Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Int J Mol Sci ; 22(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33915913

ABSTRACT

Most chemotherapeutics target DNA integrity and thereby trigger tumour cell death through activation of DNA damage responses that are tightly coupled to the cell cycle. Disturbances in cell cycle regulation can therefore lead to treatment resistance. Here, a comprehensive analysis of cell cycle checkpoint activation following doxorubicin (doxo) treatment was performed using flow cytometry, immunofluorescence and live-cell imaging in a panel of TP53 mutated ultra high-risk neuroblastoma (NB) cell lines, SK-N-DZ, Kelly, SK-N-AS, SK-N-FI, and BE(2)-C. Following treatment, a dose-dependent accumulation in either S- and/or G2/M-phase was observed. This coincided with a heterogeneous increase of cell cycle checkpoint proteins, i.e., phos-ATM, phos-CHK1, phos-CHK2, Wee1, p21Cip1/Waf1, and p27Kip among the cell lines. Combination treatment with doxo and a small-molecule inhibitor of ATM showed a delay in regrowth in SK-N-DZ, of CHK1 in BE(2)-C, of Wee1 in SK-N-FI and BE(2)-C, and of p21 in Kelly and BE(2)-C. Further investigation revealed, in all tested cell lines, a subset of cells arrested in mitosis, indicating independence on the intra-S- and/or G2/M-checkpoints. Taken together, we mapped distinct cell cycle checkpoints in ultra high-risk NB cell lines and identified checkpoint dependent and independent druggable targets.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Cell Cycle Checkpoints/drug effects , Doxorubicin/therapeutic use , Neuroblastoma/drug therapy , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Genes, p53 , Humans , Molecular Targeted Therapy , Neuroblastoma/genetics
2.
PLoS One ; 13(1): e0190970, 2018.
Article in English | MEDLINE | ID: mdl-29342186

ABSTRACT

In this study chemotherapy response in neuroblastoma (NB) was assessed for the first time in a transplantation model comprising non-malignant human embryonic microenvironment of pluripotent stem cell teratoma (PSCT) derived from diploid bona fide hESC. Two NB cell lines with known high-risk phenotypes; the multi-resistant BE(2)-C and the drug sensitive IMR-32, were transplanted to the PSCT model and the tumour growth was exposed to single or repeated treatments with doxorubicin, and thereafter evaluated for cell death, apoptosis, and proliferation. Dose dependent cytotoxic effects were observed, this way corroborating the experimental platform for this type of analysis. Notably, analysis of doxorubicin-resilient BE(2)-C growth in the PSCT model revealed an unexpected 1,5-fold increase in Ki67-index (p<0.05), indicating that non-cycling (G0) cells entered the cell cycle following the doxorubicin exposure. Support for this notion was obtained also in vitro. A pharmacologically relevant dose (1µM) resulted in a marked accumulation of Ki67 positive BE(2)-C cells (p<0.0001), as well as a >3-fold increase in active cell cycle (i.e. cells positive staining for PH3 together with incorporation of EdU) (p<0.01). Considering the clinical challenge for treating high-risk NB, the discovery of a therapy-provoked growth-stimulating effect in the multi-resistant and p53-mutated BE(2)-C cell line, but not in the drug-sensitive p53wt IMR-32 cell line, warrants further studies concerning generality and clinical significance of this new observation.


Subject(s)
Doxorubicin/pharmacology , Mitosis/drug effects , Neuroblastoma/pathology , Resting Phase, Cell Cycle , Animals , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Models, Biological
3.
Cell Death Dis ; 8(5): e2759, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28471449

