Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Genes Dev ; 37(19-20): 883-900, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37890975

RESUMEN

Loss-of-function mutations in MECP2 cause Rett syndrome (RTT), a severe neurological disorder that mainly affects girls. Mutations in MECP2 do occur in males occasionally and typically cause severe encephalopathy and premature lethality. Recently, we identified a missense mutation (c.353G>A, p.Gly118Glu [G118E]), which has never been seen before in MECP2, in a young boy who suffered from progressive motor dysfunction and developmental delay. To determine whether this variant caused the clinical symptoms and study its functional consequences, we established two disease models, including human neurons from patient-derived iPSCs and a knock-in mouse line. G118E mutation partially reduces MeCP2 abundance and its DNA binding, and G118E mice manifest RTT-like symptoms seen in the patient, affirming the pathogenicity of this mutation. Using live-cell and single-molecule imaging, we found that G118E mutation alters MeCP2's chromatin interaction properties in live neurons independently of its effect on protein levels. Here we report the generation and characterization of RTT models of a male hypomorphic variant and reveal new insight into the mechanism by which this pathological mutation affects MeCP2's chromatin dynamics. Our ability to quantify protein dynamics in disease models lays the foundation for harnessing high-resolution single-molecule imaging as the next frontier for developing innovative therapies for RTT and other diseases.


Asunto(s)
Cromatina , Síndrome de Rett , Femenino , Humanos , Masculino , Ratones , Animales , Cromatina/metabolismo , Encéfalo/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/genética , Mutación , Neuronas/metabolismo
2.
Am J Hum Genet ; 111(5): 841-862, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38593811

RESUMEN

RNA sequencing (RNA-seq) has recently been used in translational research settings to facilitate diagnoses of Mendelian disorders. A significant obstacle for clinical laboratories in adopting RNA-seq is the low or absent expression of a significant number of disease-associated genes/transcripts in clinically accessible samples. As this is especially problematic in neurological diseases, we developed a clinical diagnostic approach that enhanced the detection and evaluation of tissue-specific genes/transcripts through fibroblast-to-neuron cell transdifferentiation. The approach is designed specifically to suit clinical implementation, emphasizing simplicity, cost effectiveness, turnaround time, and reproducibility. For clinical validation, we generated induced neurons (iNeurons) from 71 individuals with primary neurological phenotypes recruited to the Undiagnosed Diseases Network. The overall diagnostic yield was 25.4%. Over a quarter of the diagnostic findings benefited from transdifferentiation and could not be achieved by fibroblast RNA-seq alone. This iNeuron transcriptomic approach can be effectively integrated into diagnostic whole-transcriptome evaluation of individuals with genetic disorders.


Asunto(s)
Transdiferenciación Celular , Fibroblastos , Neuronas , Análisis de Secuencia de ARN , Humanos , Transdiferenciación Celular/genética , Fibroblastos/metabolismo , Fibroblastos/citología , Análisis de Secuencia de ARN/métodos , Neuronas/metabolismo , Neuronas/citología , Transcriptoma , Reproducibilidad de los Resultados , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/diagnóstico , RNA-Seq/métodos , Femenino , Masculino
3.
Proc Natl Acad Sci U S A ; 119(13): e2023784119, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35333654

RESUMEN

Neural stem cells, the source of newborn neurons in the adult hippocampus, are intimately involved in learning and memory, mood, and stress response. Despite considerable progress in understanding the biology of neural stem cells and neurogenesis, regulating the neural stem cell population precisely has remained elusive because we have lacked the specific targets to stimulate their proliferation and neurogenesis. The orphan nuclear receptor TLX/NR2E1 governs neural stem and progenitor cell self-renewal and proliferation, but the precise mechanism by which it accomplishes this is not well understood because its endogenous ligand is not known. Here, we identify oleic acid (18:1ω9 monounsaturated fatty acid) as such a ligand. We first show that oleic acid is critical for neural stem cell survival. Next, we demonstrate that it binds to TLX to convert it from a transcriptional repressor to a transcriptional activator of cell-cycle and neurogenesis genes, which in turn increases neural stem cell mitotic activity and drives hippocampal neurogenesis in mice. Interestingly, oleic acid-activated TLX strongly up-regulates cell cycle genes while only modestly up-regulating neurogenic genes. We propose a model in which sufficient quantities of this endogenous ligand must bind to TLX to trigger the switch to proliferation and drive the progeny toward neuronal lineage. Oleic acid thus serves as a metabolic regulator of TLX activity that can be used to selectively target neural stem cells, paving the way for future therapeutic manipulations to counteract pathogenic impairments of neurogenesis.


