Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Trends Biotechnol ; 41(11): 1343-1359, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37302911

ABSTRACT

Exosome-based strategies constitute a promising tool for therapeutics, avoiding potential immunogenic and tumorigenic side-effects of cell therapies. However, the collection of a suitable exosome pool, and the need for high doses with conventional administration approaches, hamper their clinical translation. To overcome these challenges, versatile exosome collection strategies together with advanced delivery platforms may represent major progress in this field. Microfluidics enables large-scale gathering of both natural and synthetic exosomes for their implementation into bioinks, while 3D-bioprinting holds great promise in regenerative medicine with the use of exosome-loaded scaffolds that mimic the target tissue with controlled pharmacokinetics and pharmacodynamics. Hence, the combination of both strategies might become the key for the translation of exosome therapies to clinical practice.

2.
Int J Mol Sci ; 24(11)2023 May 31.
Article in English | MEDLINE | ID: mdl-37298538

ABSTRACT

New therapeutic options for liver cirrhosis are needed. Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have emerged as a promising tool for delivering therapeutic factors in regenerative medicine. Our aim is to establish a new therapeutic tool that employs EVs derived from MSCs to deliver therapeutic factors for liver fibrosis. EVs were isolated from supernatants of adipose tissue MSCs, induced-pluripotent-stem-cell-derived MSCs, and umbilical cord perivascular cells (HUCPVC-EVs) by ion exchange chromatography (IEC). To produce engineered EVs, HUCPVCs were transduced with adenoviruses that code for insulin-like growth factor 1 (AdhIGF-I-HUCPVC-EVs) or green fluorescent protein. EVs were characterized by electron microscopy, flow cytometry, ELISA, and proteomic analysis. We evaluated EVs' antifibrotic effect in thioacetamide-induced liver fibrosis in mice and on hepatic stellate cells in vitro. We found that IEC-isolated HUCPVC-EVs have an analogous phenotype and antifibrotic activity to those isolated by ultracentrifugation. EVs derived from the three MSCs sources showed a similar phenotype and antifibrotic potential. EVs derived from AdhIGF-I-HUCPVC carried IGF-1 and showed a higher therapeutic effect in vitro and in vivo. Remarkably, proteomic analysis revealed that HUCPVC-EVs carry key proteins involved in their antifibrotic process. This scalable MSC-derived EV manufacturing strategy is a promising therapeutic tool for liver fibrosis.


Subject(s)
Extracellular Vesicles , Mesenchymal Stem Cells , Mice , Animals , Proteomics , Liver Cirrhosis/chemically induced , Liver Cirrhosis/therapy , Liver Cirrhosis/metabolism , Hepatic Stellate Cells/metabolism , Mesenchymal Stem Cells/metabolism , Extracellular Vesicles/metabolism
3.
Shock ; 59(6): 941-947, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37036956

ABSTRACT

ABSTRACT: Background : Mesenchymal stem cells (MSCs) can be activated by different bacterial toxins. Lipopolysaccharides and Shiga Toxin (Stx) are the main toxins necessary for hemolytic uremic syndrome development. The main etiological event in this disease is endothelial damage that causes glomerular destruction. Considering the repairing properties of MSC, we aimed to study the response of MSC derived from induced pluripotent stem cells (iPSC-MSC) to LPS and/or Stx and its effect on the restoration of injured endothelial cells. Methods : iPSC-MSC were treated with LPS and or/Stx for 24 h and secretion of cytokines, adhesion, and migration were measured in response to these toxins. In addition, conditioned media from treated iPSC-MSC were collected and used for proteomics analysis and evaluation of endothelial cell healing and tubulogenesis using human microvascular endothelial cells 1 as a source of endothelial cells. Results : The results obtained showed that LPS induced a proinflammatory profile on iPSC-MSC, whereas Stx effects were less evident, even though cells expressed the Gb 3 receptor. Moreover, LPS induced on iPSC-MSC an increment in migration and adhesion to a gelatin substrate. Addition of conditioned media of iPSC-MSC treated with LPS + Stx, decreased the capacity of human microvascular endothelial cells 1 to close a wound, and did not favor tubulogenesis. Proteomic analysis of iPSC-MSC treated with LPS and/or Stx revealed specific protein secretion patterns that support the functional results described. Conclusions : iPSC-MSC activated by LPS acquired a proinflammatory profile that induces migration and adhesion to extracellular matrix proteins but the addition of Stx did not activate any repair program to ameliorate endothelial damage, indicating that the use of iPSC-MSC to regenerate endothelial injury caused by LPS and/or Stx in hemolytic uremic syndrome could not be the best option to consider to regenerate a tissue injury.


