Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Heliyon ; 9(1): e12675, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36685404

ABSTRACT

Background: Imaging-guided access to the brain has become a routine procedure for various research and clinical applications, including drug administration, neurophysiological recording, and sampling tissue. Therefore, open-source software is required to handle such datasets in these specific applications. New methods: Here, we proposed an open-source tool utilizing different imaging modalities for automating the steps to access the brain. This tool provides means for easily calculating the coordination of the area of interest concerning a specific point of entry. The source and documentation are available at this link. Results: We have used this software for three different applications: electrophysiological recording, drug infusion in the nonhuman primate brain, and guided biopsy procedure in the human brain. We performed a neural recording of two monkeys' prefrontal cortex and inferior temporal cortex using this software in submillimeter resolution. We also applied our procedure for infusion in the putamen and caudate nuclei in both hemispheres of another group of rhesus monkeys with histological proof in one animal. More so, we validated this software in the human subjects that underwent biopsy surgery with the commercial software used in human biopsy surgery. Comparison with existing methods: Our software uses different imaging modalities by co-registering them. This will provide structural details of the skull and brain tissue. We can calculate each brain region's coordination at the point of entry by re-slicing the images. Atlas-based image segmentation were implemented in our software. Three mentioned applications of our software in neuroscience will be further discussed in this paper. Conclusion: In our procedure, working with different imaging modalities provides a precise estimation of the specific region in the brain related to the location of implants or stereotaxic frames. There is no limitation to using metal implants in this procedure.

2.
Cell Prolif ; 53(10): e12892, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32918782

ABSTRACT

OBJECTIVES: Genetic engineering of human-induced pluripotent stem cell-derived neural stem cells (hiPSC-NSC) may increase the risk of genomic aberrations. Therefore, we asked whether genetic modification of hiPSC-NSCs exacerbates chromosomal abnormalities that may occur during passaging and whether they may cause any functional perturbations in NSCs in vitro and in vivo. MATERIALS AND METHODS: The transgenic cassette was inserted into the AAVS1 locus, and the genetic integrity of zinc-finger nuclease (ZFN)-modified hiPSC-NSCs was assessed by the SNP-based karyotyping. The hiPSC-NSC proliferation was assessed in vitro by the EdU incorporation assay and in vivo by staining of brain slices with Ki-67 antibody at 2 and 8 weeks after transplantation of ZFN-NSCs with and without chromosomal aberration into the striatum of immunodeficient rats. RESULTS: During early passages, no chromosomal abnormalities were detected in unmodified or ZFN-modified hiPSC-NSCs. However, at higher passages both cell populations acquired duplication of the entire long arm of chromosome 1, dup(1)q. ZNF-NSCs carrying dup(1)q exhibited higher proliferation rate than karyotypically intact cells, which was partly mediated by increased expression of AKT3 located on Chr1q. Compared to karyotypically normal ZNF-NSCs, cells with dup(1)q also exhibited increased proliferation in vivo 2 weeks, but not 2 months, after transplantation. CONCLUSIONS: These results demonstrate that, independently of ZFN-editing, hiPSC-NSCs have a propensity for acquiring dup(1)q and this aberration results in increased proliferation which might compromise downstream hiPSC-NSC applications.


Subject(s)
Chromosomes, Human, Pair 1/genetics , Gene Editing/methods , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Brain/metabolism , Brain/pathology , Cell Differentiation , Cell Proliferation , Cells, Cultured , Gene Duplication , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Karyotype , Neural Stem Cells/cytology , Proto-Oncogene Proteins c-akt/metabolism , Zinc Fingers/genetics
3.
Mol Cell Proteomics ; 17(9): 1670-1684, 2018 09.
Article in English | MEDLINE | ID: mdl-29848781

ABSTRACT

Despite the progress in safety and efficacy of cell replacement therapy with pluripotent stem cells (PSCs), the presence of residual undifferentiated stem cells or proliferating neural progenitor cells with rostral identity remains a major challenge. Here we report the generation of a LIM homeobox transcription factor 1 alpha (LMX1A) knock-in GFP reporter human embryonic stem cell (hESC) line that marks the early dopaminergic progenitors during neural differentiation to find reliable membrane protein markers for isolation of midbrain dopaminergic neurons. Purified GFP positive cells in vitro exhibited expression of mRNA and proteins that characterized and matched the midbrain dopaminergic identity. Further quantitative proteomics analysis of enriched LMX1A+ cells identified several membrane-associated proteins including a polysialylated embryonic form of neural cell adhesion molecule (PSA-NCAM) and contactin 2 (CNTN2), enabling prospective isolation of LMX1A+ progenitor cells. Transplantation of human-PSC-derived purified CNTN2+ progenitors enhanced dopamine release from transplanted cells in the host brain and alleviated Parkinson's disease-related phenotypes in animal models. This study establishes an efficient approach for purification of large numbers of human-PSC-derived dopaminergic progenitors for therapeutic applications.


