplasms/genetics , Wnt Signaling Pathway/genetics
7.
Life Sci ; 254: 117632, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32437796

ABSTRACT

AIMS: Liver cancer is one of the leading causes of cancer death worldwide owing to its delayed diagnosis and absence of efficient treatment at advanced TNM stages. Increasing evidence demonstrated that microRNAs are implicated in tumorgenesis and cancer development by regulating cancer-related proteins. This study aimed to explore the effect of miR-3619-5p on cell growth in liver cancer. MAIN METHODS: The effect of miR-3619-5p on cell proliferation was measured by quantitative real-time PCR, MTT assay, flow cytometry, and Immunofluorescence assay. The interaction between miR-3619-5p and PSMD10 was validated using dual-luciferase. The expression of PSMD10 and Ki67 was further determined by immunohistochemistry. KEY FINDINGS: MiR-3619-5p over-expression remarkably inhibited cell proliferation and induced G1 phase arrest, accompanied with reduced expression of proliferating cell nuclear antigen. The expression of miR-3619-5p was negatively correlated to that of PSMD10, and PSMD10 was validated to be a downstream target of miR-3619-5p. Moreover, miR-3619-5p induced suppressed proliferation and G1 phase arrest were abrogated by elevated the expression of PSMD10 in liver cancer cells. PSMD10 over-expression also induced phosphorylation of signal transducer and activator of transcription 3 (STAT3) and retinoblastoma protein (Rb1). Besides, elevated cyclin A, cyclin D1 and cyclin E expression supported that PSMD10 promoted the progress of cell cycle. In addition, miR-3619-5p inhibited tumor growth in vivo by targeting PSMD10, accompanied with blocked cell cycle. SIGNIFICANCE: In conclusion, our findings revealed that miR-3619-5p inhibits cancer cell proliferation by targeting PSMD10, and miR-3619-5p as a potential therapeutic target for the treatment of liver cancer.


Subject(s)
Liver Neoplasms/pathology , MicroRNAs/physiology , Proteasome Endopeptidase Complex/physiology , Proto-Oncogene Proteins/physiology , Cell Line, Tumor , Cell Proliferation , Humans , In Vitro Techniques
8.
Br J Haematol ; 187(2): 163-173, 2019 10.
Article in English | MEDLINE | ID: mdl-31222725

ABSTRACT

We established mutated and non-mutated induced pluripotent stem cell (iPSC) clones from a patient with PTPN11 (c.226G>A)-mutated juvenile myelomonocytic leukaemia (JMML). Both types of iPSCs fulfilled the quality criteria. Mutated iPSC colonies generated significantly more CD34+ and CD34+ CD45+ cells compared to non-mutated iPSC colonies in a culture coated with irradiated AGM-S3 cells to which four growth factors were added sequentially or simultaneously. The haematopoietic differentiation potential of non-mutated JMML iPSC colonies was similar to or lower than that of iPSC colonies from a healthy individual. The PTPN11 mutation coexisted with the OSBP2 c.389C>T mutation. Zinc-finger nuclease-mediated homologous recombination revealed that correction of PTPN11 mutation in iPSCs with PTPN11 and OSBP2 mutations resulted in reduced CD34+ cell generation to a level similar to that obtained with JMML iPSC colonies with the wild-type of both genes, and interestingly, to that obtained with normal iPSC colonies. Transduction of the PTPN11 mutation into JMML iPSCs with the wild-type of both genes increased CD34+ cell production to a level comparable to that obtained with JMML iPSC colonies harbouring the two genetic mutations. Thus, PTPN11 mutation may be the most essential abnormality to confer an aberrant haematopoietic differentiation potential in this disorder.