ABSTRACT

Deregulation of microRNAs (miRNAs) contributes to the development and progression of many cancer types; however, their functions in the pathogenesis of testicular germ cell tumor (TGCT) remain unclear. Here, we determined miRNA expression profiles of TGCTs and normal testes using small RNA sequencing, and identified several deregulated miRNAs in TGCTs, including the miR-506~514 cluster. In functional studies in vitro we demonstrated that miR-514a-3p induced apoptosis through direct regulation of the paternally expressed gene 3 (PEG3), and ectopically expressed PEG3 could rescue the apoptotic effect of miR-514a-3p overexpression. Silencing of PEG3 or miR-514a-3p overexpression reduced nuclear accumulation of p50 and NF-κB reporter activity. Furthermore, PEG3 was co-immunoprecipitated with tumor necrosis factor receptor-associated factor 2 (TRAF2) in TGCT cell lysates. We propose a model of PEG3-mediated activation of NF-κB in TGCT. Loss of miR-514a-3p expression in TGCT increases PEG3 expression that recruits TRAF2 and activates the NF-kappa B pathway, which protects germ cells from apoptosis. Importantly, we observed strong expression of PEG3 and nuclear p50 in the majority of TGCTs (83% and 78%, respectively). In conclusion, our study describes a novel function for miR-514a-3p in TGCT and highlights an unrecognized mechanism of PEG3 regulation and NF-κB activation in TGCT.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , NF-kappa B/metabolism , Neoplasms, Germ Cell and Embryonal/pathology , Testicular Neoplasms/pathology , Antagomirs/metabolism , Apoptosis , Base Sequence , Cell Line, Tumor , DNA Methylation , Humans , Immunoprecipitation , Kruppel-Like Transcription Factors/antagonists & inhibitors , Kruppel-Like Transcription Factors/genetics , Male , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , NF-kappa B p50 Subunit/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Promoter Regions, Genetic , RNA Interference , Sequence Alignment , Signal Transduction , TNF Receptor-Associated Factor 2/metabolism , Testicular Neoplasms/metabolism , Transcriptome
4.
Am J Stem Cells ; 5(1): 19-28, 2016.
Article in English | MEDLINE | ID: mdl-27335699

ABSTRACT

Hearing impairment most often involves loss of sensory hair cells and auditory neurons. As this loss is permanent in humans, a cell therapy approach has been suggested to replace damaged cells. It is thus of interest to generate lineage restricted progenitor cells appropriate for cell based therapies. Human long-term self-renewing neuroepithelial stem (lt-NES) cell lines exhibit in vitro a developmental potency to differentiate into CNS neural lineages, and importantly lack this potency in vivo, i.e do not form teratomas. Small-molecules-driven differentiation is today an established route obtain specific cell derivatives from stem cells. In this study, we have investigated the effects of three small molecules SB431542, ISX9 and Metformin to direct differentiation of lt-NES cells into sensory neurons. Exposure of lt-NES cells to Metformin or SB431542 did not induce any marked induction of markers for sensory neurons. However, a four days exposure to the ISX9 small molecule resulted in reduced expression of NeuroD1 mRNA as well as enhanced mRNA levels of GATA3, a marker and important player in auditory neuron specification and development. Subsequent culture in the presence of the neurotrophic factors BDNF and NT3 for another seven days yielded a further increase of mRNA expression for GATA3. This regimen resulted in a frequency of up to 25-30% of cells staining positive for Brn3a/Tuj1. We conclude that an approach with ISX9 small molecule induction of lt-NES cells into auditory like neurons may thus be an attractive route for obtaining safe cell replacement therapy of sensorineural hearing loss.

5.
Nat Commun ; 7: 10458, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26822956

ABSTRACT

Non-CG methylation is an unexplored epigenetic hallmark of pluripotent stem cells. Here we report that a reduction in non-CG methylation is associated with impaired differentiation capacity into endodermal lineages. Genome-wide analysis of 2,670 non-CG sites in a discovery cohort of 25 phenotyped human induced pluripotent stem cell (hiPSC) lines revealed unidirectional loss (Δß=13%, P<7.4 × 10(-4)) of non-CG methylation that correctly identifies endodermal differentiation capacity in 23 out of 25 (92%) hiPSC lines. Translation into a simplified assay of only nine non-CG sites maintains predictive power in the discovery cohort (Δß=23%, P<9.1 × 10(-6)) and correctly identifies endodermal differentiation capacity in nine out of ten pluripotent stem cell lines in an independent replication cohort consisting of hiPSCs reprogrammed from different cell types and different delivery systems, as well as human embryonic stem cell (hESC) lines. This finding infers non-CG methylation at these sites as a biomarker when assessing endodermal differentiation capacity as a readout.


