Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Article in English | MEDLINE | ID: mdl-34035170

ABSTRACT

Heterozygous NRXN1 deletions constitute the most prevalent currently known single-gene mutation associated with schizophrenia, and additionally predispose to multiple other neurodevelopmental disorders. Engineered heterozygous NRXN1 deletions impaired neurotransmitter release in human neurons, suggesting a synaptic pathophysiological mechanism. Utilizing this observation for drug discovery, however, requires confidence in its robustness and validity. Here, we describe a multicenter effort to test the generality of this pivotal observation, using independent analyses at two laboratories of patient-derived and newly engineered human neurons with heterozygous NRXN1 deletions. Using neurons transdifferentiated from induced pluripotent stem cells that were derived from schizophrenia patients carrying heterozygous NRXN1 deletions, we observed the same synaptic impairment as in engineered NRXN1-deficient neurons. This impairment manifested as a large decrease in spontaneous synaptic events, in evoked synaptic responses, and in synaptic paired-pulse depression. Nrxn1-deficient mouse neurons generated from embryonic stem cells by the same method as human neurons did not exhibit impaired neurotransmitter release, suggesting a human-specific phenotype. Human NRXN1 deletions produced a reproducible increase in the levels of CASK, an intracellular NRXN1-binding protein, and were associated with characteristic gene-expression changes. Thus, heterozygous NRXN1 deletions robustly impair synaptic function in human neurons regardless of genetic background, enabling future drug discovery efforts.


Subject(s)
Calcium-Binding Proteins/genetics , Mutation , Neural Cell Adhesion Molecules/genetics , Neurons/metabolism , Neurotransmitter Agents/metabolism , Schizophrenia/metabolism , Case-Control Studies , Cell Transdifferentiation , Cells, Cultured , Cohort Studies , Embryonic Stem Cells/cytology , Gene Expression , Guanylate Kinases/metabolism , Heterozygote , Humans , Induced Pluripotent Stem Cells/cytology
2.
Mol Cell Neurosci ; 109: 103562, 2020 12.
Article in English | MEDLINE | ID: mdl-32987141

ABSTRACT

Abnormal dendritic arbor development has been implicated in a number of neurodevelopmental disorders, such as autism and Rett syndrome, and the neuropsychiatric disorder schizophrenia. Postmortem brain samples from subjects with schizophrenia show elevated levels of NOS1AP in the dorsolateral prefrontal cortex, a region of the brain associated with cognitive function. We previously reported that the long isoform of NOS1AP (NOS1AP-L), but not the short isoform (NOS1AP-S), negatively regulates dendrite branching in rat hippocampal neurons. To investigate the role that NOS1AP isoforms play in human dendritic arbor development, we adapted methods to generate human neural progenitor cells and neurons using induced pluripotent stem cell (iPSC) technology. We found that increased protein levels of either NOS1AP-L or NOS1AP-S decrease dendrite branching in human neurons at the developmental time point when primary and secondary branching actively occurs. Next, we tested whether pharmacological agents can decrease the expression of NOS1AP isoforms. Treatment of human iPSC-derived neurons with d-serine, but not clozapine, haloperidol, fluphenazine, or GLYX-13, results in a reduction in endogenous NOS1AP-L, but not NOS1AP-S, protein expression; however, d-serine treatment does not reverse decreases in dendrite number mediated by overexpression of NOS1AP isoforms. In summary, we demonstrate how an in vitro model of human neuronal development can help in understanding the etiology of schizophrenia and can also be used as a platform to screen drugs for patients.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Dendrites/ultrastructure , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neurons/cytology , Adaptor Proteins, Signal Transducing/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Cells, Cultured , Clozapine/pharmacology , Drug Evaluation, Preclinical , Fluphenazine/pharmacology , Gene Expression Regulation/drug effects , Glutamic Acid/physiology , Haloperidol/pharmacology , Humans , Induced Pluripotent Stem Cells/metabolism , Ion Channels/physiology , Nerve Tissue Proteins/physiology , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/metabolism , Oligopeptides/pharmacology , Patch-Clamp Techniques , Protein Isoforms/physiology , Schizophrenia/etiology , Schizophrenia/genetics , Serine/pharmacology
3.
Stem Cell Res ; 47: 101926, 2020 Jul 25.
Article in English | MEDLINE | ID: mdl-32738631