Subject(s)
Cell Differentiation/genetics , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Leukemia, Myelomonocytic, Juvenile , Neoplastic Stem Cells/metabolism , Point Mutation , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Animals , Hematopoietic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/pathology , Male , Mice, SCID , Neoplastic Stem Cells/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism
9.
Cell Tissue Res ; 375(3): 697-707, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30284085

ABSTRACT

Cancer stem cells (CSCs) are specific targets for therapeutic applications, but the rarity of CSCs within tumors makes the isolation of CSCs difficult. To overcome these problems, we generated CSCs in vitro using established reprogramming techniques. We transduced four previously established reprogramming factors, Oct3/4, Sox2, Klf4, and L-myc, into the colon cancer cell lines LoVo and OUMS-23, and investigated the biological characteristics of these lines. Tra-1-60+ cells were obtained from reprogrammed induced pluripotent stem (iPS) cell-like colonies and showed CSC properties, including colony formation, maintenance of colonies by repeated passages, and feeder cell dependency, as well as increased expressions of CSC markers such as CD133 and ALDH1. The CSC-like cells showed increased chemoresistance to 5-fluorouracil and elevated tumorigenicity upon transplantation into kidneys of immune-deficient mice. These tumors shifted to a poorly differentiated stage with many atypical cells, cytoplasmic mucin, and focal papillary components, with demonstrated dedifferentiation. The principal component analysis from DNA microarrays showed that though both cell lines moved to iPS cells after reprogramming, they were not completely identical to iPS cells. Significantly elevated gene expression of Decorin and CD90 was observed in CSC-like cells. Together, these results show that reprogramming of cancer cells produced not pluripotent stem cells but CSC-like cells, and these findings will provide biological information about genuine CSCs and help establish new CSC-targeted therapies.


Subject(s)
Cellular Reprogramming , Colonic Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Animals , Biomarkers, Tumor/metabolism , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Mice, SCID , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Curr Med Chem ; 25(24): 2826-2839, 2018.
Article in English | MEDLINE | ID: mdl-28782468

ABSTRACT

Understanding the biological and molecular processes underlying human pathologies is fundamental in order to develop innovative approaches to treat or prevent them. Among the technologies that could provide innovative disease models, induced pluripotent stem cells (iPSCs) is one of the most promising. Indeed, one application of this technology is patient-specific disease modeling. iPSCs obtained by reprogramming patients' cells collected from accessible tissues, have the unique capability to differentiate, under an adequate stimulus, into any human cell type. In particular, iPSCs technology can be applied to study drug adverse effects, that is a key part of the drug discovery process. Indeed, drug induced adverse effects are among the most common causes that lead to abandon the development of new candidate therapeutic molecules, increasing the cost of drug discovery. An innovative strategy that could be used in drug design to solve drug attrition rate, and to establish innovative pharmacological models, could be the application of iPSCs technology in the early stage of the drug discovery process to model druginduced adverse events. In this review, recently developed disease models based on iPSCs will be discussed, with a particular focus on available models of drugs' adverse effect, in particular hepatic/pancreatic toxicity.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Models, Biological , Cellular Reprogramming , Child , Drug-Related Side Effects and Adverse Reactions/etiology , Granulomatous Disease, Chronic/drug therapy , Granulomatous Disease, Chronic/pathology , Humans , Induced Pluripotent Stem Cells/cytology , Liver/drug effects , Liver/metabolism , Pancreas/drug effects , Pancreas/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/toxicity
11.
Anat Sci Int ; 93(1): 82-87, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27659947

ABSTRACT

The gastric cardia-the small area around the cardiac orifice including the abdominal esophagus-is an important target area for abdominal and thoracic surgeries, especially for laparoscopic procedures. In this study of 28 cadavers, a peritoneal earlobe-like appendage near the angle of His was identified as a useful indicator of the lateral margin of the abdominal esophagus, which is otherwise obscure because the peritoneum continues to the diaphragm without definite demarcation of this margin. This structure, which appears equivalent to the epiploic appendages, was commonly found to be present (in 22/28, 78.6 % of the 28 cadavers) and was 4-21 mm × 6-40 mm × 1-4 mm in size, triangular, round, or leaf-like in shape, contained fat, and was on an imaginary line along which the lesser omentum adheres to the lesser curvature and continues to the diaphragm (18/22, 81.8 %). This indicator is associated with the lesser omentum and is part of the gastrophrenic ligament, and could serve as a useful indicator of the margin of the gastric cardia, thus aiding surgeons performing laparoscopic surgery in this region.