Subject(s)
Hemolytic-Uremic Syndrome , Induced Pluripotent Stem Cells , Humans , Shiga Toxin , Lipopolysaccharides/pharmacology , Endothelial Cells/metabolism , Culture Media, Conditioned , Proteomics
4.
R Soc Open Sci ; 9(1): 211510, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35242349

ABSTRACT

Microfluidic tools have recently made possible many advances in biological and biomedical research. Research in fields such as physics, engineering, chemistry and biology have combined to produce innovation in microfluidics which has positively impacted diverse areas such as nucleotide sequencing, functional genomics, single-cell studies, single molecules assays and biomedical diagnostics. Among these areas, regenerative medicine and stem cells have benefited from microfluidics since these tools have had a profound impact on their applications. In this study, we present a high-performance droplet-based system for transfecting individual human-induced pluripotent stem cells. We will demonstrate that this system has great efficiency in single cells and captured droplets, like other microfluidic methods but with lower cost. Moreover, this microfluidic approach can be associated with the PiggyBac transposase-based system to increase its transfection efficiency. Our results provide a starting point for subsequent applications in more complex transfection systems, single-cell differentiation interactions, cell subpopulations and cell therapy, among other potential applications.

5.
PLoS One ; 16(6): e0253666, 2021.
Article in English | MEDLINE | ID: mdl-34166446

ABSTRACT

Cell death experiments are routinely done in many labs around the world, these experiments are the backbone of many assays for drug development. Cell death detection is usually performed in many ways, and requires time and reagents. However, cell death is preceded by slight morphological changes in cell shape and texture. In this paper, we trained a neural network to classify cells undergoing cell death. We found that the network was able to highly predict cell death after one hour of exposure to camptothecin. Moreover, this prediction largely outperforms human ability. Finally, we provide a simple python tool that can broadly be used to detect cell death.


Subject(s)
Deep Learning , Image Interpretation, Computer-Assisted , Programming Languages , Cell Death , Humans , MCF-7 Cells , Microscopy
6.
Cell Transplant ; 30: 963689721993774, 2021.
Article in English | MEDLINE | ID: mdl-33975446

ABSTRACT

In normal physiological conditions, restoration of a functional epidermal barrier is highly efficient; nevertheless, when it fails, one of the main consequences is a chronic ulcerative skin defect, one of the most frequently recognized complications of diabetes. Most of these chronic venous ulcers do not heal with conventional treatment, leading to the appearance of infections and complications in the patient. Treatments based on the use of autologous mesenchymal stem cells (MSC) have been successful; however, its implementation entails complications. The umbilical cord offers an unlimited source of adult MSC (ucMSC) from the Wharton's jelly tissue with the same relevant features for clinical applicability and avoiding difficulties. It has recently been characterized by one specific subpopulation derived from ucMSC, the differentiated mesenchymal cells (DMCs). This subpopulation expresses the human leukocyte antigen-G (HLA-G) molecule, a strong immunosuppressive checkpoint, and vascular endothelial growth factor (VEGF), the most potent angiogenic factor. Considering the importance of developing a more effective therapy for wound treatment, especially ulcerative skin lesions, we analyzed DMC safety, efficacy, and therapeutic potential. By immunohistochemistry, umbilical cords HLA-G and VEGF positive were selected. Flow cytometry revealed that 90% of the DMC subpopulation are HLA-G+, CD44+, CD73+, CD29+, CD105+, CD90+, and HLA-DR-. Reverse transcription-polymerase chain reaction revealed the expression of HLA-G in all of DMC subpopulations. Upon co-culture with the DMC, peripheral blood mononuclear cell proliferation was inhibited by 50%. In a xenograft transplantation assay, DMC improved wound healing with no signs of rejection of the transplanted cells in immunocompetent mice. This study confirms that HLA-G allows allogeneic cell transplantation, and VEGF is fundamental for the restoration of the failure in blood supply. DMC population has positive effects on wound healing by promoting local angiogenesis in skin lesions. DMC could play a very important role in regenerative medicine and could be a novel allogeneic cell-therapeutic tool for wound healing.