ABSTRACT

We have generated two iPSC lines from skin biopsies of two healthy individuals. Skin fibroblasts were derived and reprogrammed using a Sendai virus-based approach. The resulting iPSC lines have normal karyotype, express stemness markers and can generate endoderm, mesoderm and ectoderm in vitro. These iPSC lines can be used as healthy controls in differentiation paradigms as well as backbone for gene editing experiments.

4.
Neurology ; 94(12): 526-537, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32098856

ABSTRACT

OBJECTIVE: Dystonia is a complex movement disorder. Research progress has been difficult, particularly in developing widely effective therapies. This is a review of the current state of knowledge, research gaps, and proposed research priorities. METHODS: The NIH convened leaders in the field for a 2-day workshop. The participants addressed the natural history of the disease, the underlying etiology, the pathophysiology, relevant research technologies, research resources, and therapeutic approaches and attempted to prioritize dystonia research recommendations. RESULTS: The heterogeneity of dystonia poses challenges to research and therapy development. Much can be learned from specific genetic subtypes, and the disorder can be conceptualized along clinical, etiology, and pathophysiology axes. Advances in research technology and pooled resources can accelerate progress. Although etiologically based therapies would be optimal, a focus on circuit abnormalities can provide a convergent common target for symptomatic therapies across dystonia subtypes. The discussions have been integrated into a comprehensive review of all aspects of dystonia. CONCLUSION: Overall research priorities include the generation and integration of high-quality phenotypic and genotypic data, reproducing key features in cellular and animal models, both of basic cellular mechanisms and phenotypes, leveraging new research technologies, and targeting circuit-level dysfunction with therapeutic interventions. Collaboration is necessary both for collection of large data sets and integration of different research methods.


Subject(s)
Dystonic Disorders , Neurology/trends , Research/trends , Animals , Dystonia , Humans
5.
Mol Psychiatry ; 25(7): 1406-1419, 2020 07.
Article in English | MEDLINE | ID: mdl-31481756

ABSTRACT

The OPRM1 A118G single nucleotide polymorphism (SNP rs1799971) gene variant encoding the N40D µ-opioid receptor (MOR) has been associated with dependence on opiates and other drugs of abuse but its mechanism is unknown. The frequency of G-allele carriers is ~40% in Asians, ~16% in Europeans, and ~3% in African-Americans. With opioid abuse-related deaths rising at unprecedented rates, understanding these mechanisms may provide a path to therapy. Here we generated homozygous N40D subject-specific induced inhibitory neuronal cells (iNs) from seven human-induced pluripotent stem (iPS) cell lines from subjects of European descent (both male and female) and probed the impact of N40D MOR regulation on synaptic transmission. We found that D40 iNs exhibit consistently stronger suppression (versus N40) of spontaneous inhibitory postsynaptic currents (sIPSCs) across multiple subjects. To mitigate the confounding effects of background genetic variation on neuronal function, the regulatory effects of MORs on synaptic transmission were recapitulated in two sets of independently engineered isogenic N40D iNs. In addition, we employed biochemical analysis and observed differential N-linked glycosylation of human MOR N40D. This study identifies neurophysiological and molecular differences between human MOR variants that may predict altered opioid responsivity and/or dependence in this subset of individuals.


