Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Gene Ther ; 30(3-4): 386-397, 2023 04.
Article in English | MEDLINE | ID: mdl-36258038

ABSTRACT

Gene editing for the cure of inborn errors of metabolism (IEMs) has been limited by inefficiency of adult hepatocyte targeting. Here, we demonstrate that in utero CRISPR/Cas9-mediated gene editing in a mouse model of hereditary tyrosinemia type 1 provides stable cure of the disease. Following this, we performed an extensive gene expression analysis to explore the inherent characteristics of fetal/neonatal hepatocytes that make them more susceptible to efficient gene editing than adult hepatocytes. We showed that fetal and neonatal livers are comprised of proliferative hepatocytes with abundant expression of genes involved in homology-directed repair (HDR) of DNA double-strand breaks (DSBs), key for efficient gene editing by CRISPR/Cas9. We demonstrated the same is true of hepatocytes after undergoing a regenerative stimulus (partial hepatectomy), where post-hepatectomy cells show a higher efficiency of HDR and correction. Specifically, we demonstrated that HDR-related genome correction is most effective in the replicative phase, or S-phase, of an actively proliferating cell. In conclusion, this study shows that taking advantage of or triggering cell proliferation, specifically DNA replication in S-phase, may serve as an important tool to improve efficiency of CRISPR/Cas9-mediated genome editing in the liver and provide a curative therapy for IEMs in both children and adults.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , Mice , Recombinational DNA Repair , DNA Breaks, Double-Stranded , DNA , DNA Repair
2.
Cells ; 10(8)2021 07 31.
Article in English | MEDLINE | ID: mdl-34440722

ABSTRACT

Human stem cell-derived neurons are increasingly considered powerful models in drug discovery and disease modeling, despite limited characterization of their molecular properties. Here, we have conducted a detailed study of the properties of a commercial human induced Pluripotent Stem Cell (iPSC)-derived neuron line, iCell [GABA] neurons, maintained for up to 3 months in vitro. We confirmed that iCell neurons display neurite outgrowth within 24 h of plating and label for the pan-neuronal marker, ßIII tubulin within the first week. Our multi-electrode array (MEA) recordings clearly showed neurons generated spontaneous, spike-like activity within 2 days of plating, which peaked at one week, and rapidly decreased over the second week to remain at low levels up to one month. Extracellularly recorded spikes were reversibly inhibited by tetrodotoxin. Patch-clamp experiments showed that iCell neurons generated spontaneous action potentials and expressed voltage-gated Na and K channels with membrane capacitances, resistances and membrane potentials that are consistent with native neurons. Our single neuron recordings revealed that reduced spiking observed in the MEA after the first week results from development of a dominant inhibitory tone from GABAergic neuron circuit maturation. GABA evoked concentration-dependent currents that were inhibited by the convulsants, bicuculline and picrotoxin, and potentiated by the positive allosteric modulators, diazepam, chlordiazepoxide, phenobarbital, allopregnanolone and mefenamic acid, consistent with native neuronal GABAA receptors. We also show that glycine evoked robust concentration-dependent currents that were inhibited by the neurotoxin, strychnine. Glutamate, AMPA, Kainate and NMDA each evoked concentration-dependent currents in iCell neurons that were blocked by their selective antagonists, consistent with the expression of ionotropic glutamate receptors. The NMDA currents required the presence of the co-agonist glycine and were blocked in a highly voltage-dependent manner by Mg2+ consistent with the properties of native neuronal NMDA receptors. Together, our data suggest that such human iPSC-derived neurons may have significant value in drug discovery and development and may eventually largely replace the need for animal tissues in human biomedical research.


Subject(s)
Drug Discovery , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , GABAergic Neurons/drug effects , Induced Pluripotent Stem Cells/drug effects , Neural Stem Cells/drug effects , Cell Line , Dose-Response Relationship, Drug , GABAergic Neurons/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Ion Channels/drug effects , Ion Channels/metabolism , Membrane Potentials , Neural Stem Cells/metabolism , Phenotype
3.
Heterocycles ; 101(1): 145-164, 2020.
Article in English | MEDLINE | ID: mdl-32773946