Subject(s)
Biomarkers/metabolism , Cell Membrane/metabolism , Cell Separation/methods , Dopaminergic Neurons/transplantation , Embryonic Stem Cells/cytology , Parkinson Disease/therapy , Animals , Cell Differentiation , Contactin 2/metabolism , Disease Models, Animal , Embryonic Stem Cells/metabolism , Female , Green Fluorescent Proteins/metabolism , Humans , LIM-Homeodomain Proteins/metabolism , Parkinson Disease/pathology , Proteomics , Rats, Sprague-Dawley , Reproducibility of Results , Transcription Factors/metabolism
4.
Methods ; 133: 21-28, 2018 01 15.
Article in English | MEDLINE | ID: mdl-28939501

ABSTRACT

Recent investigations have demonstrated that defined sets of exogenous factors (chemical and/or biochemical) can convert human and mouse somatic cells into induced neural stem cells (iNSCs). Considering the self-renewal and multi-potential differentiation capabilities of iNSCs, generation of these cells has considerably enhanced cell therapy for treatment of neurodegenerative disorders. These cells can also serve as models for investigation of the mechanism(s) underlying neurodegenerative diseases and as an asset in drug discovery. Meanwhile, using the process of direct conversion/transdifferentiation, by bypassing pluripotent state and consequently reducing tumorigenesis and genetic instability risks, establishment of several desired cells are feasible. In this review, we describe the pros and cons of different methods employed to directly reprogram somatic cells to iNSCs along with the progress of iNSCs applications and the future challenges.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/transplantation , Neural Stem Cells/transplantation , Neurodegenerative Diseases/therapy , Animals , Cell Differentiation/genetics , Cell Transdifferentiation/genetics , Fibroblasts/cytology , Humans , Mice
5.
Cell J ; 19(4): 545-551, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29105388

ABSTRACT

OBJECTIVES: Motor neuron differentiation from human embryonic stem cells (hESCs) is a goal of regenerative medicine to provide cell therapy as treatments for diseases that damage motor neurons. Most protocols lack adequate efficiency in generating functional motor neurons. However, small molecules present a new approach to overcome this challenge. The aim of this research is to replace morphogen factors with a cocktail of efficient, affordable small molecules for effective, low cost motor neuron differentiation. MATERIALS AND METHODS: In this experimental study, hESCs were differentiated into motor neuron by the application of a small molecule cocktail that consisted of dorsomorphin, A8301, and XAV939. During the differentiation protocol, we selected five stages and assessed expressions of neural markers by real-time polymerase chain reaction (PCR), immunofluorescence staining, and flow cytometry. Motor neuron ion currents were determined by whole cell patch clamp recording. RESULTS: Immunofluorescence staining and flow cytometry analysis of hESC-derived neural ectoderm (NE) indicated that they were positive for NESTIN (92.68%), PAX6 (64.40%), and SOX1 (82.11%) in a chemically defined adherent culture. The replated (hESC)-derived NE differentiated cells were positive for TUJ1, MAP2, HB9 and ISL1. We evaluated the gene expression levels with real-time reverse transcriptase-PCR at different stages of the differentiation protocol. Voltage gated channel currents of differentiated cells were examined by the whole-cell patch clamp technique. The hESC-derived motor neurons showed voltage gated delay rectifier K+, Na+ and Ca2+ inward currents. CONCLUSIONS: Our results indicated that hESC-derived neurons expressed the specific motor neuron markers specially HB9 and ISL1 but voltage clamp recording showed small ionic currents therefore it seems that voltage gated channel population were inadequate for firing action potentials.

6.
Biochem Biophys Res Commun ; 491(1): 166-172, 2017 09 09.
Article in English | MEDLINE | ID: mdl-28711494

ABSTRACT

OBJECTIVE: For human embryonic stem cells (hESCs) to differentiate into neurons, enormous changes has to occur leading to trigger action potential and neurotransmitter release. We attempt to determine the changes in expression of voltage gated channels (VGCs) and their electrophysiological properties during neural differentiation. MATERIALS AND METHODS: The relative expressions of α-subunit of voltage gated potassium, sodium and calcium channels were characterized by qRT-PCR technique. Patch clamp recording was performed to characterize the electrophysiological properties of hESCs during their differentiation into neuron-like cells. RESULTS: Relative expression of α-subunit of channels changed significantly. 4-AP and TEA sensitive outward currents were observed in all stages, although TEA sensitive currents were recorded once in rosette structure. Nifedipine and QX314 sensitive inward currents were recorded only in neuron-like cells. CONCLUSION: K+ currents were recorded in hESCs and rosette structure cells. Inward currents, sensitive to Nifedipine and QX314, were recorded in neuron-like cells.