Subject(s)
Neurons/metabolism , Opioid-Related Disorders/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Synapses/metabolism , Cell Line , Female , Humans , Inhibitory Postsynaptic Potentials , Male , Neurons/cytology , Receptors, Opioid, mu/chemistry
6.
Article in English | MEDLINE | ID: mdl-30065644

ABSTRACT

Non-coding RNA, including microRNA (miRNA) serves critical regulatory functions in the developing brain. The let-7 family of miRNAs has been shown to regulate neuronal differentiation, neural subtype specification, and synapse formation in animal models. However, the regulatory role of human let-7c (hsa-let-7c) in human neuronal development has yet to be examined. Let-7c is encoded on chromosome 21 in humans and therefore may be overexpressed in human brains in Trisomy 21 (T21), a complex neurodevelopmental disorder. Here, we employ recent developments in stem cell biology to show that hsa-let-7c mediates important regulatory epigenetic functions that control the development and functional activity of human induced neuronal cells (iNs). We show that overexpression of hsa-let-7c in human iNs derived from induced pluripotent stem (iPS), as well as embryonic stem (ES), cells leads to morphological as well as functional deficits including impaired neuronal morphologic development, synapse formation and synaptic strength, as well as a marked reduction of neuronal excitability. Importantly, we have assessed these findings over three independent genetic backgrounds, showing that some of these effects are subject to influence by background genetic variability with the most robust and reproducible effect being a striking reduction in spontaneous neural firing. Collectively, these results suggest an important function for let-7 family miRNAs in regulation of human neuronal development and raise implications for understanding the complex molecular etiology of neurodevelopmental disorders, such as T21, where let-7c gene dosage is increased.

7.
Sci Rep ; 6: 34341, 2016 10 04.
Article in English | MEDLINE | ID: mdl-27698409

ABSTRACT

Genetic variation in nicotinic receptor alpha 5 (CHRNA5) has been associated with increased risk of addiction-associated phenotypes in humans yet little is known the underlying neural basis. Induced pluripotent stem cells (iPSCs) were derived from donors homozygous for either the major (D398) or the minor (N398) allele of the nonsynonymous single nucleotide polymorphism (SNP), rs16969968, in CHRNA5. To understand the impact of these nicotinic receptor variants in humans, we differentiated these iPSCs to dopamine (DA) or glutamatergic neurons and then tested their functional properties and response to nicotine. Results show that N398 variant human DA neurons differentially express genes associated with ligand receptor interaction and synaptic function. While both variants exhibited physiological properties consistent with mature neuronal function, the N398 neuronal population responded more actively with an increased excitatory postsynaptic current response upon the application of nicotine in both DA and glutamatergic neurons. Glutamatergic N398 neurons responded to lower nicotine doses (0.1 µM) with greater frequency and amplitude but they also exhibited rapid desensitization, consistent with previous analyses of N398-associated nicotinic receptor function. This study offers a proof-of-principle for utilizing human neurons to study gene variants contribution to addiction.


Subject(s)
Alleles , Induced Pluripotent Stem Cells/drug effects , Nerve Tissue Proteins/genetics , Neurons/drug effects , Nicotine/pharmacology , Receptors, Nicotinic/genetics , Cells, Cultured , Gene Expression Profiling , Genetic Variation , Glutamic Acid/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism
8.
Mol Brain ; 9(1): 51, 2016 05 10.
Article in English | MEDLINE | ID: mdl-27160314

ABSTRACT

BACKGROUND: Alcohol abuse produces an enormous impact on health, society, and the economy. Currently, there are very limited therapies available, largely due to the poor understanding of mechanisms underlying alcohol use disorders (AUDs) in humans. Oxidative damage of mitochondria and cellular proteins aggravates the progression of neuroinflammation and neurological disorders initiated by alcohol abuse. RESULTS: Here we show that ethanol exposure causes neuroinflammation in both human induced pluripotent stem (iPS) cells and human neural progenitor cells (NPCs). Ethanol exposure for 24 hours or 7 days does not affect the proliferation of iPS cells and NPCs, but primes an innate immune-like response by activating the NLR family pyrin domain containing 3 (NLRP3) inflammasome pathway. This leads to an increase of microtubule-associated protein 1A/1B-light chain 3(+) (LC3B(+)) autophagic puncta and impairment of the mitochondrial and lysosomal distribution. In addition, a decrease of mature neurons derived from differentiating NPCs is evident in ethanol pre-exposed compared to control NPCs. Moreover, a second insult of a pro-inflammatory factor in addition to ethanol preexposure enhances innate cellular inflammation in human iPS cells. CONCLUSIONS: This study provides strong evidence that neuronal inflammation contributes to the pathophysiology of AUDs through the activation of the inflammasome pathway in human cellular models.