Subject(s)
Mesenchymal Stem Cells/metabolism , Transplantation, Homologous/methods , Umbilical Cord/metabolism , Wound Healing/physiology , Animals , Disease Models, Animal , Female , Humans , Mice
7.
PLoS One ; 15(5): e0232715, 2020.
Article in English | MEDLINE | ID: mdl-32369512

ABSTRACT

PIWI-interacting RNAs (piRNAs) are a class of non-coding RNAs initially thought to be restricted exclusively to germline cells. In recent years, accumulating evidence has demonstrated that piRNAs are actually expressed in pluripotent, neural, cardiac and even cancer cells. However, controversy remains around the existence and function of somatic piRNAs. Using small RNA-seq samples from H9 pluripotent cells differentiated to mesoderm progenitors and cardiomyocytes we identified the expression of 447 piRNA transcripts, of which 241 were detected in pluripotency, 218 in mesoderm and 171 in cardiac cells. The majority of them originated from the sense strand of protein coding and lncRNAs genes in all stages of differentiation, though no evidences of amplification loop (ping-pong) were found. Genes hosting piRNA transcripts in cardiac samples were related to critical biological processes in the heart, like contraction and cardiac muscle development. Our results indicate that these piRNAs might have a role in fine-tuning the expression of genes involved in differentiation of pluripotent cells to cardiomyocytes.


Subject(s)
Human Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , RNA, Small Interfering/genetics , Adult , Cell Differentiation , Cell Line , Gene Expression Regulation, Developmental , Human Embryonic Stem Cells/metabolism , Humans , Male , Middle Aged , Myocytes, Cardiac/metabolism
8.
Sci Rep ; 9(1): 18077, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792288

ABSTRACT

The stem cell niche has a strong influence in the differentiation potential of human pluripotent stem cells with integrins playing a major role in communicating cells with the extracellular environment. However, it is not well understood how interactions between integrins and the extracellular matrix are involved in cardiac stem cell differentiation. To evaluate this, we performed a profile of integrins expression in two stages of cardiac differentiation: mesodermal progenitors and cardiomyocytes. We found an active regulation of the expression of different integrins during cardiac differentiation. In particular, integrin α5 subunit showed an increased expression in mesodermal progenitors, and a significant downregulation in cardiomyocytes. To analyze the effect of α5 subunit, we modified its expression by using a CRISPRi technique. After its downregulation, a significant impairment in the process of epithelial-to-mesenchymal transition was seen. Early mesoderm development was significantly affected due to a downregulation of key genes such as T Brachyury and TBX6. Furthermore, we observed that repression of integrin α5 during early stages led to a reduction in cardiomyocyte differentiation and impaired contractility. In summary, our results showed the link between changes in cell identity with the regulation of integrin α5 expression through the alteration of early stages of mesoderm commitment.


Subject(s)
Human Embryonic Stem Cells/cytology , Integrin alpha5/genetics , Myocytes, Cardiac/cytology , CRISPR-Cas Systems , Cell Differentiation , Cell Line , Down-Regulation , Gene Expression Regulation, Developmental , HEK293 Cells , Human Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Stem Cell Niche
9.
Stem Cell Reports ; 12(4): 845-859, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30880077

ABSTRACT

Deep learning is a significant step forward for developing autonomous tasks. One of its branches, computer vision, allows image recognition with high accuracy thanks to the use of convolutional neural networks (CNNs). Our goal was to train a CNN with transmitted light microscopy images to distinguish pluripotent stem cells from early differentiating cells. We induced differentiation of mouse embryonic stem cells to epiblast-like cells and took images at several time points from the initial stimulus. We found that the networks can be trained to recognize undifferentiated cells from differentiating cells with an accuracy higher than 99%. Successful prediction started just 20 min after the onset of differentiation. Furthermore, CNNs displayed great performance in several similar pluripotent stem cell (PSC) settings, including mesoderm differentiation in human induced PSCs. Accurate cellular morphology recognition in a simple microscopic set up may have a significant impact on how cell assays are performed in the near future.