Subject(s)
Cell Differentiation/physiology , Embryonic Development/physiology , Human Embryonic Stem Cells/physiology , Ion Channels/physiology , Neural Stem Cells/physiology , Neurogenesis/physiology , Calcium/metabolism , Cells, Cultured , Human Embryonic Stem Cells/cytology , Humans , Ion Channel Gating/physiology , Membrane Potentials/physiology , Neural Stem Cells/cytology , Potassium/metabolism , Sodium/metabolism
7.
Stem Cell Reports ; 6(4): 539-551, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27052315

ABSTRACT

Direct conversion of somatic cells into neural stem cells (NSCs) by defined factors holds great promise for mechanistic studies, drug screening, and potential cell therapies for different neurodegenerative diseases. Here, we report that a single zinc-finger transcription factor, Zfp521, is sufficient for direct conversion of human fibroblasts into long-term self-renewable and multipotent NSCs. In vitro, Zfp521-induced NSCs maintained their characteristics in the absence of exogenous factor expression and exhibited morphological, molecular, developmental, and functional properties that were similar to control NSCs. In addition, the single-seeded induced NSCs were able to form NSC colonies with efficiency comparable with control NSCs and expressed NSC markers. The converted cells were capable of surviving, migrating, and attaining neural phenotypes after transplantation into neonatal mouse and adult rat brains, without forming tumors. Moreover, the Zfp521-induced NSCs predominantly expressed rostral genes. Our results suggest a facilitated approach for establishing human NSCs through Zfp521-driven conversion of fibroblasts.


Subject(s)
Cell Self Renewal/genetics , Fibroblasts/metabolism , Multipotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Transcription Factors/metabolism , 3T3 Cells , Animals , Animals, Newborn , Cell Survival/genetics , Cells, Cultured , Fibroblasts/cytology , Foreskin/cytology , Gene Expression Profiling/methods , Humans , Infant, Newborn , Male , Mice , Microscopy, Fluorescence , Multipotent Stem Cells/cytology , Multipotent Stem Cells/transplantation , Neural Stem Cells/cytology , Neural Stem Cells/transplantation , Rats, Nude , Stem Cell Transplantation/methods , Transcription Factors/genetics , Transfection , Transplantation, Heterologous
8.
PLoS One ; 10(8): e0135479, 2015.
Article in English | MEDLINE | ID: mdl-26266943

ABSTRACT

BACKGROUND: A number of studies generated induced neural progenitor cells (iNPCs) from human fibroblasts by viral delivering defined transcription factors. However, the potential risks associated with gene delivery systems have limited their clinical use. We propose it would be safer to induce neural progenitor-like cells from human adult fibroblasts via a direct non-genetic alternative approach. METHODOLOGY/PRINCIPAL FINDINGS: Here, we have reported that seven rounds of TAT-SOX2 protein transduction in a defined chemical cocktail under a 3D sphere culture gradually morphed fibroblasts into neuroepithelial-like colonies. We were able to expand these cells for up to 20 passages. These cells could give rise to cells that expressed neurons and glia cell markers both in vitro and in vivo. CONCLUSIONS/SIGNIFICANCE: These results show that our approach is beneficial for the genetic material-free generation of iNPCs from human fibroblasts where small chemical molecules can provide a valuable, viable strategy to boost and improve induction in a 3D sphere culture.


Subject(s)
Fibroblasts/cytology , Neural Stem Cells/cytology , Adult , Cell Differentiation/physiology , Cells, Cultured , Fibroblasts/metabolism , Humans , Immunohistochemistry , Neural Stem Cells/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
9.
Stem Cells Dev ; 21(12): 2262-72, 2012 Aug 10.
Article in English | MEDLINE | ID: mdl-22145677

ABSTRACT

We describe a new, efficient protocol that involves the serial addition of noggin, basic fibroblast growth factor (bFGF), retinoic acid, and sonic hedgehog (Shh) for the differentiation of human induced pluripotent stem cells (hiPSC) to retinal pigmented epithelium (RPE) in a serum- and feeder-free adherent condition. hiPSC-RPE cells exhibited RPE morphology and specific molecular markers. Additionally, several hiPSC lines were generated from retinal-specific patients with Leber's congenital amaurosis, Usher syndrome, two patients with retinitis pigmentosa, and a patient with Leber's hereditary optic neuropathy. The RPE cells generated from these disease-specific hiPSCs expressed specific markers by the same RPE lineage-directed differentiation protocol. These findings indicate a new short-term, simple, and efficient protocol for differentiation of hiPSCs to RPE cells. Such specific retinal disease-specific hiPSCs offer an unprecedented opportunity to recapitulate normal and pathologic formation of human retinal cells in vitro, thereby enabling pharmaceutical screening, and potentially autologous cell replacement therapies for retinal diseases.