Subject(s)
Cell Differentiation , DNA Methylation , Endoderm/cytology , Induced Pluripotent Stem Cells/cytology , Biomarkers/metabolism , Cohort Studies , Endoderm/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism
6.
Biomed Res Int ; 2015: 754876, 2015.
Article in English | MEDLINE | ID: mdl-26557696

ABSTRACT

We have previously shown that human embryonic stem cell (hESC) therapy to injured rabbit vocal folds (VFs) induces human tissue generation with regained VF vibratory capacity. The aims of this study were to test the sustainability of such effect and to what extent derivatives of the transplanted hESCs are propagated in the VFs. The VFs of 14 New Zealand rabbits were injured by a localized resection. HESCs were transplanted to 22 VFs which were analyzed for persistence of hESCs after six weeks and after three months. At three months, the VFs were also analyzed for viscoelasticity, measured as dynamic viscosity and elastic modulus, for the lamina propria (Lp) thickness and relative content of collagen type I. Three months after hESC cell therapy, the dynamic viscosity and elastic modulus of the hESC treated VFs were similar to normal controls and lower than untreated VFs (p ≤ 0.011). A normalized VF architecture, reduction in collagen type I, and Lp thickness were found compared with untreated VFs (p ≤ 0.031). At three months, no derivatives of hESCs were detected. HESCs transplanted to injured rabbit VFs restored the vibratory characteristics of the VFs, with maintained restored function for three months without remaining hESCs or derivatives.


Subject(s)
Heterografts/physiology , Human Embryonic Stem Cells/transplantation , Mesenchymal Stem Cell Transplantation/methods , Transplantation, Heterologous/methods , Vocal Cords/injuries , Wound Healing/physiology , Animals , Cell Line , Elastic Modulus/physiology , Female , Human Embryonic Stem Cells/cytology , Humans , Rabbits , Viscosity
7.
PLoS One ; 9(9): e107712, 2014.
Article in English | MEDLINE | ID: mdl-25229469

ABSTRACT

Stem cells contribute to regeneration of tissues and organs. Cells with stem cell-like properties have been identified in tumors from a variety of origins, but to our knowledge there are yet no reports on tumor-related stem cells in the human upper respiratory tract. In the present study, we show that a tracheal mucoepidermoid tumor biopsy obtained from a 6 year-old patient contained a subpopulation of cells with morphology, clonogenicity and surface markers that overlapped with bone marrow mesenchymal stromal cells (BM-MSCs). These cells, designated as MEi (mesenchymal stem cell-like mucoepidermoid tumor) cells, could be differentiated towards mesenchymal lineages both with and without induction, and formed spheroids in vitro. The MEi cells shared several multipotent characteristics with BM-MSCs. However, they displayed differences to BM-MSCs in growth kinectics and gene expression profiles relating to cancer pathways and tube development. Despite this, the MEi cells did not possess in vivo tumor-initiating capacity, as proven by the absence of growth in situ after localized injection in immunocompromised mice. Our results provide an initial characterization of benign tracheal cancer-derived niche cells. We believe that this report could be of importance to further understand tracheal cancer initiation and progression as well as therapeutic development.