ABSTRACT

Neuropathic pain, epilepsy, insomnia, and tremor disorder may arrive from an increase of intracellular Ca2+ concentration through a dysfunction of T-type Ca2+ channels. Thus, T-type calcium channels could be a target in drug discovery for the treatments of neuropathic pain and epilepsy. From rational drug design approach, a group of 2,5-disubstituted 1,3,4-oxadiazole molecules was synthesized and their selective T-type channel inhibitions were evaluated. The synthetic strategy consists of a short sequence of three reactions: (i) condensation of thiosemicarbazide with acid chlorides; (ii) ring closing by 1,3-dibromo-5,5- dimethylhydantoin; and (iii) coupling with various acid chlorides. 5-Chloro-N-(5- phenyl-1,3,4-oxadiazol-2-yl)thiophene-2-carboxamide (11) was found to selectively inhibit T-type Ca2+ channel over Na+ and K+ channels in mouse dorsal root ganglion neurons and/or human embryonic kidney (HEK)-293 cells and to suppress seizure-induced death in mouse model. Consequently, compound 11 is a useful probe for investigation of physiologic and pathophysiologic roles of the T-channel, and provides a basis to develop a novel therapeutic to treat chronic neuropathic and inflammatory pains.

4.
Sleep ; 42(3)2019 03 01.
Article in English | MEDLINE | ID: mdl-30535004

ABSTRACT

STUDY OBJECTIVES: A major challenge in treating insomnia is to find effective medicines with fewer side effects. Activation of G-protein-gated inward rectifying K+ channels (GIRKs) by GABAB agonists baclofen or γ-hydroxybutyric acid (GHB) promotes nonrapid eye movement (NREM) sleep and consolidates sleep. However, baclofen has poor brain penetration, GHB possesses abuse liability, and in rodents both drugs cause spike-wave discharges (SWDs), an absence seizure activity. We tested the hypothesis that direct GIRK activation promotes sleep without inducing SWD using ML297, a potent and selective GIRK activator. METHODS: Whole-cell patch-clamp recordings from hypocretin/orexin or hippocampal neurons in mouse brain slices were made to study neuronal excitability and synaptic activity; spontaneous activity, locomotion, contextual and tone-conditioned memory, and novel object recognition were assessed. Electroencephalogram/electromyogram (EEG/EMG) recordings were used to study GIRK modulation of sleep. RESULTS: ML297, like baclofen, caused membrane hyperpolarization, decreased input resistance, and blockade of spontaneous action potentials. Unlike baclofen, ML297 (5-10 µM) did not cause significant depression of postsynaptic excitatory and inhibitory currents (EPSCs-IPSCs), indicating preferential postsynaptic inhibition. ML297 (30 mg/kg, i.p.) inhibited wake activity and locomotion, and preferentially increased NREM sleep without altering EEG delta power, REM sleep, inducing SWDs, or impairing conditioned memory and novel object recognition. CONCLUSIONS: This study finds that direct activation of neuronal GIRK channels modulates postsynaptic membrane excitability and prolongs NREM sleep without changing sleep intensity, inducing SWDs, or impairing memory in rodents. These results suggest that direct GIRK activation with a selective compound may present an innovative approach for the treatment of chronic insomnia.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/agonists , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Phenylurea Compounds/pharmacology , Pyrazoles/pharmacology , Sleep Stages/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Electromyography/drug effects , Electromyography/methods , Female , Hippocampus/drug effects , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Neurons/physiology , Organ Culture Techniques , Patch-Clamp Techniques/methods , Sleep Stages/drug effects
5.
Cell Stem Cell ; 23(5): 758-771.e8, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30318302

ABSTRACT

Comprehensive identification of factors that can specify neuronal fate could provide valuable insights into lineage specification and reprogramming, but systematic interrogation of transcription factors, and their interactions with each other, has proven technically challenging. We developed a CRISPR activation (CRISPRa) approach to systematically identify regulators of neuronal-fate specification. We activated expression of all endogenous transcription factors and other regulators via a pooled CRISPRa screen in embryonic stem cells, revealing genes including epigenetic regulators such as Ezh2 that can induce neuronal fate. Systematic CRISPR-based activation of factor pairs allowed us to generate a genetic interaction map for neuronal differentiation, with confirmation of top individual and combinatorial hits as bona fide inducers of neuronal fate. Several factor pairs could directly reprogram fibroblasts into neurons, which shared similar transcriptional programs with endogenous neurons. This study provides an unbiased discovery approach for systematic identification of genes that drive cell-fate acquisition.