Subject(s)
Cardia/anatomy & histology , Aged , Aged, 80 and over , Diaphragm/anatomy & histology , Esophagus/anatomy & histology , Female , Humans , Laparoscopy , Male , Margins of Excision , Middle Aged , Omentum/anatomy & histology , Peritoneum/anatomy & histology
12.
Biochem Biophys Res Commun ; 482(4): 549-555, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27856246

ABSTRACT

Retinoblastoma is the most common intraocular malignancy in pediatric patients. It develops rapidly in the retina and can be fatal if not treated promptly. It has been proposed that a small population of cancer cells, termed cancer stem cells (CSCs), initiate tumorigenesis from immature tissue stem cells or progenitor cells. Reprogramming technology, which can convert mature cells into pluripotent stem cells (iPS), provides the possibility of transducing malignant cancer cells back to CSCs, a type of early stage of cancer. We herein took advantage of reprogramming technology to induce CSCs from retinoblastoma cancer cells. In the present study, the 4 Yamanaka transcription factors, Oct4, Sox2, Klf4 and c-myc, were transduced into retinoblastoma cells (Rbc51). iPS-like colonies were observed 15 days after transduction and showed significantly enhanced CSC properties. The gene and protein expression levels of pluripotent stem cell markers (Tra-1-60, Oct4, Nanog) and cancer stem cell markers (CD133, CD44) were up-regulated in transduced Rbc51 cells compared to control cells. Moreover, iPS-like CSCs could be sorted using the Magnetic-activated cell sorting (MACS) method. A sphere formation assay demonstrated spheroid formation in transduced Rbc51 cells cultured in serum free media, and these spheroids could be differentiated into Pax6-, Nestin-positive neural progenitors and rhodopsin- and recoverin-positive mature retinal cells. The cell viability after 5-Fu exposure was higher in transduced Rbc51 cells. In conclusion, CSCs were generated from retinoblastoma cancer cells using reprogramming technology. Our novel method can generate CSCs, the study of which can lead to better understanding of cancer-specific initiation, cancer epigenetics, and the overlapping mechanisms of cancer development and pluripotent stem cell behavior.


Subject(s)
Cellular Reprogramming Techniques/methods , Cellular Reprogramming , Induced Pluripotent Stem Cells/cytology , Neoplastic Stem Cells/cytology , Retinal Neoplasms/genetics , Retinoblastoma/genetics , Cell Line, Tumor , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Neoplastic Stem Cells/metabolism , Retina/cytology , Retina/metabolism , Transcription Factors/genetics , Transduction, Genetic
13.
PLoS One ; 11(9): e0162693, 2016.
Article in English | MEDLINE | ID: mdl-27632182

ABSTRACT

Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) are potential resources for the regeneration of defective organs, including the liver. However, some obstacles must be overcome before this becomes reality. Undifferentiated cells that remain following differentiation have teratoma-forming potential. Additionally, practical applications require a large quantity of differentiated cells, so the differentiation process must be economical. Here we describe a DNA microarray-based global analysis of the gene expression profiles of differentiating human pluripotent stem cells. We identified differences and commonalities among six human pluripotent stem cell lines: the hESCs KhES1, KhES2, KhES3, and H1, and the iPSCs 201B7 and 243G1. Embryoid bodies (EBs) formed without requiring supplementation with inducing factors. EBs also expressed some liver-specific metabolic genes including the ammonia-metabolizing enzymes glutamine synthetase and carbamoyl-phosphate synthase 1. Real-time PCR analysis revealed hepatocyte-like differentiation of EBs treated with ammonia in Lanford medium. Analysis of DNA microarray data suggested that hepatocyte-like cells were the most abundant population in ammonia-treated cells. Furthermore, expression levels of undifferentiated pluripotent stem cell markers were drastically reduced, suggesting a reduced teratoma-forming capacity. These results indicate that treatment of EBs with ammonia in Lanford medium may be an effective inducer of hepatic differentiation in absence of expensive inducing factors.