Subject(s)
Mucoepidermoid Tumor/pathology , Neoplastic Stem Cells/pathology , Tracheal Neoplasms/pathology , Animals , Cell Separation , Child , Female , Gene Expression Profiling , Genomics , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Mucoepidermoid Tumor/diagnosis , Mucoepidermoid Tumor/genetics , Tracheal Neoplasms/diagnosis , Tracheal Neoplasms/genetics
8.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25079316

ABSTRACT

Mesenchymal stem cells occupy niches in stromal tissues where they provide sources of cells for specialized mesenchymal derivatives during growth and repair. The origins of mesenchymal stem cells have been the subject of considerable discussion, and current consensus holds that perivascular cells form mesenchymal stem cells in most tissues. The continuously growing mouse incisor tooth offers an excellent model to address the origin of mesenchymal stem cells. These stem cells dwell in a niche at the tooth apex where they produce a variety of differentiated derivatives. Cells constituting the tooth are mostly derived from two embryonic sources: neural crest ectomesenchyme and ectodermal epithelium. It has been thought for decades that the dental mesenchymal stem cells giving rise to pulp cells and odontoblasts derive from neural crest cells after their migration in the early head and formation of ectomesenchymal tissue. Here we show that a significant population of mesenchymal stem cells during development, self-renewal and repair of a tooth are derived from peripheral nerve-associated glia. Glial cells generate multipotent mesenchymal stem cells that produce pulp cells and odontoblasts. By combining a clonal colour-coding technique with tracing of peripheral glia, we provide new insights into the dynamics of tooth organogenesis and growth.


Subject(s)
Cell Differentiation , Cell Lineage , Incisor/cytology , Mesenchymal Stem Cells/cytology , Neuroglia/cytology , Animals , Cell Tracking , Clone Cells/cytology , Dental Pulp/cytology , Female , Incisor/embryology , Male , Mice , Models, Biological , Neural Crest/cytology , Odontoblasts/cytology , Regeneration , Schwann Cells/cytology
10.
Semin Cancer Biol ; 29: 75-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25153353

ABSTRACT

Xenografting is the so far only available in vivo model for assessing pluripotency of human stem cells. This review describes known biological features of experimental teratoma from human pluripotent stem cells. We focus on the dual nature mimicking both normal and abnormal development, and propose this model system to be particularly interesting for investigations of the relationship between developmentally controlled differentiation and neoplasia of embryonic origin. In resemblance to the wide range of clinical teratomas, pluripotent stem cell (PSC) induced teratoma (PSCT) typically shows a mixture of developing tissues in randomly distributed compartments. The combined literature suggests that for teratomas derived from human diploid bona fide PSC the embryonic development in the separate tissue-niches can show a controlled differentiation into organoid patterns closely mimicking early development. In the experimental situation such PSCT human homologous in vivo tissue-niches have been shown to provide also matching microenvironment for a micrometastatic colonization and outgrowth of embryonic tumors transplanted directly from patients. Single or small clusters of normal and neoplastic cells can easily be visualized together in microscope-based imaging systems, enabling multi-parameter detection of in the scans of tissue slides/specimens.


Subject(s)
Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Teratoma/pathology , Animals , Cell Differentiation , Cellular Microenvironment , Embryonic Development , Humans , Mice , Neoplasm Transplantation , Transplantation, Heterologous
11.
Int J Cancer ; 134(7): 1630-7, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24122295

ABSTRACT

Experimental teratoma induced from human pluripotent stem cells with normal karyotype can be described as a failed embryonic process and includes besides advanced organoid development also large elements of tissue with a prolonged occurrence of immature neural components. Such immature components, although benign, exhibit strong morphological resemblance with tumors of embryonic neuroectodermal origin. Here, we demonstrate that biopsy material from childhood tumors of neural embryonic origin transplanted to mature experimental teratoma can show an exclusive preference for matching tissue. Tumor specimens from five children with; Supratentorial primitive neuroectodermal tumor (sPNET); Pilocytic astrocytoma of the brainstem; Classic medulloblastoma; peripheral primitive neuroectodermal tumor (pPNET) or neuroblastoma (NB), respectively, were transplanted. Analysis of up to 120 sections of each tumor revealed an engraftment for three of the transplanted tumors: pPNET, sPNET, and NB, with a protruding growth from the latter two that were selected for detailed examination. The histology revealed a strict tropism with a non-random integration into what morphologically appeared as matched embryonic microenvironment recuperating the patient tumor histology. The findings suggest specific advantages over xenotransplantation and lead us to propose that transplantation to the human embryonic microenvironment in experimental teratoma can be a well-needed complement for preclinical in vivo studies of childhood neuroectodermal tumors.