Subject(s)
Ethanol/pharmacology , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neural Stem Cells/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Cell Count , Cell Differentiation/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Peroxides/pharmacology
9.
Nat Commun ; 7: 10862, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26983594

ABSTRACT

Cell replacement therapy with human pluripotent stem cell-derived neurons has the potential to ameliorate neurodegenerative dysfunction and central nervous system injuries, but reprogrammed neurons are dissociated and spatially disorganized during transplantation, rendering poor cell survival, functionality and engraftment in vivo. Here, we present the design of three-dimensional (3D) microtopographic scaffolds, using tunable electrospun microfibrous polymeric substrates that promote in situ stem cell neuronal reprogramming, neural network establishment and support neuronal engraftment into the brain. Scaffold-supported, reprogrammed neuronal networks were successfully grafted into organotypic hippocampal brain slices, showing an ∼ 3.5-fold improvement in neurite outgrowth and increased action potential firing relative to injected isolated cells. Transplantation of scaffold-supported neuronal networks into mouse brain striatum improved survival ∼ 38-fold at the injection site relative to injected isolated cells, and allowed delivery of multiple neuronal subtypes. Thus, 3D microscale biomaterials represent a promising platform for the transplantation of therapeutic human neurons with broad neuro-regenerative relevance.


Subject(s)
Brain/cytology , Cellular Reprogramming , Imaging, Three-Dimensional , Neurons/cytology , Neurons/transplantation , Tissue Scaffolds/chemistry , Cell Differentiation , Cell Proliferation , Cell Survival , Humans , Polymers/chemistry , Transcription Factors/metabolism
10.
Stem Cell Reports ; 5(6): 1097-1108, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26677768

ABSTRACT

A spontaneously reverted iPSC line was identified from an A-T subject with heterozygous ATM truncation mutations. The reverted iPSC line expressed ATM protein and was capable of radiation-induced phosphorylation of CHK2 and H2A.X. Genome-wide SNP analysis confirmed a match to source T cells and also to a distinct, non-reverted iPSC line from the same subject. Rearranged T cell receptor sequences predict that the iPSC culture originated as several independently reprogrammed cells that resolved into a single major clone, suggesting that gene correction likely occurred early in the reprogramming process. Gene expression analysis comparing ATM(-/-) iPSC lines to unrelated ATM(+/-) cells identifies a large number of differences, but comparing only the isogenic pair of A-T iPSC lines reveals that the primary pathway affected by loss of ATM is a diminished expression of p53-related mRNAs. Gene reversion in culture, although likely a rare event, provided a novel, reverted cell line for studying ATM function.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , Induced Pluripotent Stem Cells/cytology , Base Sequence , Cell Line , Cellular Reprogramming , Gene Deletion , Heterozygote , Humans , Induced Pluripotent Stem Cells/metabolism , Mutation , Polymorphism, Single Nucleotide
11.
Schizophr Bull ; 41(1): 123-32, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24622295