Subject(s)
Ammonia/pharmacology , Hepatocytes/drug effects , Pluripotent Stem Cells/drug effects , Animals , Cell Differentiation , Cell Lineage , Hepatocytes/cytology , Humans , Oligonucleotide Array Sequence Analysis , Pluripotent Stem Cells/cytology
14.
Curr Drug Metab ; 17(1): 91-8, 2015.
Article in English | MEDLINE | ID: mdl-26526832

ABSTRACT

Induced pluripotent stem cells (iPSC) can be produced from adult cells by transfecting them with a definite set of pluripotency-associated genes. Under adequate growth conditions and stimulation iPSC can differentiate to almost every somatic lineage in the body. Patients' derived iPSC are an innovative model to study mechanisms of adverse drug reactions in individual patients and in cell types that cannot be easily obtained from human subjects. Proof-of concept studies with known toxicants have been performed for liver, cardiovascular and central nervous system cells: neurons obtained from iPSC have been used to elucidate the mechanism of chemotherapy-induced peripheral neuropathy by evaluating the effects of neurotoxic drugs such as vincristine. However, no study has been performed yet on pancreatic tissue and drug induced pancreatitis. Thiopurines (azathioprine and mercaptopurine) are immunosuppressive antimetabolite drugs, commonly used to treat Crohn's disease. About 5% of Crohn's disease patients treated with thiopurines develop pancreatitis, a severe idiosyncratic adverse event; these patients have to stop thiopurine administration and may require medical treatment, with significant personal and social costs. Molecular mechanism of thiopurine induced pancreatitis (TIP) is currently unknown and no fully validated biomarker is available to assist clinicians in preventing this adverse event. Hence, in this review we have reflected upon the probable research applications of exocrine pancreatic cells generated from patient specific iPS cells. Such pancreatic cells can provide excellent insights into the molecular mechanism of TIP. In particular three hypotheses on the mechanism of TIP could be explored: drug biotransformation, innate immunity and adaptative immunity.


Subject(s)
Azathioprine/adverse effects , Induced Pluripotent Stem Cells/immunology , Mercaptopurine/adverse effects , Pancreatitis/chemically induced , Pancreatitis/immunology , Adaptive Immunity/drug effects , Adaptive Immunity/physiology , Animals , Humans , Immunosuppressive Agents/adverse effects , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Pancreatitis/metabolism , Predictive Value of Tests , Risk Factors
15.
World J Stem Cells ; 7(5): 883-93, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-26131319

ABSTRACT

AIM: To facilitate close contacts between transplanted cardiomyocytes and host skeletal muscle using cell fusion mediated by hemagglutinating virus of Japan envelope (HVJ-E) and tissue maceration. METHODS: Cardiomyocytes (1.5 × 10(6)) from fetal rats were first cultured. After proliferation, some cells were used for fusion with adult muscle fibers using HVJ-E. Other cells were used to create cardiomyocyte sheets (area: about 3.5 cm(2) including 2.1 × 10(6) cells), which were then treated with Nile blue, separated, and transplanted between the latissimus dorsi and intercostal muscles of adult rats with four combinations of HVJ-E and/or NaOH maceration: G1: HVJ-E(+), NaOH(+), Cardiomyocytes(+); G2: HVJ-E(-), NaOH(+), Cardiomyocytes(+); G3: HVJ-E(+), NaOH(-), Cardiomyocytes(+); G4: HVJ-E(-), NaOH(-), Cardiomyocytes(-). At 1 and 2 wk after transplantation, the four groups were compared by detection of beating domains, motion images using moving target analysis software, action potentials, gene expression of MLC-2v and Mesp1 by reverse transcription-polymerase chain reaction, hematoxylin-eosin staining, and immunostaining for cardiac troponin and skeletal myosin. RESULTS: In vitro cardiomyocytes were fused with skeletal muscle fibers using HVJ-E. Cardiomyocyte sheets remained in the primary transplanted sites for 2 wk. Although beating domains were detected in G1, G2, and G3 rats, G1 rats prevailed in the number, size, motion image amplitudes, and action potential compared with G2 and G3 rats. Close contacts were only found in G1 rats. At 1 wk after transplantation, the cardiomyocyte sheets showed adhesion at various points to the myoblast layer in the latissimus dorsi muscle. At 2 wk after transplantation, close contacts were seen over a broad area. Part of the skeletal muscle sarcoplasma seemed to project into the myocardiocyte plasma and some nuclei appeared to share both sarcoplasmas. CONCLUSION: The present results show that close contacts were acquired and facilitated the beating function, thereby providing a new cellular transplantation method using HVJ-E and NaOH maceration.