Subject(s)
Neuroectodermal Tumors, Primitive/pathology , Teratoma/pathology , Tropism/physiology , Animals , Astrocytoma/pathology , Biopsy/methods , Child , Child, Preschool , Female , Humans , Infant , Male , Medulloblastoma/pathology , Mice , Neuroblastoma/pathology , Pluripotent Stem Cells/pathology , Transplantation, Heterologous/methods
12.
Cancer Res ; 72(8): 1915-20, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22508696

ABSTRACT

Nodal is a TGF-ß-related embryonic morphogen that is expressed in multiple human cancers. Detection of Nodal expression in these tissues can be challenging if issues related to Nodal transcription and protein processing are not considered. Here, we discuss certain characteristics related to Nodal expression and function and how these can facilitate acquisition and interpretation of expression data, contributing to our understanding of the potential role of Nodal in human cancer. We also discuss how Nodal could be exploited clinically as a novel biomarker for cancer progression and therapeutic target.


Subject(s)
Neoplasms , Nodal Protein/physiology , Humans
13.
Stem Cells ; 30(1): 55-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22069231

ABSTRACT

Hematopoietic cell transplantation (HCT) has become a standard practice to treat a number of malignant and nonmalignant hematologic diseases. Bone marrow, mobilized peripheral blood, and umbilical cord blood can all serve as primary sources of cells for HCT. The number of cord blood units currently stored is large, although it represents only a fraction of potential collections. With much of the collection being sequestered in private banks for possible autologous use, there is a reason to expect that public banks may not be able to provide for the demand in coming years as use of cord blood for treatment of patients with diseases such as leukemia and lymphoma continues to increase. We suggest that a possible solution to encourage private banks to share their valuable units is to apply recent methodologies to generate induced pluripotent stem cells from cord cells and to optimize techniques to generate hematopoietic lineages from them. This strategy would allow us to take advantage of the units already collected under appropriate regulatory guidelines, to access a pristine cell that can be converted to a pluripotent cell at a much higher efficiency and in a shorter time period than other cells. The ability to potentially replenish a used cord unit with new cells, as well as extend the potential utility of cord blood for additional therapeutic applications, should allow banks to develop an appropriate business model for both private and public cord blood banks to flourish.


Subject(s)
Blood Banks , Cord Blood Stem Cell Transplantation , Fetal Blood/cytology , Induced Pluripotent Stem Cells/cytology , Blood Banks/ethics , Bone Marrow/physiology , Hematopoietic Stem Cell Transplantation , Humans , Induced Pluripotent Stem Cells/physiology , Private Sector , Public Sector , Transplantation, Autologous
14.
PLoS One ; 6(11): e27741, 2011.
Article in English | MEDLINE | ID: mdl-22140465

ABSTRACT

Xenografting is widely used for assessing in vivo pluripotency of human stem cell populations. Here, we report on early to late events in the development of mature experimental teratoma from a well-characterized human embryonic stem cell (HESC) line, HS181. The results show an embryonic process, increasingly chaotic. Active proliferation of the stem cell derived cellular progeny was detected already at day 5, and characterized by the appearance of multiple sites of engraftment, with structures of single or pseudostratified columnar epithelium surrounding small cavities. The striking histological resemblance to developing embryonic ectoderm, and the formation of epiblast-like structures was supported by the expression of the markers OCT4, NANOG, SSEA-4 and KLF4, but a lack of REX1. The early neural marker NESTIN was uniformly expressed, while markers linked to gastrulation, such as BMP-4, NODAL or BRACHYURY were not detected. Thus, observations on day 5 indicated differentiation comparable to the most early transient cell populations in human post implantation development. Confirming and expanding on previous findings from HS181 xenografts, these early events were followed by an increasingly chaotic development, incorporated in the formation of a benign teratoma with complex embryonic components. In the mature HS181 teratomas not all types of organs/tissues were detected, indicating a restricted differentiation, and a lack of adequate spatial developmental cues during the further teratoma formation. Uniquely, a kinetic alignment of rare complex structures was made to human embryos at diagnosed gestation stages, showing minor kinetic deviations between HS181 teratoma and the human counterpart.