ABSTRACT

BACKGROUND: Herpes simplex virus, type 1 (HSV-1) commonly produces lytic mucosal lesions. It invariably initiates latent infection in sensory ganglia enabling persistent, lifelong infection. Acute HSV-1 encephalitis is rare and definitive evidence of latent infection in the brain is lacking. However, exposure untraceable to encephalitis has been repeatedly associated with impaired working memory and executive functions, particularly among schizophrenia patients. METHODS: Patterns of HSV-1 infection and gene expression changes were examined in human induced pluripotent stem cell (iPSC)-derived neurons. Separately, differences in blood oxygenation level-dependent (BOLD) responses to working memory challenges using letter n-back tests were investigated using functional magnetic resonance imaging (fMRI) among schizophrenia cases/controls. RESULTS: HSV-1 induced lytic changes in iPSC-derived glutamatergic neurons and neuroprogenitor cells. In neurons, HSV-1 also entered a quiescent state following coincubation with antiviral drugs, with distinctive changes in gene expression related to functions such as glutamatergic signaling. In the fMRI studies, main effects of schizophrenia (P = .001) and HSV-1 exposure (1-back, P = 1.76 × 10(-4); 2-back, P = 1.39 × 10(-5)) on BOLD responses were observed. We also noted increased BOLD responses in the frontoparietal, thalamus, and midbrain regions among HSV-1 exposed schizophrenia cases and controls, compared with unexposed persons. CONCLUSIONS: The lytic/quiescent cycles in iPSC-derived neurons indicate that persistent neuronal infection can occur, altering cellular function. The fMRI studies affirm the associations between nonencephalitic HSV-1 infection and functional brain changes linked with working memory impairment. The fMRI and iPSC studies together provide putative mechanisms for the cognitive impairments linked to HSV-1 exposure.


Subject(s)
Brain/physiopathology , Cognition Disorders/physiopathology , Herpes Simplex/physiopathology , Memory, Short-Term/physiology , Neurons/metabolism , RNA, Messenger/metabolism , RNA, Viral/genetics , Schizophrenia/physiopathology , Schizophrenic Psychology , Adolescent , Adult , Case-Control Studies , Cognition Disorders/etiology , Cognition Disorders/virology , Female , Functional Neuroimaging , Gene Expression , Gene Expression Profiling , Herpes Simplex/pathology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/immunology , Humans , Induced Pluripotent Stem Cells/cytology , Magnetic Resonance Imaging , Male , Neurons/cytology , Schizophrenia/complications , Schizophrenia/virology , Young Adult
12.
Methods Mol Biol ; 997: 35-43, 2013.
Article in English | MEDLINE | ID: mdl-23546746

ABSTRACT

Human somatic cells can be reprogrammed to the pluripotent state to become human-induced pluripotent stem cells (hiPSC). This reprogramming is achieved by activating signaling pathways that are expressed during early development. These pathways can be induced by ectopic expression of four transcription factors-Oct4, Sox2, Klf4, and c-Myc. Although there are many ways to deliver these transcription factors into the somatic cells, this chapter will provide protocols that can be used to generate hiPSC from lentiviruses.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Lentivirus/genetics , Animals , Coculture Techniques , Cryopreservation , Culture Media , Genetic Vectors , HEK293 Cells , Humans , Kruppel-Like Factor 4 , Mice , Recombinant Proteins/genetics , Transcription Factors/genetics , Transduction, Genetic
13.
Elife ; 2: e01749, 2013 Dec 31.
Article in English | MEDLINE | ID: mdl-24381249

ABSTRACT

Many studies are uncovering functional roles for long noncoding RNAs (lncRNAs), yet few have been tested for in vivo relevance through genetic ablation in animal models. To investigate the functional relevance of lncRNAs in various physiological conditions, we have developed a collection of 18 lncRNA knockout strains in which the locus is maintained transcriptionally active. Initial characterization revealed peri- and postnatal lethal phenotypes in three mutant strains (Fendrr, Peril, and Mdgt), the latter two exhibiting incomplete penetrance and growth defects in survivors. We also report growth defects for two additional mutant strains (linc-Brn1b and linc-Pint). Further analysis revealed defects in lung, gastrointestinal tract, and heart in Fendrr(-/-) neonates, whereas linc-Brn1b(-/-) mutants displayed distinct abnormalities in the generation of upper layer II-IV neurons in the neocortex. This study demonstrates that lncRNAs play critical roles in vivo and provides a framework and impetus for future larger-scale functional investigation into the roles of lncRNA molecules. DOI: http://dx.doi.org/10.7554/eLife.01749.001.