Subject(s)
Cell Differentiation , Induced Pluripotent Stem Cells/physiology , Retinal Diseases/pathology , Retinal Pigment Epithelium/pathology , Adolescent , Adult , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Culture Techniques , Cell Shape , Cells, Cultured , Child , Female , Gene Expression , Humans , Induced Pluripotent Stem Cells/metabolism , Karyotype , Male , Middle Aged , Phenotype , Real-Time Polymerase Chain Reaction , Retinal Pigment Epithelium/metabolism
10.
Stem Cells ; 29(12): 1933-41, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21997905

ABSTRACT

A major goal of regenerative medicine is to produce cells to participate in the generation, maintenance, and repair of tissues that are damaged by disease, aging, or trauma, such that function is restored. The establishment of induced pluripotent stem cells, followed by directed differentiation, offers a powerful strategy for producing patient-specific therapies. Given how laborious and lengthy this process can be, the conversion of somatic cells into lineage-specific stem/progenitor cells in one step, without going back to, or through, a pluripotent stage, has opened up tremendous opportunities for regenerative medicine. However, there are a number of obstacles to overcome before these cells can be widely considered for clinical applications. Here, we focus on induced transdifferentiation strategies to convert mature somatic cells to other mature cell types or progenitors, and we summarize the challenges that need to be met if the potential applications of transdifferentiation technology are to be achieved.


Subject(s)
Cell Transdifferentiation , Epigenesis, Genetic , Induced Pluripotent Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/immunology , Humans , Induced Pluripotent Stem Cells/immunology , Mice , Regenerative Medicine , Stem Cell Niche , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Tissue Eng Part A ; 17(23-24): 3021-31, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21740132

ABSTRACT

In this study, the impact of randomly oriented electrospun polyamide nanofibrous architecture on neurogenic differentiation of human embryonic stem cells (hESCs) compared with the lack of nanofibrous features in vitro in a neural-inducing condition was examined. Flow cytometry analysis of hESC-derived neural ectoderm (NE) showed nanofibrous surfaces capable of supporting NE by expression of higher percentages of related markers NESTIN, SOX1, and PAX6 in addition to significantly greater total cell proliferation as shown by Ki67 in the neurogenic condition. After replating hESC-derived NE, the differentiated cells expressed higher neuronal markers (TUJ1 and MAP2) and motor neuron markers (HB9, ISL1, and ChAT) at both the protein and mRNA levels on nanofibers. The presence of developed spread neurites and plausible neurite connections were shown by scanning electron microscopy. Additionally, Na(+) and Ca(2+) currents in differentiated neurons on nanofibers were significantly greater than both control and generated action potentials. Moreover, less duration of inward currents, greater negative resting membrane potential, and enhanced expression and functionality of ionic channel genes were observed in neuronal cells on nanofibers. These results indicated that a nanofibrillar surface along with neurogenic growth factors provided a better environment for hESC neurogenic differentiation and function, which holds great promise in prospective tissue engineering applications.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Nanofibers/chemistry , Neurons/cytology , Tissue Engineering/methods , Cell Line , Ectoderm/cytology , Electrophysiological Phenomena , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/ultrastructure , Gene Expression Profiling , Gene Expression Regulation , Humans , Ion Channels/genetics , Ion Channels/metabolism , Nanofibers/ultrastructure , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Tube/growth & development , Neurons/metabolism , Neurons/ultrastructure , Surface Properties
12.
In Vitro Cell Dev Biol Anim ; 46(3-4): 395-402, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20177993

ABSTRACT

In this study, we focused on the derivation of human embryonic stem cell (hESC) from preimplantation genetic screening (PGS)-analyzed and preimplantation genetic diagnosis (PGD)-analyzed embryos. Out of 62 fresh PGD/PGS-analyzed embryos, 22 embryos reached the blastocyst stage. From 12 outgrowth blastocysts, we derived four hESC lines onto a feeder layer. Surprisingly, karyotype analysis showed that hESC lines derived from aneuploid embryos had diploid female karyotype. One hESC line was found to carry a balanced Robertsonian translocation. All the cell lines showed hESC markers and had the pluripotent ability to differentiate into derivatives of the three embryonic germ layers. The established lines had clonal propagation with 22-31% efficiency in the presence of ROCK inhibitor. These results further indicate that hESC lines can be derived from PGD/PGS-analyzed embryos that are destined to be discarded and can serve as an alternative source for normal euploid lines.