Subject(s)
Cell Differentiation , Deep Learning , Neural Networks, Computer , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cells, Cultured , Humans , Image Processing, Computer-Assisted , Machine Learning , Microscopy
10.
PLoS One ; 13(12): e0207074, 2018.
Article in English | MEDLINE | ID: mdl-30507934

ABSTRACT

Cell reprogramming has been well described in mouse and human cells. The expression of specific microRNAs has demonstrated to be essential for pluripotent maintenance and cell differentiation, but not much information is available in domestic species. We aim to generate horse iPSCs, characterize them and evaluate the expression of different microRNAs (miR-302a,b,c,d, miR-205, miR-145, miR-9, miR-96, miR-125b and miR-296). Two equine iPSC lines (L2 and L3) were characterized after the reprogramming of equine fibroblasts with the four human Yamanaka's factors (OCT-4/SOX-2/c-MYC/KLF4). The pluripotency of both lines was assessed by phosphatase alkaline activity, expression of OCT-4, NANOG and REX1 by RT-PCR, and by immunofluorescence of OCT-4, SOX-2 and c-MYC. In vitro differentiation to embryo bodies (EBs) showed the capacity of the iPSCs to differentiate into ectodermal, endodermal and mesodermal phenotypes. MicroRNA analyses resulted in higher expression of the miR-302 family, miR-9 and miR-96 in L2 and L3 vs. fibroblasts (p<0.05), as previously shown in human pluripotent cells. Moreover, downregulation of miR-145 and miR-205 was observed. After differentiation to EBs, higher expression of miR-96 was observed in the EBs respect to the iPSCs, and also the expression of miR-205 was induced but only in the EB-L2. In addition, in silico alignments of the equine microRNAs with mRNA targets suggested the ability of miR-302 family to regulate cell cycle and epithelial mesenchymal transition genes, miR-9 and miR-96 to regulate neural determinant genes and miR-145 to regulate pluripotent genes, similarly as in humans. In conclusion, we could obtain equine iPSCs, characterize them and determine for the first time the expression level of microRNAs in equine pluripotent cells.


Subject(s)
Horses , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/genetics , Animals , Cell Differentiation/genetics , Fibroblasts/cytology , Gene Expression Profiling , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Nuclear Transfer Techniques
11.
Exp Mol Med ; 50(9): 1-12, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30201949

ABSTRACT

Mesenchymal stem/stromal cells (MSCs) obtained from pluripotent stem cells (PSCs) constitute an interesting alternative to classical MSCs in regenerative medicine. Among their many mechanisms of action, MSC extracellular vesicles (EVs) are a potential suitable substitute for MSCs in future cell-free-based therapeutic approaches. Unlike cells, EVs do not elicit acute immune rejection, and they can be produced in large quantities and stored until ready to use. Although the therapeutic potential of MSC EVs has already been proven, a thorough characterization of MSC EVs is lacking. In this work, we used a label-free liquid chromatography tandem mass spectrometry proteomic approach to identify the most abundant proteins in EVs that are secreted from MSCs derived from PSCs (PD-MSCs) and from their parental induced PSCs (iPSCs). Next, we compared both datasets and found that while iPSC EVs enclose proteins that modulate RNA and microRNA stability and protein sorting, PD-MSC EVs are rich in proteins that organize extracellular matrix, regulate locomotion, and influence cell-substrate adhesion. Moreover, compared to their respective cells, iPSCs and iPSC EVs share a greater proportion of proteins, while the PD-MSC proteome appears to be more specific. Correlation and principal component analysis consistently aggregate iPSCs and iPSC EVs but segregate PD-MSC and their EVs. Altogether, these findings suggest that during differentiation, compared with their parental iPSC EVs, PD-MSC EVs acquire a more specific set of proteins; arguably, this difference might confer their therapeutic properties.