16.
Cell Tissue Res ; 354(3): 751-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23996199

ABSTRACT

The major molecular signals of pancreatic exocrine development are largely unknown. We examine the role of fibroblast growth factor 7 (FGF7) in the final induction of pancreatic amylase-containing exocrine cells from induced-pancreatic progenitor cells derived from human embryonic stem (hES) cells. Our protocol consisted in three steps: Step I, differentiation of definitive endoderm (DE) by activin A treatment of hES cell colonies; Step II, differentiation of pancreatic progenitor cells by re-plating of the cells of Step I onto 24-well plates at high density and stimulation with all-trans retinoic acid; Step III, differentiation of pancreatic exocrine cells with a combination of FGF7, glucagon-like peptide 1 and nicotinamide. The expression levels of pancreatic endodermal markers such as Foxa2, Sox17 and gut tube endoderm marker HNF1ß were up-regulated in both Step I and II. Moreover, in Step III, the induced cells expressed pancreatic markers such as amylase, carboxypeptidase A and chymotrypsinogen B, which were similar to those in normal human pancreas. From day 8 in Step III, cells immunohistochemically positive for amylase and for carboxypeptidase A, a pancreatic exocrine cell product, were induced by FGF7. Pancreatic progenitor Pdx1-positive cells were localized in proximity to the amylase-positive cells. In the absence of FGF7, few amylase-positive cells were identified. Thus, our three-step culture protocol for human ES cells effectively induces the differentiation of amylase- and carboxypeptidase-A-containing pancreatic exocrine cells.


Subject(s)
Amylases/biosynthesis , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Fibroblast Growth Factor 7/metabolism , Pancreas, Exocrine/cytology , Pancreas, Exocrine/drug effects , Activins/pharmacology , Cell Communication/drug effects , Cell Communication/physiology , Cell Count , Cell Culture Techniques/methods , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Cytological Techniques , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/pharmacology , Glucagon-Like Peptide 1/pharmacology , Humans , Niacinamide/pharmacology , Pancreas, Exocrine/enzymology , Pancreas, Exocrine/metabolism , Tretinoin/pharmacology
17.
Ann Anat ; 195(1): 50-6, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22727933

ABSTRACT

To investigate the effects of the Rho-dependent protein kinase (ROCK) inhibitor Y-27632 on the kinetics of E-cadherin, F-actin, and Oct3/4 distributions in dissociated human embryonic stem (hES) cells and to analyze their interactions morphologically, Y-27632-treated [R(i) (+)] and untreated [R(i) (-)] cells were immunohistochemically stained for E-cadherin and Oct3/4 within 24h of dissociation and also for F-actin. Furthermore, the gene expression of E-cadherin, Oct3/4, and RhoA was confirmed by quantitative real-time RT-PCR. E-cadherin expression intensified linearly along the membranes of R(i) (+) cells or intercellular junctions in cell clusters. F-actin accumulated along the periphery of cells and expanded in a web-like manner along junctions in cell clusters, and Oct3/4 was restricted to the nucleus within few hours of dissociation. However, R(i) (-) cells exhibited deformation and blebbing and appeared to die over time. E-cadherin exhibited a punctate pattern along the periphery, after which it accumulated on one or both sides of the cytoplasm. Actin filaments were concentrated at the bleb bases. Oct3/4 was detected in the cytoplasm, not in the nucleus the recovery of integrated E-cadherin distribution. Quantitative real-time RT-PCR revealed RhoA upregulation and E-cadherin downregulation at 12h after dissociation. Oct3/4 gene expression was unaffected by ROCK inhibition. These results revealed that the cooperative nature of hES cells is maintained by the E-cadherin-actin cytoskeleton system along with the restricted distribution of Oct3/4 in the nucleus. RhoA activation followed by dissociation disorders this system and accelerates cell death, which is partially suppressed by ROCK inhibition.