Subject(s)
Brain/growth & development , RNA, Long Noncoding/physiology , Animals , Mice , Mice, Knockout , RNA, Long Noncoding/genetics
14.
Differentiation ; 84(2): 176-84, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22721678

ABSTRACT

Multipotent P19CL6 cells differentiate into cardiac myocytes or neural lineages when stimulated with dimethyl sulfoxide (DMSO) or retinoic acid (RA), respectively. Expression of the transcription factor Tbx6 was found to increase during cardiac myocyte differentiation and to decrease during neural differentiation. Overexpression of Tbx6 was not sufficient to drive P19CL6 cells to a cardiac myocyte fate or to accelerate DMSO-induced differentiation. In contrast, knockdown of Tbx6 dramatically inhibited DMSO-induced differentiation of P19CL6 cells to cardiac myocytes, as evidenced by the loss of striated muscle-specific markers and spontaneous beating. Tbx6 knockdown was also accompanied by almost complete loss of Nkx2.5, a transcription factor involved in the specification of the cardiac myocyte lineage, indicating that Nkx2.5 is downstream of Tbx6. In distinction to its positive role in cardiac myocyte differentiation, Tbx6 knockdown augmented RA-induced differentiation of P19CL6 cells to both neurons and glia, and accelerated the rate of neurite formation. Conversely, Tbx6 overexpression attenuated differentiation to neural lineages. Thus, in the P19CL6 model, Tbx6 is required for cardiac myocyte differentiation and represses neural differentiation. We propose a model in which Tbx6 is a part of a molecular switch that modulates divergent differentiation programs within a single progenitor cell.


Subject(s)
Cell Differentiation , Multipotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Neurons/cytology , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Lineage , Dimethyl Sulfoxide/pharmacology , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/metabolism , Mice , Multipotent Stem Cells/drug effects , RNA, Small Interfering , T-Box Domain Proteins , Transcription Factors/antagonists & inhibitors , Tretinoin/pharmacology
15.
FASEB J ; 26(8): 3240-51, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22542683

ABSTRACT

Substrates used to culture human embryonic stem cells (hESCs) are typically 2-dimensional (2-D) in nature, with limited ability to recapitulate in vivo-like 3-dimensional (3-D) microenvironments. We examined critical determinants of hESC self-renewal in poly-d-lysine-pretreated synthetic polymer-based substrates with variable microgeometries, including planar 2-D films, macroporous 3-D sponges, and microfibrous 3-D fiber mats. Completely synthetic 2-D substrates and 3-D macroporous scaffolds failed to retain hESCs or support self-renewal or differentiation. However, synthetic microfibrous geometries made from electrospun polymer fibers were found to promote cell adhesion, viability, proliferation, self-renewal, and directed differentiation of hESCs in the absence of any exogenous matrix proteins. Mechanistic studies of hESC adhesion within microfibrous scaffolds indicated that enhanced cell confinement in such geometries increased cell-cell contacts and altered colony organization. Moreover, the microfibrous scaffolds also induced hESCs to deposit and organize extracellular matrix proteins like laminin such that the distribution of laminin was more closely associated with the cells than the Matrigel treatment, where the laminin remained associated with the coated fibers. The production of and binding to laminin was critical for formation of viable hESC colonies on synthetic fibrous scaffolds. Thus, synthetic substrates with specific 3-D microgeometries can support hESC colony formation, self-renewal, and directed differentiation to multiple lineages while obviating the stringent needs for complex, exogenous matrices. Similar scaffolds could serve as tools for developmental biology studies in 3-D and for stem cell differentiation in situ and transplantation using defined humanized conditions.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Tissue Scaffolds , Biopolymers , Cell Adhesion , Cell Differentiation , Cell Proliferation , Collagen , Drug Combinations , Embryonic Stem Cells/drug effects , Humans , Laminin/biosynthesis , Polylysine/pharmacology , Proteoglycans , Stereoisomerism , Tyrosine/analogs & derivatives
16.
Mycopathologia ; 173(1): 13-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21858547