Subject(s)
Cell Differentiation , Extracellular Vesicles/metabolism , Mesenchymal Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Proteomics , Cell Line , Extracellular Vesicles/ultrastructure , Humans , Principal Component Analysis , Stromal Cells/metabolism , Tandem Mass Spectrometry , Wharton Jelly/cytology
12.
Sci Rep ; 8(1): 8072, 2018 05 23.
Article in English | MEDLINE | ID: mdl-29795287

ABSTRACT

MicroRNAs are small non-coding RNAs involved in post-transcriptional regulation of gene expression related to many cellular functions. We performed a small-RNAseq analysis of cardiac differentiation from pluripotent stem cells. Our analyses identified some new aspects about microRNA expression in this differentiation process. First, we described a dynamic expression profile of microRNAs where some of them are clustered according to their expression level. Second, we described the extensive network of isomiRs and ADAR modifications. Third, we identified the microRNAs families and clusters involved in the establishment of cardiac lineage and define the mirRNAome based on these groups. Finally, we were able to determine a more accurate miRNAome associated with cardiomyocytes by comparing the expressed microRNAs with other mature cells. MicroRNAs exert their effect in a complex and interconnected way, making necessary a global analysis to better understand their role. Our data expands the knowledge of microRNAs and their implications in cardiomyogenesis.


Subject(s)
Biomarkers/metabolism , Cell Lineage/genetics , Gene Expression Regulation , Mesoderm/metabolism , MicroRNAs/genetics , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Cell Differentiation , Cells, Cultured , Genome, Human , High-Throughput Nucleotide Sequencing , Humans , Mesoderm/cytology , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology
13.
Stem Cell Rev Rep ; 13(4): 491-498, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28214945

ABSTRACT

Intercellular communication is one of the most important events in cell population behavior. In the last decade, tunneling nanotubes (TNTs) have been recognized as a new form of long distance intercellular connection. TNT function is to allow molecular and subcellular structure exchange between neighboring cells via the transfer of molecules and organelles such as calcium ions, prions, viral and bacterial pathogens, small lysosomes and mitochondria. New findings support the concept that mesenchymal stem cells (MSCs) can affect cell microenvironment by the release of soluble factors or the transfer of cellular components to neighboring cells, in a way which significantly contributes to cell regulation and tissue repair, although the underlying mechanisms remain poorly understood. MSCs have many advantages for their implementation in regenerative medicine. The TNTs in these cell types are heterogeneous in both structure and function, probably due to their highly dynamic behavior. In this work we report an extensive and detailed description of types, structure, components, dynamics and functionality of the TNTs bridging neighboring human umbilical cord MSCs obtained from Wharton"s jelly. Characterization studies were carried out through phase contrast, fluorescence, electron microscopy and time lapse images with the aim of describing cells suitable for an eventual regenerative medicine.


Subject(s)
Cell Communication , Mesenchymal Stem Cells/metabolism , Nanotubes/chemistry , Humans , Mesenchymal Stem Cells/cytology
14.
Stem Cell Rev Rep ; 13(1): 68-78, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27815690

ABSTRACT

Mesenchymal stem cells (MSC) have been extensively studied over the past years for the treatment of different diseases. Most of the ongoing clinical trials currently involve the use of MSC derived from adult tissues. This source may have some limitations, particularly with therapies that may require extensive and repetitive cell dosage. However, nowadays, there is a staggering growth in literature on a new source of MSC. There is now increasing evidence about the mesenchymal differentiation from pluripotent stem cell (PSC). Here, we summarize the current knowledge of pluripotent-derived mesenchymal stem cells (PD-MSC). We present a historical perspective on the subject, and then discuss some critical questions that remain unanswered.


Subject(s)
Cell Differentiation , Cell Proliferation , Mesenchymal Stem Cells/cytology , Pluripotent Stem Cells/cytology , Adult , Animals , Embryonic Stem Cells/cytology , Epithelial-Mesenchymal Transition , Humans , Induced Pluripotent Stem Cells/cytology
15.
Elife ; 52016 10 14.
Article in English | MEDLINE | ID: mdl-27740454

ABSTRACT

The levels of the cyclin-dependent kinase (CDK) inhibitor p21 are low in S phase and insufficient to inhibit CDKs. We show here that endogenous p21, instead of being residual, it is functional and necessary to preserve the genomic stability of unstressed cells. p21depletion slows down nascent DNA elongation, triggers permanent replication defects and promotes the instability of hard-to-replicate genomic regions, namely common fragile sites (CFS). The p21's PCNA interacting region (PIR), and not its CDK binding domain, is needed to prevent the replication defects and the genomic instability caused by p21 depletion. The alternative polymerase kappa is accountable for such defects as they were not observed after simultaneous depletion of both p21 and polymerase kappa. Hence, in CDK-independent manner, endogenous p21 prevents a type of genomic instability which is not triggered by endogenous DNA lesions but by a dysregulation in the DNA polymerase choice during genomic DNA synthesis.