Asunto(s)
Hipocampo , Neurogénesis , Ácido Oléico , Receptores Citoplasmáticos y Nucleares , Animales , Proliferación Celular , Hipocampo/crecimiento & desarrollo , Hipocampo/metabolismo , Ligandos , Ratones , Neurogénesis/fisiología , Ácido Oléico/metabolismo , Receptores Nucleares Huérfanos , Receptores Citoplasmáticos y Nucleares/metabolismo
4.
J Neurosci ; 40(2): 459-477, 2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31748376

RESUMEN

α-Synuclein (α-Syn) accumulation is a pathological hallmark of Parkinson's disease. Duplications and triplications of SNCA, the gene coding for α-Syn, cause genetic forms of the disease, which suggests that increased α-Syn dosage can drive PD. To identify the proteins that regulate α-Syn, we previously performed a screen of potentially druggable genes that led to the identification of 60 modifiers. Among them, Doublecortin-like kinase 1 (DCLK1), a microtubule binding serine threonine kinase, emerged as a promising target due to its potent effect on α-Syn and potential druggability as a neuron-expressed kinase. In this study, we explore the relationship between DCLK1 and α-Syn in human cellular and mouse models of PD. First, we show that DCLK1 regulates α-Syn levels post-transcriptionally. Second, we demonstrate that knockdown of Dclk1 reduces phosphorylated species of α-Syn and α-Syn-induced neurotoxicity in the SNc in two distinct mouse models of synucleinopathy. Last, silencing DCLK1 in human neurons derived from individuals with SNCA triplications reduces phosphorylated and total α-Syn, thereby highlighting DCLK1 as a potential therapeutic target to reduce pathological α-Syn in disease.SIGNIFICANCE STATEMENT DCLK1 regulates α-Syn protein levels, and Dclk1 knockdown rescues α-Syn toxicity in mice. This study provides evidence for a novel function for DCLK1 in the mature brain, and for its potential as a new therapeutic target for synucleinopathies.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Enfermedad de Parkinson/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , alfa-Sinucleína/metabolismo , Animales , Quinasas Similares a Doblecortina , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo
5.
Am J Hum Genet ; 101(6): 874-887, 2017 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-29129316

RESUMEN

Copy-number variants (CNVs) of chromosome 15q13.3 manifest clinically as neuropsychiatric disorders with variable expressivity. CHRNA7, encoding for the α7 nicotinic acetylcholine receptor (nAChR), has been suggested as a candidate gene for the phenotypes observed. Here, we used induced pluripotent stem cells (iPSCs) and neural progenitor cells (NPCs) derived from individuals with heterozygous 15q13.3 deletions and heterozygous 15q13.3 duplications to investigate the CHRNA7-dependent molecular consequences of the respective CNVs. Unexpectedly, both deletions and duplications lead to decreased α7 nAChR-associated calcium flux. For deletions, this decrease in α7 nAChR-dependent calcium flux is expected due to haploinsufficiency of CHRNA7. For duplications, we found that increased expression of CHRNA7 mRNA is associated with higher expression of nAChR-specific and resident ER chaperones, indicating increased ER stress. This is likely a consequence of inefficient chaperoning and accumulation of α7 subunits in the ER, as opposed to being incorporated into functional α7 nAChRs at the cell membrane. Here, we showed that α7 nAChR-dependent calcium signal cascades are downregulated in both 15q13.3 deletion and duplication NPCs. While it may seem surprising that genomic changes in opposite direction have consequences on downstream pathways that are in similar direction, it aligns with clinical data, which suggest that both individuals with deletions and duplications of 15q13.3 manifest neuropsychiatric disease and cognitive deficits.