Subject(s)
CRISPR-Cas Systems/genetics , Cellular Reprogramming/genetics , Gene Editing , Mutagenesis, Site-Directed/methods , Neurons/cytology , Neurons/metabolism , Transcription Factors/genetics , Animals , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Transcription Factors/metabolism
6.
J Alzheimers Dis ; 58(2): 559-574, 2017.
Article in English | MEDLINE | ID: mdl-28482635

ABSTRACT

There is an urgent unmet need for new therapeutics for Alzheimer's disease (AD), the most common cause of dementia in the elderly. Therapeutic approaches targeting amyloid-ß (Aß) and its downstream toxicities have become major strategies in AD drug development. We have taken a rational design approach and synthesized a class of tricyclic pyrone (TP) compounds that show anti-Aß and other neuroprotective actions. The in vivo efficacy of a lead TP named CP2 to ameliorate AD-like pathologies has been shown in mouse models. Here we report the selection and initial characterization of a new lead TP70, which exhibited an anti-Aß therapeutic index even higher than CP2. Moreover, TP70 was able to reduce oxidative stress, inhibit acyl-coenzyme A:cholesterol acyltransferase (ACAT), and upregulate the expression of ATP-binding cassette subfamily A, member 1 (ABCA1), actions considered neuroprotective in AD. TP70 further showed excellent pharmacokinetic properties, including brain penetration and oral availability. When administered to 5xFAD mice via intraperitoneal or oral route, TP70 enhanced the overall solubility and decreased the level of cerebral Aß, including both fibrillary and soluble Aß species. Interestingly, TP70 enhanced N-methyl-D-aspartate (NMDA) receptor-mediated excitatory post-synaptic potential (EPSP) in the hippocampal CA1 area, increased the magnitude of NMDA-dependent hippocampal long-term potentiation (LTP), a cellular model of learning and memory, and prevented the Aß oligomer-impaired LTP. Significantly, a single dose of TP70 administered to aged 5xFAD mice was effective in mitigating the impaired LTP induction, recorded at 24 h after administration. Our results support a potential of TP70 in clinical development for AD in view of its synergistic neuroprotective actions, ability to positively modulate NMDA receptor-mediated hippocampal plasticity, and favorable pharmacokinetic properties in rodents.


Subject(s)
Alzheimer Disease/drug therapy , Amyloidogenic Proteins/metabolism , Brain/drug effects , Brain/metabolism , Neuroprotective Agents/therapeutic use , Pyrones/therapeutic use , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Amyloidogenic Proteins/toxicity , Animals , Brain/pathology , Cell Line, Tumor , Disease Models, Animal , Drinking Behavior/drug effects , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Humans , Locomotion/drug effects , Locomotion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/genetics , Mutation/genetics , Neuroblastoma/pathology , Neuroprotective Agents/chemistry , Presenilin-1/genetics , Pyrones/chemical synthesis , Pyrones/chemistry
7.
Toxicol In Vitro ; 29(3): 592-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637331

ABSTRACT

Determination of the impact of a drug on human brain development relies instead on surrogate animal studies. Here we have exploited the human stem cell line, TERA2.cl.SP12 to differentiate into neurons and addressed their value as an in vitro model to evaluate the risk of developmental neurotoxicity with antiepileptic drugs (AEDs). The effects of four AEDs were investigated on cell viability, cell cycle and neural differentiation. Exposure to either phenobarbital (10-1000 µM), valproic acid (10-1000 µM), lamotrigine (1-100 µM) or carbamazepine (1-100 µM) for 3 days reduced viability in non-differentiating cells only at the highest concentrations tested. Viability was also reduced with lower concentrations of all AEDs in cells undergoing neural differentiation. Valproic acid and carbamazepine increased DNA fragmentation and reduced cell cycle progression. 3 days exposure at the start of neural differentiation to phenobarbital, valproic acid or lamotrigine also significantly reduced the proportion of stem cells that subsequently differentiated into neurons at 15 days in vitro. The two control agents tested, ciprofloxacin and perfluorooctanoic acid had no impact on neurogenesis in vitro. These new data show that modelling neurogenesis in vitro using a human stem cell line may be a powerful method to predict risks of developmental neurotoxicity in vivo with psychotropic drugs.


Subject(s)
Anticonvulsants/toxicity , Nervous System Diseases/chemically induced , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Neurons/drug effects , Neurotoxicity Syndromes/pathology , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , DNA Fragmentation/drug effects , Humans , Neurons/pathology , Risk
SELECTION OF CITATIONS
SEARCH DETAIL
...