Subject(s)
Cell Division , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Replication , DNA/biosynthesis , Genomic Instability , Cells, Cultured , Humans
16.
PLoS One ; 11(10): e0164049, 2016.
Article in English | MEDLINE | ID: mdl-27732616

ABSTRACT

The demand for equine cloning as a tool to preserve high genetic value is growing worldwide; however, nuclear transfer efficiency is still very low. To address this issue, we first evaluated the effects of time from cell fusion to activation (<1h, n = 1261; 1-2h, n = 1773; 2-3h, n = 1647) on in vitro and in vivo development of equine embryos generated by cloning. Then, we evaluated the effects of using different nuclear donor cell types in two successive experiments: I) induced pluripotent stem cells (iPSCs) vs. adult fibroblasts (AF) fused to ooplasts injected with the pluripotency-inducing genes OCT4, SOX2, MYC and KLF4, vs. AF alone as controls; II) umbilical cord-derived mesenchymal stromal cells (UC-MSCs) vs. fetal fibroblasts derived from an unborn cloned foetus (FF) vs. AF from the original individual. In the first experiment, both blastocyst production and pregnancy rates were higher in the 2-3h group (11.5% and 9.5%, respectively), respect to <1h (5.2% and 2%, respectively) and 1-2h (5.6% and 4.7%, respectively) groups (P<0.05). However, percentages of born foals/pregnancies were similar when intervals of 2-3h (35.2%) or 1-2h (35.7%) were used. In contrast to AF, the iPSCs did not generate any blastocyst-stage embryos. Moreover, injection of oocytes with the pluripotency-inducing genes did not improve blastocyst production nor pregnancy rates respect to AF controls. Finally, higher blastocyst production was obtained using UC-MSC (15.6%) than using FF (8.9%) or AF (9.3%), (P<0.05). Despite pregnancy rates were similar for these 3 groups (17.6%, 18.2% and 22%, respectively), viable foals (two) were obtained only by using FF. In summary, optimum blastocyst production rates can be obtained using a 2-3h interval between cell fusion and activation as well as using UC-MSCs as nuclear donors. Moreover, FF line can improve the efficiency of an inefficient AF line. Overall, 24 healthy foals were obtained from a total of 29 born foals.


Subject(s)
Cell Nucleus/physiology , Fetus/cytology , Fibroblasts/cytology , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Animals , Blastocyst/cytology , Cell Differentiation , Cell Lineage , Cells, Cultured , Embryo Transfer , Female , Horses , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Mesenchymal Stem Cells/cytology , Microinjections , Nuclear Transfer Techniques , Oocytes/cytology , Plasmids/genetics , Plasmids/metabolism , Pregnancy , Transcription Factors/genetics , Transcription Factors/metabolism , Umbilical Cord/cytology
17.
Sci Rep ; 6: 35660, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27762303

ABSTRACT

Human embryonic and induced pluripotent stem cells are self-renewing pluripotent stem cells (PSC) that can differentiate into a wide range of specialized cells. Basic fibroblast growth factor is essential for PSC survival, stemness and self-renewal. PI3K/AKT pathway regulates cell viability and apoptosis in many cell types. Although it has been demonstrated that PI3K/AKT activation by bFGF is relevant for PSC stemness maintenance its role on PSC survival remains elusive. In this study we explored the molecular mechanisms involved in the regulation of PSC survival by AKT. We found that inhibition of AKT with three non-structurally related inhibitors (GSK690693, AKT inhibitor VIII and AKT inhibitor IV) decreased cell viability and induced apoptosis. We observed a rapid increase in phosphatidylserine translocation and in the extent of DNA fragmentation after inhibitors addition. Moreover, abrogation of AKT activity led to Caspase-9, Caspase-3, and PARP cleavage. Importantly, we demonstrated by pharmacological inhibition and siRNA knockdown that GSK3ß signaling is responsible, at least in part, of the apoptosis triggered by AKT inhibition. Moreover, GSK3ß inhibition decreases basal apoptosis rate and promotes PSC proliferation. In conclusion, we demonstrated that AKT activation prevents apoptosis, partly through inhibition of GSK3ß, and thus results relevant for PSC survival.