Asunto(s)
Señalización del Calcio/genética , Trastornos de los Cromosomas/genética , Variaciones en el Número de Copia de ADN/genética , Estrés del Retículo Endoplásmico/genética , Dosificación de Gen/genética , Células Madre Pluripotentes Inducidas/citología , Discapacidad Intelectual/genética , Células-Madre Neurales/citología , Convulsiones/genética , Receptor Nicotínico de Acetilcolina alfa 7/genética , Adolescente , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 15/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Receptor Nicotínico de Acetilcolina alfa 7/biosíntesis
6.
J Neurosci ; 38(43): 9286-9301, 2018 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-30249792

RESUMEN

Accumulation of α-Synuclein (α-Syn) causes Parkinson's disease (PD) as well as other synucleopathies. α-Syn is the major component of Lewy bodies and Lewy neurites, the proteinaceous aggregates that are a hallmark of sporadic PD. In familial forms of PD, mutations or copy number variations in SNCA (the α-Syn gene) result in a net increase of its protein levels. Furthermore, common risk variants tied to PD are associated with small increases of wild-type α-Syn levels. These findings are further bolstered by animal studies which show that overexpression of α-Syn is sufficient to cause PD-like features. Thus, increased α-Syn levels are intrinsically tied to PD pathogenesis and underscore the importance of identifying the factors that regulate its levels. In this study, we establish a pooled RNAi screening approach and validation pipeline to probe the druggable genome for modifiers of α-Syn levels and identify 60 promising targets. Using a cross-species, tiered validation approach, we validate six strong candidates that modulate α-Syn levels and toxicity in cell lines, Drosophila, human neurons, and mouse brain of both sexes. More broadly, this genetic strategy and validation pipeline can be applied for the identification of therapeutic targets for disorders driven by dosage-sensitive proteins.SIGNIFICANCE STATEMENT We present a research strategy for the systematic identification and validation of genes modulating the levels of α-Synuclein, a protein involved in Parkinson's disease. A cell-based screen of the druggable genome (>7,500 genes that are potential therapeutic targets) yielded many modulators of α-Synuclein that were subsequently confirmed and validated in Drosophila, human neurons, and mouse brain. This approach has broad applicability to the multitude of neurological diseases that are caused by mutations in genes whose dosage is critical for brain function.


Asunto(s)
Genoma/genética , Neuronas/fisiología , Interferencia de ARN/fisiología , Análisis de Secuencia de ARN/métodos , alfa-Sinucleína/genética , Animales , Animales Recién Nacidos , Drosophila , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Reproducibilidad de los Resultados , Especificidad de la Especie
7.
Stem Cell Res ; 74: 103292, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38154383

RESUMEN

MECP2 Duplication Syndrome (MDS) is a rare, severe neurodevelopmental disorder arising from duplications in the Xq28 region containing the MECP2 gene that predominantly affects males. We generated five human induced pluripotent stem cell (iPSC) lines from the fibroblasts of individuals carrying between 0.355 and 11.2 Mb size duplications in the chromosomal locus containing MECP2. All lines underwent extensive testing to confirm MECP2 duplication and iPSC-related features such as morphology, pluripotency markers, and trilineage differentiation potential. These lines are a valuable resource for molecular and functional studies of MDS as well as screening for a variety of therapeutic approaches.


Asunto(s)
Células Madre Pluripotentes Inducidas , Discapacidad Intelectual Ligada al Cromosoma X , Proteína 2 de Unión a Metil-CpG , Humanos , Masculino , Diferenciación Celular , Duplicación de Gen , Discapacidad Intelectual Ligada al Cromosoma X/genética , Proteína 2 de Unión a Metil-CpG/genética
8.
Nat Commun ; 14(1): 3911, 2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37400440