ABSTRACT

Microbial growth in damp indoor environments has been correlated with risks to human health. This study was aimed to determine the cytotoxicity of 1-octen-3-ol ("mushroom alcohol"), a major fungal volatile organic compound (VOC) associated with mushroom and mold odors. Using an airborne exposure technique, human embryonic stem cells were exposed for 1 h to different concentrations (0-1,000 ppm) of racemic 1-octen-3-ol and its enantiomers, (R)-(-)-1-octen-3-ol and (S)-(+)-1-octen-3-ol. Cytotoxicity was assayed using both the MTS (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) assay and the fluorescently tagged Calcein AM-mediated "live and dead" assay. Racemic 1-octen-3-ol and (S)-(+)-1-octen-3-ol exhibited greater cytotoxicity to the undifferentiated human cell line H1 than did (R)-(-)-1-octen-3-ol. The inhibition concentration 50 (IC(50)) values assessed by the MTS assay for racemic 1-octen-3-ol, (S)-(+)-1-octen-3-ol and (R)-(-)-1-octen-3-ol were, respectively, 109, 98, and 258 ppm. These IC(50) values were 40-80-fold lower than that of vapor phase toluene, an industrial chemical used as a positive control in this study. Our report pioneers the modeling of human embryonic stem cells as an in vitro approach to screen the potential toxicity of fungal VOCs. Human embryonic stem cells exposed to 1-octen-3-ol, and its enantiomers in the vapor phase showed more cytotoxicity than those exposed to toluene.


Subject(s)
Embryonic Stem Cells/drug effects , Octanols/toxicity , Volatile Organic Compounds/toxicity , Cell Survival/drug effects , Cells, Cultured , Humans , Inhibitory Concentration 50
17.
Stem Cell Res Ther ; 1(3): 23, 2010 Jul 26.
Article in English | MEDLINE | ID: mdl-20659329

ABSTRACT

INTRODUCTION: Genetic manipulation of human embryonic stem cells (hESC) has been limited by their general resistance to common methods used to introduce exogenous DNA or RNA. Efficient and high throughput transfection of nucleic acids into hESC would be a valuable experimental tool to manipulate these cells for research and clinical applications. METHODS: We investigated the ability of two commercially available electroporation systems, the Nucleofection® 96-well Shuttle® System from Lonza and the Neon™ Transfection System from Invitrogen to efficiently transfect hESC. Transfection efficiency was measured by flow cytometry for the expression of the green fluorescent protein and the viability of the transfected cells was determined by an ATP catalyzed luciferase reaction. The transfected cells were also analyzed by flow cytometry for common markers of pluripotency. RESULTS: Both systems are capable of transfecting hESC at high efficiencies with little loss of cell viability. However, the reproducibility and the ease of scaling for high throughput applications led us to perform more comprehensive tests on the Nucleofection® 96-well Shuttle® System. We demonstrate that this method yields a large fraction of transiently transfected cells with minimal loss of cell viability and pluripotency, producing protein expression from plasmid vectors in several different hESC lines. The method scales to a 96-well plate with similar transfection efficiencies at the start and end of the plate. We also investigated the efficiency with which stable transfectants can be generated and recovered under antibiotic selection. Finally, we found that this method is effective in the delivery of short synthetic RNA oligonucleotides (siRNA) into hESC for knockdown of translation activity via RNA interference. CONCLUSIONS: Our results indicate that these electroporation methods provide a reliable, efficient, and high-throughput approach to the genetic manipulation of hESC.


Subject(s)
Electroporation/methods , Embryonic Stem Cells/cytology , Transfection/methods , Cell Differentiation/genetics , Cell Survival , Cells, Cultured , DNA/genetics , Electroporation/instrumentation , Embryonic Stem Cells/metabolism , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Pluripotent Stem Cells/metabolism , RNA Interference , RNA, Small Interfering/genetics
18.
Stem Cell Res ; 4(2): 92-106, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20060795