Subject(s)
Embryonic Stem Cells/cytology , Germ Layers/cytology , Transplantation, Heterologous , Biomarkers/metabolism , Cell Differentiation , Cell Line , Embryonic Stem Cells/metabolism , Germ Layers/metabolism , Humans , Kruppel-Like Factor 4 , Limb Buds/metabolism , Limb Buds/pathology , Neurons/metabolism , Neurons/pathology , Teratoma/metabolism , Teratoma/pathology , Time Factors
15.
Laryngoscope ; 121(10): 2185-90, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21898432

ABSTRACT

OBJECTIVES: Using a xenograft model the aim was to analyze if injection of human mesenchymal stem cells (hMSC) into the rabbit vocal fold (VF), after excision of an established scar, can improve the functional healing of the VF. STUDY DESIGN: Prospective design with an experimental xenograft model. METHODS: The VFs of 12 New Zealand rabbits were injured by a bilateral localized resection. After 9 weeks the scar after the resection was excised and hMSC were injected into the VFs. After another 10 weeks 10 VFs were dissected and stained for histology. Lamina propria thickness and relative content of collagen type I were measured. Viscoelasticity of 14 VFs at phonatory frequencies was quantified by a simple-shear rheometer. The hMSC survival was determined using a human DNA specific reference probe, that is, FISH analysis. RESULTS: The viscoelastic measurements, that is, dynamic viscosity and elastic shear modulus for the hMSC-treated VFs, were found to be similar to those of normal controls and were significantly lower than those of untreated controls (P < .05). A significant reduction in lamina propria thickness was also shown for the hMSC treated VFs compared with the untreated VFs (P < .05). This histologic finding corresponded with the viscoelastic results. No hMSC survived 10 weeks after the injection. CONCLUSIONS: Human mesenchymal stem cells injected into the rabbit VF following the excision of a chronic scar, were found to enhance the functional healing of the VF with reduced lamina propria thickness and restored viscoelastic shear properties.


Subject(s)
Cicatrix/surgery , Mesenchymal Stem Cell Transplantation/methods , Mucous Membrane/pathology , Wound Healing/physiology , Animals , Cicatrix/pathology , Disease Models, Animal , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Injections, Intralesional , Rabbits , Random Allocation , Reference Values , Statistics, Nonparametric , Transplantation, Heterologous , Vocal Cords/pathology , Vocal Cords/surgery
16.
J Clin Invest ; 120(9): 3127-36, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20739751

ABSTRACT

Human induced pluripotent stem (iPS) cells hold great promise for advancements in developmental biology, cell-based therapy, and modeling of human disease. Here, we examined the use of human iPS cells for modeling inherited metabolic disorders of the liver. Dermal fibroblasts from patients with various inherited metabolic diseases of the liver were used to generate a library of patient-specific human iPS cell lines. Each line was differentiated into hepatocytes using what we believe to be a novel 3-step differentiation protocol in chemically defined conditions. The resulting cells exhibited properties of mature hepatocytes, such as albumin secretion and cytochrome P450 metabolism. Moreover, cells generated from patients with 3 of the inherited metabolic conditions studied in further detail (alpha1-antitrypsin deficiency, familial hypercholesterolemia, and glycogen storage disease type 1a) were found to recapitulate key pathological features of the diseases affecting the patients from which they were derived, such as aggregation of misfolded alpha1-antitrypsin in the endoplasmic reticulum, deficient LDL receptor-mediated cholesterol uptake, and elevated lipid and glycogen accumulation. Therefore, we report a simple and effective platform for hepatocyte generation from patient-specific human iPS cells. These patient-derived hepatocytes demonstrate that it is possible to model diseases whose phenotypes are caused by pathological dysregulation of key processes within adult cells.