RESUMEN

Batten disease, one of the most devastating types of neurodegenerative lysosomal storage disorders, is caused by mutations in CLN3. Here, we show that CLN3 is a vesicular trafficking hub connecting the Golgi and lysosome compartments. Proteomic analysis reveals that CLN3 interacts with several endo-lysosomal trafficking proteins, including the cation-independent mannose 6 phosphate receptor (CI-M6PR), which coordinates the targeting of lysosomal enzymes to lysosomes. CLN3 depletion results in mis-trafficking of CI-M6PR, mis-sorting of lysosomal enzymes, and defective autophagic lysosomal reformation. Conversely, CLN3 overexpression promotes the formation of multiple lysosomal tubules, which are autophagy and CI-M6PR-dependent, generating newly formed proto-lysosomes. Together, our findings reveal that CLN3 functions as a link between the M6P-dependent trafficking of lysosomal enzymes and lysosomal reformation pathway, explaining the global impairment of lysosomal function in Batten disease.


Asunto(s)
Glicoproteínas de Membrana , Lipofuscinosis Ceroideas Neuronales , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Lipofuscinosis Ceroideas Neuronales/genética , Lipofuscinosis Ceroideas Neuronales/metabolismo , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Proteómica , Chaperonas Moleculares/metabolismo , Lisosomas/metabolismo , Hidrolasas/metabolismo , Autofagia
9.
Nat Cell Biol ; 24(5): 737-747, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35484250

RESUMEN

Human NANOG expression resets stem cells to ground-state pluripotency. Here we identify the unique features of human NANOG that relate to its dose-sensitive function as a master transcription factor. NANOG is largely disordered, with a C-terminal prion-like domain that phase-transitions to gel-like condensates. Full-length NANOG readily forms higher-order oligomers at low nanomolar concentrations, orders of magnitude lower than typical amyloids. Using single-molecule Förster resonance energy transfer and fluorescence cross-correlation techniques, we show that NANOG oligomerization is essential for bridging DNA elements in vitro. Using chromatin immunoprecipitation sequencing and Hi-C 3.0 in cells, we validate that NANOG prion-like domain assembly is essential for specific DNA recognition and distant chromatin interactions. Our results provide a physical basis for the indispensable role of NANOG in shaping the pluripotent genome. NANOG's unique ability to form prion-like assemblies could provide a cooperative and concerted DNA bridging mechanism that is essential for chromatin reorganization and dose-sensitive activation of ground-state pluripotency.


Asunto(s)
Cromatina , Priones , Cromatina/genética , ADN/genética , Humanos , Proteína Homeótica Nanog/genética , Priones/genética
10.
Gen Physiol Biophys ; 28 Spec No: 212-8, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19893103

RESUMEN

Amyotrophic lateral sclerosis (ALS) is a devastating neurological disorder affecting upper and lower motoneurons. The transgenic ALS rat model (hSOD-1(G93A)) was used for magnetic resonance imaging (MRI) study using a low field wide bore magnet. T2-weighted hyperintensities were observed in the brainstem, rubrospinal tract and vagus motor nuclei with prominent lateral ventricle and cerebral aqueduct enlargements. These changes could be observed already in presymptomatic animals. T2*-weighted MRI with magnetically labeled antibodies (against CD4) revealed lymphocyte infiltration in the brainstem-midbrain region corresponding to the areas of dilated lateral ventricles. Confocal imaging revealed reactive astroglia in these areas. Thus, with the use of wide bore MRI new sites of neurodegeneration and inflammation were revealed in the hSOD-1(G93A) rat model.


Asunto(s)
Esclerosis Amiotrófica Lateral/complicaciones , Encefalopatías/diagnóstico , Encefalopatías/patología , Supervivencia Tisular , Esclerosis Amiotrófica Lateral/patología , Animales , Encefalopatías/complicaciones , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Imagen por Resonancia Magnética , Microscopía Confocal , Ratas , Ratas Sprague-Dawley
11.
Sci Rep ; 9(1): 15935, 2019 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-31685878