Subject(s)
Glycogen Synthase Kinase 3 beta/metabolism , Oncogene Protein v-akt/metabolism , Pluripotent Stem Cells/physiology , Signal Transduction , Cell Survival , Cells, Cultured , Gene Expression Regulation , Humans
18.
Stem Cell Res ; 16(2): 300-3, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27345989

ABSTRACT

Human foreskin fibroblasts were used to generate the iPSC line iPSC-FH2.1 using the EF1a-hSTEMCCA-loxP vector expressing OCT4, SOX2, c-MYC and KLF4, in 5% O2 culture conditions. Stemness was confirmed, as was pluripotency both in vivo and in vitro, in normoxia and hypoxia. Human Embryonic Stem Cell (hESC) line WA-09 and reprogrammed fibroblast primary culture HFF-FM were used as controls.


Subject(s)
Fibroblasts/cytology , Foreskin/cytology , Induced Pluripotent Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Cellular Reprogramming , Comparative Genomic Hybridization , DNA Methylation , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotype , Kruppel-Like Factor 4 , Male , Microscopy, Fluorescence , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Biochem Biophys Res Commun ; 473(1): 194-199, 2016 Apr 22.
Article in English | MEDLINE | ID: mdl-27012206

ABSTRACT

Addition of methyl groups to arginine residues is catalyzed by a group of enzymes called Protein Arginine Methyltransferases (Prmt). Although Prmt1 is essential in development, its paralogue Prmt8 has been poorly studied. This gene was reported to be expressed in nervous system and involved in neurogenesis. In this work, we found that Prmt8 is expressed in mouse embryonic stem cells (ESC) and in induced pluripotent stem cells, and modulated along differentiation to neural precursor cells. We found that Prmt8 promoter activity is induced by the pluripotency transcription factors Oct4, Sox2 and Nanog. Moreover, endogenous Prmt8 mRNA levels were reduced in ESC transfected with Sox2 shRNA vector. As a whole, our results indicate that Prmt8 is expressed in pluripotent stem cells and its transcription is modulated by pluripotency transcription factors. These findings suggest that besides its known function in nervous system, Prmt8 could play a role in pluripotent stem cells.


Subject(s)
Gene Expression Regulation, Enzymologic , Pluripotent Stem Cells/cytology , Protein-Arginine N-Methyltransferases/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cell Differentiation , Down-Regulation , Fibroblasts/metabolism , Homeodomain Proteins/metabolism , Mice , NIH 3T3 Cells , Nanog Homeobox Protein , Neurons/cytology , Octamer Transcription Factor-3/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
20.
PLoS One ; 10(12): e0144336, 2015.
Article in English | MEDLINE | ID: mdl-26642061

ABSTRACT

Pluripotent stem cells possess complex systems that protect them from oxidative stress and ensure genomic stability, vital for their role in development. Even though it has been reported that antioxidant activity diminishes along stem cell differentiation, little is known about the transcriptional regulation of the involved genes. The reported modulation of some of these genes led us to hypothesize that some of them could be regulated by the transcription factors critical for self-renewal and pluripotency in embryonic stem cells (ESCs) and in induced pluripotent stem cells (iPSCs). In this work, we studied the expression profile of multiple genes involved in antioxidant defense systems in both ESCs and iPSCs. We found that Manganese superoxide dismutase gene (Mn-Sod/Sod2) was repressed during diverse differentiation protocols showing an expression pattern similar to Nanog gene. Moreover, Sod2 promoter activity was induced by Oct4 and Nanog when we performed a transactivation assay using two different reporter constructions. Finally, we studied Sod2 gene regulation by modulating the expression of Oct4 and Nanog in ESCs by shRNAs and found that downregulation of any of them reduced Sod2 expression. Our results indicate that pluripotency transcription factors positively modulate Sod2 gene transcription.


Subject(s)
Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/physiology , Octamer Transcription Factor-3/metabolism , Superoxide Dismutase/genetics , Animals , Cell Differentiation , Cell Line , Embryonic Stem Cells/cytology , Gene Expression Regulation , Gene Expression Regulation, Enzymologic , Homeodomain Proteins/genetics , Induced Pluripotent Stem Cells/cytology , Mice , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Promoter Regions, Genetic , RNA, Small Interfering , Superoxide Dismutase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...