Subject(s)
Embryonic Stem Cells/metabolism , Embryonic Stem Cells/ultrastructure , Organic Cation Transport Proteins/metabolism , Actins/metabolism , Amides/pharmacology , Animals , Cadherins/metabolism , Cell Line , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Cytoplasm/metabolism , Cytoplasm/ultrastructure , Enzyme Inhibitors/pharmacology , Humans , Immunohistochemistry , Kinetics , Mice , Pyridines/pharmacology , Real-Time Polymerase Chain Reaction , rho-Associated Kinases/antagonists & inhibitors
18.
Cryobiology ; 64(1): 12-22, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22133891

ABSTRACT

Cryopreservation is an essential technique in basic research and clinical applications of human embryonic stem (hES) cells. Cryopreserved hES cells are fragile and undergo post-thaw apoptosis. We performed gene pathway analysis on cryopreserved and thawed hES cells to examine the effect of Y-27632, a Rho-associated kinase (ROCK) inhibitor, on apoptosis and associated molecular events. Y-27632 was added to the cryopreservation solution and/or the post-thaw medium of two hES cell lines (KhES-1, KhES-3). Post-thaw apoptosis was recorded as a function of time using Giemsa staining and the terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Apoptosis plateaued 12h after the untreated hES cells were thawed. Gene pathway analysis showed the activation of IL-1ß, TGF-ß, and their respective receptors (IL-1R, ACVR1C) in the mitogen-activated protein kinase (MAPK) pathway, which resulted in the upregulation of caspase-8 and -10. Quantitative RT-PCR confirmed the upregulation of IL-1ß, TGF-ß, their respective receptors, and caspase-10 and -3. As these molecules were suppressed by Y-27632, gene pathways involving these molecules probably depend on ROCK activation. The TGF-ß receptor antagonist, SB-431542, and an inhibitor of p38MAPK, SB-203580, did not affect apoptosis. Combining Y-27632 with SB-203580, however, resulted in an increase in the survival rate compared with the control. This suggests that the initiation of apoptosis depends on cytokine interactions and multiple ways exist to reduce post-thaw apoptosis in hES cells. Y-27632 can suppress cytokine interactions and the MAPK pathway, thereby reducing the occurrence of apoptosis, and is an effective cryoprotectant for hES cells.


Subject(s)
Amides/pharmacology , Apoptosis/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/physiology , Pyridines/pharmacology , rho-Associated Kinases/antagonists & inhibitors , Benzamides/pharmacology , Caspase 10/biosynthesis , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Cell Line , Cell Survival/drug effects , Cryopreservation , Cryoprotective Agents/metabolism , Dioxoles/pharmacology , Embryonic Stem Cells/cytology , Humans , Imidazoles/pharmacology , Interleukin-1beta/biosynthesis , Transforming Growth Factor beta/biosynthesis , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Pancreas ; 40(8): 1245-52, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21989025