RESUMEN

MAGEL2 is a maternally imprinted, paternally expressed gene, located in the Prader-Willi region of human chromosome 15. Pathogenic variants in the paternal copy of MAGEL2 cause Schaaf-Yang syndrome (SHFYNG), a neurodevelopmental disorder related to Prader-Willi syndrome (PWS). Patients with SHFYNG, like PWS, manifest neonatal hypotonia, feeding difficulties, hypogonadism, intellectual disability and sleep apnea. However, individuals with SHFYNG have joint contractures, greater cognitive impairment, and higher prevalence of autism than seen in PWS. Additionally, SHFYNG is associated with a lower prevalence of hyperphagia and obesity than PWS. Previous studies have shown that truncating variants in MAGEL2 lead to SHFYNG. However, the molecular pathways involved in manifestation of the SHFYNG disease phenotype are still unknown. Here we show that a Magel2 null mouse model and fibroblast cell lines from individuals with SHFYNG exhibit increased expression of mammalian target of rapamycin (mTOR) and decreased autophagy. Additionally, we show that SHFYNG induced pluripotent stem cell (iPSC)-derived neurons exhibit impaired dendrite formation. Alterations in SHFYNG patient fibroblast lines and iPSC-derived neurons are rescued by treatment with the mTOR inhibitor rapamycin. Collectively, our findings identify mTOR as a potential target for the development of pharmacological treatments for SHFYNG.


Asunto(s)
Autofagia , Síndrome de Prader-Willi/patología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/efectos de los fármacos , Dendritas/fisiología , Modelos Animales de Enfermedad , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/citología , Neuronas/metabolismo , Fenotipo , Síndrome de Prader-Willi/metabolismo , Proteínas/genética , Proteínas/metabolismo , ARN Largo no Codificante/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Regulación hacia Arriba
12.
Methods Mol Biol ; 1842: 265-281, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30196417

RESUMEN

Over the past decade, advances in systems biology or 'omics techniques have enabled unprecedented insights into the biological processes that occur in cells, tissues, and on the organism level. One of these technologies is the metabolomics, which examines the whole content of the metabolites in a given sample. In a biological system, a stem cell for instance, there are thousands of single components, such as genes, RNA, proteins, and metabolites. These multiple molecular species interact with each other and these interactions may change over the life-time of a cell or in response to specific stimuli, adding to the complexity of the system. Using metabolomics, we can obtain an instantaneous snapshot of the biological status of a cell, tissue, or organism and gain insights on the pattern(s) of numerous analytes, both known and unknown, that result because of a given biological condition. Here, we outline the main methods to study the metabolism of stem cells, including a relatively recent technology of mass spectrometry imaging. Given the abundant and increasing interest in stem cell metabolism in both physiological and pathological conditions, we hope that this chapter will provide incentives for more research in these areas to ultimately reach wide network of applications in biomedical, pharmaceutical, and nutritional research and clinical medicine.


Asunto(s)
Metabolismo Energético , Metabolómica , Células Madre/metabolismo , Aminoácidos/metabolismo , Glucólisis , Humanos , Espectroscopía de Resonancia Magnética , Espectrometría de Masas , Metabolómica/métodos , Células-Madre Neurales/metabolismo
13.
Neuron ; 97(6): 1235-1243.e5, 2018 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-29526553

RESUMEN

Polyglutamine (polyQ) diseases are caused by expansion of translated CAG repeats in distinct genes leading to altered protein function. In spinocerebellar ataxia type 1 (SCA1), a gain of function of polyQ-expanded ataxin-1 (ATXN1) contributes to cerebellar pathology. The extent to which cerebellar toxicity depends on its cognate partner capicua (CIC), versus other interactors, remains unclear. It is also not established whether loss of the ATXN1-CIC complex in the cerebellum contributes to disease pathogenesis. In this study, we exclusively disrupt the ATXN1-CIC interaction in vivo and show that it is at the crux of cerebellar toxicity in SCA1. Importantly, loss of CIC in the cerebellum does not cause ataxia or Purkinje cell degeneration. Expression profiling of these gain- and loss-of-function models, coupled with data from iPSC-derived neurons from SCA1 patients, supports a mechanism in which gain of function of the ATXN1-CIC complex is the major driver of toxicity.