Subject(s)
Cell Culture Techniques/methods , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Preimplantation Diagnosis/methods , Animals , Blastomeres/cytology , Cell Differentiation , Cell Line , Colony-Forming Units Assay , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Mice
13.
Biochem Biophys Res Commun ; 391(1): 329-34, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19912985

ABSTRACT

Bombay phenotype is one of the rare phenotypes in the ABO blood group system that fails to express ABH antigens on red blood cells. Nonsense or missense mutations in fucosyltransfrase1 (FUT1) and fucosyltransfrase2 (FUT2) genes are known to create this phenotype. This blood group is compatible with all other blood groups as a donor, as it does not express the H antigen on the red blood cells. In this study, we describe the establishment of human induced pluripotent stem cells (iPSCs) from the dermal fibroblasts of a Bombay blood-type individual by the ectopic expression of established transcription factors Klf4, Oct4, Sox2, and c-Myc. Sequence analyses of fibroblasts and iPSCs revealed a nonsense mutation 826C to T (276 Gln to Ter) in the FUT1 gene and a missense mutation 739G to A (247 Gly to Ser) in the FUT2 gene in the Bombay phenotype under study. The established iPSCs resemble human embryonic stem cells in morphology, passaging, surface and pluripotency markers, normal karyotype, gene expression, DNA methylation of critical pluripotency genes, and in-vitro differentiation. The directed differentiation of the iPSCs into hematopoietic lineage cells displayed increased expression of the hematopoietic lineage markers such as CD34, CD133, RUNX1, KDR, alpha-globulin, and gamma-globulin. Such specific stem cells provide an unprecedented opportunity to produce a universal blood group donor, in-vitro, thus enabling cellular replacement therapies, once the safety issue is resolved.


Subject(s)
ABO Blood-Group System , Cell Line , Erythrocytes/cytology , Hematopoiesis , Pluripotent Stem Cells/cytology , Amino Acid Sequence , Base Sequence , Blood Donors , Fibroblasts/cytology , Fibroblasts/metabolism , Fucosyltransferases/genetics , Gene Expression , Humans , Kruppel-Like Factor 4 , Mutation, Missense , Phenotype , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Galactoside 2-alpha-L-fucosyltransferase
14.
Stem Cells Dev ; 19(6): 809-18, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19788394

ABSTRACT

To evaluate the effect of dehydroepiandrosterone (DHEA) as a neurosteroid on the rate of neurogenesis, neural survival, and proliferation of pluripotent stem cell-derived neurons, we have added DHEA to mouse P19 embryonal carcinoma cell- and human embryonic stem cell-derived neural progenitors (ECC- and ESC-NPs). In ECC-derived NPs, flow cytometric analysis of nestin and Tuj1-positive cells revealed that the percentages of these cells increased significantly for the markers following DHEA treatment of the cells. Moreover, the percentages of tyrosine hydroxylase (TH)-positive cells, the marker of dopaminergic neurons, significantly increased in the presence of DHEA. The expression of neural-specific genes such as Mash1, Pax6, Tuj1, and TH was also detected by RT-PCR analysis. BrdU incorporation and estrogen receptor (EsR) were found to be increased after DHEA induction. Moreover, apoptosis was significantly decreased after DHEA treatment. DHEA effect was also confirmed on human ESC-NPs by the enhancement of Tuj1- and TH-immunofluorescent-positive cells and TH and Nurr1 transcripts, as detected by quantitative RT-PCR. In conclusion, these results have presented evidence that DHEA was able to induce neurogenesis in mouse ECC and human ESC-NPs. This observation was related to the division of NPs and the reduction of apoptosis. Moreover, DHEA has dopaminergic potential in the cells of both orders. This provides a better insight into the differentiation and maintenance of neural cells and treatment of a wide variety of neurological diseases such as Alzheimer's and Parkinson's by stem cells.


Subject(s)
Dehydroepiandrosterone/pharmacology , Dopamine/metabolism , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/drug effects , Neurogenesis/drug effects , Neurons/cytology , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Developmental/drug effects , Humans , Mice , Neurogenesis/genetics , Neurons/drug effects , Neurons/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...