Subject(s)
Induced Pluripotent Stem Cells , Liver Diseases , Liver/metabolism , Adult , Aged , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell- and Tissue-Based Therapy , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Glycogen/metabolism , Glycogen/pharmacology , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Infant , Male , Middle Aged , Models, Biological
17.
Laryngoscope ; 120(7): 1370-5, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20568271

ABSTRACT

OBJECTIVES/HYPOTHESIS: The aims were to analyze if improved histological and viscoelastic properties seen after injection of human mesenchymal stem cells (hMSCs) in scarred vocal folds (VFs) of rabbits are sustainable and if the injected hMSCs survive 3 months in the VFs. STUDY DESIGN: Experimental xenograft model. METHODS: Eighteen VFs of 11 New Zealand white rabbits were scarred by a bilateral localized resection. After 3 months the animals were sacrificed. Twelve VFs were dissected and stained for histology, lamina propria thickness, and relative collagen type I analyses. The hMSCs survival was analyzed using a human DNA-specific reference probe, that is, fluorescence in situ hybridization staining. Viscoelasticity, measured as the dynamic viscosity and elastic modulus, was analyzed in a parallel-plate rheometer for 10 VFs. RESULTS: The dynamic viscosity and elastic modulus of hMSC-treated VFs were similar to that of normal controls and significantly improved compared to untreated controls (P < .05). A reduction in lamina propria thickness and relative collagen type 1 content were also shown for the hMSC-treated VFs compared to the untreated VFs (P < .05). The histological pictures corresponded well to the viscoelastic results. No hMSCs survived. CONCLUSIONS: Human mesenchymal stem cells injected into a scarred vocal fold of rabbit enhance healing of the vocal fold with reduced lamina propria thickness and collagen type I content and restore the viscoelastic function.


Subject(s)
Mesenchymal Stem Cell Transplantation , Vocal Cords/surgery , Animals , Cell Survival/physiology , Cicatrix , Collagen Type I/analysis , Humans , Injections , Mucous Membrane/surgery , Rabbits , Time Factors , Transplantation, Heterologous , Vocal Cords/pathology , Vocal Cords/physiology , Wound Healing/physiology
18.
Stem Cells Dev ; 19(8): 1257-66, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19886822

ABSTRACT

Mesenchymal stem cells (MSCs) can differentiate into multiple mesodermal cell types in vitro; however, their differentiation capacity is influenced by their tissue of origin. To what extent epigenetic information on promoters of lineage-specification genes in human progenitors influences transcriptional activation and differentiation potential remains unclear. We produced bisulfite sequencing maps of DNA methylation in adipogenic, myogenic, and endothelial promoters in relation to gene expression and differentiation capacity, and unravel a similarity in DNA methylation profiles between MSCs isolated from human adipose tissue, bone marrow (BM), and muscle. This similarity is irrespective of promoter CpG content. Methylation patterns of MSCs are distinct from those of hematopoietic progenitor cells (HPCs), pluripotent human embryonic stem cells (hESCs), and multipotent hESC-derived mesenchymal cells (MCs). Moreover, in vitro MSC differentiation does not affect lineage-specific promoter methylation states, arguing that these methylation patterns in differentiated cells are already established at the progenitor stage. Further, we find a correlation between lineage-specific promoter hypermethylation and lack of differentiation capacity toward that lineage, but no relationship between weak promoter methylation and capacity of transcriptional activation or differentiation. Thus, only part of the restriction in differentiation capacity of tissue-specific stem cells is programmed by promoter DNA methylation: hypermethylation seems to constitute a barrier to differentiation, however, no or weak methylation has no predictive value for differentiation potential.