ABSTRACT

Meticulous characterization of human embryonic stem cells (hESC) is critical to their eventual use in cell-based therapies, particularly in view of the diverse methods for derivation and maintenance of these cell lines. However, characterization methods are generally not standardized and many currently used assays are subjective, making dependable and direct comparison of cell lines difficult. In order to address this problem, we selected 10 molecular-based high-resolution assays as components of a panel for characterization of hESC. The selection of the assays was primarily based on their quantitative or objective (rather than subjective) nature. We demonstrate the efficacy of this panel by characterizing 4 hESC lines, derived in two different laboratories using different derivation techniques, as pathogen free, genetically stable, and able to differentiate into derivatives of all three germ layers. Our panel expands and refines a characterization panel previously proposed by the International Stem Cell Initiative and is another step toward standardized hESC characterization and quality control, a crucial element of successful hESC research and clinical translation.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Flow Cytometry , Humans , Immunohistochemistry , Karyotyping , Microsatellite Repeats/genetics , Reverse Transcriptase Polymerase Chain Reaction
19.
PLoS One ; 4(9): e7192, 2009 Sep 28.
Article in English | MEDLINE | ID: mdl-19784364

ABSTRACT

BACKGROUND: MicroRNAs are required for maintenance of pluripotency as well as differentiation, but since more microRNAs have been computationally predicted in genome than have been found, there are likely to be undiscovered microRNAs expressed early in stem cell differentiation. METHODOLOGY/PRINCIPAL FINDINGS: SOLiD ultra-deep sequencing identified >10(7) unique small RNAs from human embryonic stem cells (hESC) and neural-restricted precursors that were fit to a model of microRNA biogenesis to computationally predict 818 new microRNA genes. These predicted genomic loci are associated with chromatin patterns of modified histones that are predictive of regulated gene expression. 146 of the predicted microRNAs were enriched in Ago2-containing complexes along with 609 known microRNAs, demonstrating association with a functional RISC complex. This Ago2 IP-selected subset was consistently expressed in four independent hESC lines and exhibited complex patterns of regulation over development similar to previously-known microRNAs, including pluripotency-specific expression in both hESC and iPS cells. More than 30% of the Ago2 IP-enriched predicted microRNAs are new members of existing families since they share seed sequences with known microRNAs. CONCLUSIONS/SIGNIFICANCE: Extending the classic definition of microRNAs, this large number of new microRNA genes, the majority of which are less conserved than their canonical counterparts, likely represent evolutionarily recent regulators of early differentiation. The enrichment in Ago2 containing complexes, the presence of chromatin marks indicative of regulated gene expression, and differential expression over development all support the identification of 146 new microRNAs active during early hESC differentiation.


Subject(s)
Embryonic Stem Cells/cytology , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Profiling/methods , MicroRNAs/metabolism , Neurons/metabolism , Argonaute Proteins , Cell Differentiation , Cell Line , Gene Expression Regulation , Genome , Humans , Immunoprecipitation , Karyotyping , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , RNA/metabolism
20.
Biochem Biophys Res Commun ; 377(1): 46-51, 2008 Dec 05.
Article in English | MEDLINE | ID: mdl-18823947

ABSTRACT

Gap junctions, encoded by the connexin (Cx) multi-gene family, couple adjacent cells and underlie cell-cell communications. Previous mouse studies suggest that Cxs play an important role in development but their role in human cardiogenesis is undefined. Human embryonic stem cells (hESC) provide a unique model for studying human differentiation. Lentivirus-mediated stable overexpression of Cx43 in hESC (Cx43-hESC) did not affect colony morphology, karyotype and expression of pluripotency genes such as Oct4 but completely suppressed the formation of spontaneously beating, cardiomyocyte-containing clusters in embryoid bodies (EBs). Unlike control hEBs, the transcripts of several mesodermal markers (kallikrein, delta-globin, and CMP), ventricular myosin light chain and cardiac troponin I were absent or delayed. Transcriptomic and pathway analyses showed that 194 genes crucial for movement, growth, differentiation and maintenance were differentially expressed in Cx43-hESC. We conclude that Cx43 mediates the expression of an array of genes involved in human cardiogenesis, in addition to intercellular communication.


Subject(s)
Connexin 43/metabolism , Embryonic Stem Cells/physiology , Gap Junctions/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Organogenesis/genetics , Pluripotent Stem Cells/physiology , Animals , Cell Communication/genetics , Cell Differentiation/genetics , Cell Line , Connexin 43/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Humans , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Rats , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...