Asunto(s)
Ataxina-1/deficiencia , Cerebelo/metabolismo , Mutación con Ganancia de Función/fisiología , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/metabolismo , Animales , Ataxina-1/genética , Células Cultivadas , Cerebelo/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Ataxias Espinocerebelosas/patología
14.
Elife ; 62017 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-28699891

RESUMEN

Hippocampal neural stem cells (NSCs) integrate inputs from multiple sources to balance quiescence and activation. Notch signaling plays a key role during this process. Here, we report that Lunatic fringe (Lfng), a key modifier of the Notch receptor, is selectively expressed in NSCs. Further, Lfng in NSCs and Notch ligands Delta1 and Jagged1, expressed by their progeny, together influence NSC recruitment, cell cycle duration, and terminal fate. We propose a new model in which Lfng-mediated Notch signaling enables direct communication between a NSC and its descendants, so that progeny can send feedback signals to the 'mother' cell to modify its cell cycle status. Lfng-mediated Notch signaling appears to be a key factor governing NSC quiescence, division, and fate.


Asunto(s)
Glicosiltransferasas/metabolismo , Hipocampo/fisiología , Células-Madre Neurales/fisiología , Receptores Notch/metabolismo , Transducción de Señal , Animales , Ciclo Celular , Proliferación Celular , Regulación de la Expresión Génica , Ratones
16.
Nat Commun ; 8: 16087, 2017 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-28703127

RESUMEN

Glutathione plays many important roles in biological processes; however, the dynamic changes of glutathione concentrations in living cells remain largely unknown. Here, we report a reversible reaction-based fluorescent probe-designated as RealThiol (RT)-that can quantitatively monitor the real-time glutathione dynamics in living cells. Using RT, we observe enhanced antioxidant capability of activated neurons and dynamic glutathione changes during ferroptosis. RT is thus a versatile tool that can be used for both confocal microscopy and flow cytometry based high-throughput quantification of glutathione levels in single cells. We envision that this new glutathione probe will enable opportunities to study glutathione dynamics and transportation and expand our understanding of the physiological and pathological roles of glutathione in living cells.


Asunto(s)
Colorantes Fluorescentes , Fluorometría/métodos , Glutatión/análisis , Glutatión/química , Células HeLa , Humanos , Cinética , Análisis de la Célula Individual
17.
Sci Rep ; 6: 21264, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26887909

RESUMEN

Efficient gene editing is essential to fully utilize human pluripotent stem cells (hPSCs) in regenerative medicine. Custom endonuclease-based gene targeting involves two mechanisms of DNA repair: homology directed repair (HDR) and non-homologous end joining (NHEJ). HDR is the preferred mechanism for common applications such knock-in, knock-out or precise mutagenesis, but remains inefficient in hPSCs. Here, we demonstrate that synchronizing synchronizing hPSCs in G2/M with ABT phase increases on-target gene editing, defined as correct targeting cassette integration, 3 to 6 fold. We observed improved efficiency using ZFNs, TALENs, two CRISPR/Cas9, and CRISPR/Cas9 nickase to target five genes in three hPSC lines: three human embryonic stem cell lines, neural progenitors and diabetic iPSCs. neural progenitors and diabetic iPSCs. Reversible synchronization has no effect on pluripotency or differentiation. The increase in on-target gene editing is locus-independent and specific to the cell cycle phase as G2/M phase enriched cells show a 6-fold increase in targeting efficiency compared to cells in G1 phase. Concurrently inhibiting NHEJ with SCR7 does not increase HDR or improve gene targeting efficiency further, indicating that HR is the major DNA repair mechanism after G2/M phase arrest. The approach outlined here makes gene editing in hPSCs a more viable tool for disease modeling, regenerative medicine and cell-based therapies.