ABSTRACT

OBJECTIVE: The objective of the study was to induce transdifferentiation of human hepatoma HepG2 cells into pancreatic-like cells without direct genetic intervention. METHODS: HepG2 cells were transfected with plasmids for the hepatocyte marker protein green fluorescent protein (albumin-GFP) and the pancreatic cell marker Discosoma spp red fluorescent protein (elastase-DsRed) to create FAE-HepG2 cells. Fluorescent marker expression was used to monitor in vitro transdifferentiation stimulated 100 mM CCl4, 2 mM D-galactosamine, or 200 µM ZnCl2. Concentrations were selected for optimal cell survival rate. Transdifferentiation was also characterized by immunohistochemical detection of amylase, glucagon, and insulin and by polymerase change reaction analysis of amylase and insulin mRNA production. RESULTS: Control cells expressed albumin-GFP but no elastase-DsRed. By 30 days of culture, all 3 agents induced expression of pancreatic-like cell marker elastase-DsRed. ZnCl2 was the most effective as most cells expressed elastase-DsRed in the absence of simultaneous expression of albumin-GFP. For CCl4 and D-galactosamine, elastase-DsRed was expressed in the same cells as albumin-GFP. Cells treated by each agent also expressed amylase, insulin, and glucagon proteins and mRNAs. CONCLUSIONS: Without direct genetic intervention, select low small molecules can induce in vitro transformation of hepatoma cells into pancreatic-like cells.


Subject(s)
Carbon Tetrachloride/pharmacology , Cell Transdifferentiation/drug effects , Chlorides/pharmacology , Galactosamine/pharmacology , Pancreas/cytology , Zinc Compounds/pharmacology , Albumins/genetics , Albumins/metabolism , Amylases/genetics , Amylases/metabolism , Gene Expression/drug effects , Glucagon/genetics , Glucagon/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hep G2 Cells , Humans , Immunohistochemistry , Insulin/genetics , Insulin/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Pancreas/metabolism , Pancreatic Elastase/genetics , Pancreatic Elastase/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transfection
20.
Biochem Biophys Res Commun ; 410(3): 608-13, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21684256

ABSTRACT

Mouse embryonic stem cells (ESCs) can be induced to form pancreatic exocrine enzyme-producing cells in vitro in a stepwise fashion that recapitulates the development in vivo. However, there is no protocol for the differentiation of pancreatic-like cells from human ESCs (hESCs). Based upon the mouse ESC model, we have induced the in vitro formation of pancreatic exocrine enzyme-producing cells from hESCs. The protocol took place in four stages. In Stage 1, embryoid bodies (EBs) were formed from dissociated hESCs and then treated with the growth factor activin A, which promoted the expression of Foxa2 and Sox17 mRNAs, markers of definitive endoderm. In Stage 2, the cells were treated with all-trans retinoic acid which promoted the transition to cells that expressed gut tube endoderm mRNA marker HNF1b. In Stage 3, the cells were treated with fibroblast growth factor 7 (FGF7), which induced expression of Pdx1 typical of pancreatic progenitor cells. In Stage 4, treatment with FGF7, glucagon-like peptide 1, and nicotinamide induced the expression amylase (AMY) mRNA, a marker for mature pancreatic exocrine cells. Immunohistochemical staining showed the expression of AMY protein at the edges of cell clusters. These cells also expressed other exocrine secretory proteins including elastase, carboxypeptidase A, chymotrypsin, and pancreatic lipase in culture. Production of these hESC-derived pancreatic enzyme-producing cells represents a critical step in the study of pancreatic organogenesis and in the development of a renewable source of human pancreatic-like exocrine cells.


Subject(s)
Cell Culture Techniques , Cell Differentiation , Embryoid Bodies/cytology , Pancreas, Exocrine/cytology , Activins/pharmacology , Amylases/biosynthesis , Carboxypeptidases A/biosynthesis , Chymotrypsin/biosynthesis , Embryoid Bodies/drug effects , Embryoid Bodies/enzymology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/enzymology , Fibroblast Growth Factor 7/pharmacology , Glucagon-Like Peptide 1/pharmacology , Hepatocyte Nuclear Factor 3-beta/biosynthesis , Humans , Lipase/biosynthesis , Niacinamide/pharmacology , Pancreas, Exocrine/enzymology , Pancreatic Elastase/biosynthesis , SOXF Transcription Factors/biosynthesis , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...