Subject(s)
Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Cell Lineage/physiology , DNA Methylation , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Promoter Regions, Genetic/genetics , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/cytology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/physiology , Cell Line , CpG Islands/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle, Skeletal/cytology , Platelet Endothelial Cell Adhesion Molecule-1/genetics
19.
Stem Cells ; 27(11): 2655-66, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19688839

ABSTRACT

Human pluripotent stem cells from embryonic origins and those generated from reprogrammed somatic cells share many characteristics, including indefinite proliferation and a sustained capacity to differentiate into a wide variety of cell types. However, it remains to be demonstrated whether both cell types rely on similar mechanisms to maintain their pluripotent status and to control their differentiation. Any differences in such mechanisms would suggest that reprogramming of fibroblasts to generate induced pluripotent stem cells (iPSCs) results in novel states of pluripotency. In that event, current methods for expanding and differentiating human embryonic stem cells (ESCs) might not be directly applicable to human iPSCs. However, we show here that human iPSCs rely on activin/nodal signaling to control Nanog expression and thereby maintain pluripotency, thus revealing their mechanistic similarity to human ESCs. We also show that growth factors necessary and sufficient for achieving specification of human ESCs into extraembryonic tissues, neuroectoderm, and mesendoderm also drive differentiation of human iPSCs into the same tissues. Importantly, these experiments were performed in fully chemically defined medium devoid of factors that could obscure analysis of developmental mechanisms or render the resulting tissues incompatible with future clinical applications. Together these data reveal that human iPSCs rely on mechanisms similar to human ESCs to maintain their pluripotency and to control their differentiation, showing that these pluripotent cell types are functionally equivalent.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Signal Transduction/physiology , Activin Receptors/antagonists & inhibitors , Activins/pharmacology , Adult , Animals , Benzamides/pharmacology , Bone Morphogenetic Protein 4/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Culture Media , Dioxoles/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Flow Cytometry , Fluorescent Antibody Technique , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Male , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/physiology , Polymerase Chain Reaction , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/physiology , Signal Transduction/drug effects , Signal Transduction/genetics
20.
Eur J Pharmacol ; 618(1-3): 84-90, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19622351

ABSTRACT

We have tested the protective effect of melatonin on neonate murine ovarian tissue after vitrification, thawing and heterotopic transplantation into ovariectomized recipient mice. Vitrified ovaries from neonate (CBA x C57Bl/6) F1 hybrid mice were thawed under standard condition with or without the addition of 100 microM melatonin. Following transplantation, melatonin (20 mg/kg/day) or saline solution (physiological saline) was injected i.p. to the treated and non-treated groups for 48 h respectively. Follicle survival and development, together with ovary size followed. Also, vaginal cytology was carried out for monitoring restored puberty. Histological and immunohistochemical studies showed that melatonin could promote the quality of the cumulus-oocyte complexes with uniform distribution of granulosa and stromal cells in the ovarian grafts. Furthermore, the mean follicles survival was improved and the ovary size increased (P< or = 0.001). The overall mean number of follicles entering the next maturation stage dramatically increased. However, the revascularization and restoration of puberty of ovarian grafts were similar between melatonin-treated and control groups. In conclusion, melatonin as a protection from ischemic injury and a reduce oxidative stress, was shown beneficial during the early days of transplantation.


Subject(s)
Cumulus Cells/drug effects , Melatonin/pharmacology , Oocytes/drug effects , Ovarian Follicle/drug effects , Ovary/drug effects , Tissue Survival/drug effects , Transplantation, Heterotopic , Animals , Cumulus Cells/metabolism , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Oocytes/metabolism , Organ Size/drug effects , Ovarian Follicle/blood supply , Ovarian Follicle/physiology , Ovariectomy , Ovary/anatomy & histology , Ovary/physiology , Ovary/transplantation , Vagina/cytology , Vagina/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...