Asunto(s)
Sistemas CRISPR-Cas , División Celular , Fase G2 , Marcación de Gen/métodos , Células Madre Pluripotentes/metabolismo , Reparación del ADN por Recombinación , Línea Celular , Humanos , Células Madre Pluripotentes/citología
18.
PLoS One ; 11(10): e0164407, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27732649

RESUMEN

Thyroid hormone (TH) receptors (TRs α and ß) are homologous ligand-dependent transcription factors (TFs). While the TRs display distinct actions in development, metabolic regulation and other processes, comparisons of TRα and TRß dependent gene regulation mostly reveal similar mechanisms of action and few TR subtype specific genes. Here, we show that TRα predominates in multipotent human adipose derived stem cells (hADSC) whereas TRß is expressed at lower levels and is upregulated during hADSC differentiation. The TRs display several unusual properties in parental hADSC. First, TRs display predominantly cytoplasmic intracellular distribution and major TRα variants TRα1 and TRα2 colocalize with mitochondria. Second, knockdown experiments reveal that endogenous TRs influence hADSC cell morphology and expression of hundreds of genes in the absence of hormone, but do not respond to exogenous TH. Third, TRα and TRß affect hADSC in completely distinct ways; TRα regulates cell cycle associated processes while TRß may repress aspects of differentiation. TRα splice variant specific knockdown reveals that TRα1 and TRα2 both contribute to TRα-dependent gene expression in a gene specific manner. We propose that TRs work in a non-canonical and hormone independent manner in hADSC and that prominent subtype-specific activities emerge in the context of these unusual actions.


Asunto(s)
Tejido Adiposo/citología , Regulación del Desarrollo de la Expresión Génica , Células Madre/citología , Receptores alfa de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/metabolismo , Ciclo Celular , Diferenciación Celular , Línea Celular , Silenciador del Gen , Humanos , Células Madre/metabolismo , Receptores alfa de Hormona Tiroidea/análisis , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/análisis , Receptores beta de Hormona Tiroidea/genética , Triyodotironina/análisis , Triyodotironina/genética , Triyodotironina/metabolismo
19.
Ann N Y Acad Sci ; 1048: 185-97, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16154932

RESUMEN

Short-term plasticity was studied on synapses to Purkinje cells (PC): paired-pulse facilitation in parallel fibers (PF) and paired-pulse depression in climbing fibers (CF). Both phenomena relate to synaptic strength. These forms of short-term plasticity were tested on cerebellar slices in rat by early postnatal synchronous stimulation of olivary neurons (i.e., CFs) with harmaline and by inhibition of a metabotropic glutamate receptor (mGluR) as well as in mice that were deficient in the extracellular matrix glycoprotein tenascin-C. Harmaline stimulation delayed the developmental competition between CF inputs and maintained multiple innervation. Paired-pulse depression of the CF-PC synapse after harmaline treatment was more expressed. However, paired-pulse facilitation in PF-PC synapses remained unchanged. Electrophysiological responses of postsynaptic mGluR1 in CF-PC synapses could be obtained only with AMPA receptors blocked and glutamate uptake impaired. The mGluR1-specific antagonist CPCCOEt suppressed the CF-mGluR EPSC in some PCs and potentiated it in other PCs. CF paired-pulse depression was not changed with CPCCOEt, thus excluding a presynaptic effect. The postsynaptic effect was underlined by CPCCOEt-induced rise in amplitude of EPSC and by a prolongation of its decay time. Tenascins are extracellular matrix glycoproteins that may restrict the regenerative capacity of the nervous tissue. Testing short-term presynaptic plasticity in tenascin-C-deficient mice showed that CF paired-pulse depression was less expressed while PF paired-pulse facilitation was augmented except in a group of cells where there was even depression. The results underline differences in forms of short-term plasticity with regard to susceptibility to diverse modulatory factors.


Asunto(s)
Cerebelo/patología , Trastorno Depresivo/patología , Proteínas de la Matriz Extracelular/metabolismo , Plasticidad Neuronal/fisiología , Tenascina/metabolismo , Animales , Cerebelo/metabolismo , Trastorno Depresivo/metabolismo , Electrofisiología , Proteínas de la Matriz Extracelular/deficiencia , Mutación , Fibras Nerviosas/fisiología , Células de Purkinje/fisiología , Ratas , Ratas Mutantes , Receptores de Glutamato Metabotrópico/metabolismo , Sinapsis/fisiología , Tenascina/deficiencia
20.
Ann N Y Acad Sci ; 1048: 437-40, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16154971

RESUMEN

Scotopic electroretinogram of dogfish shark (Scylliorhinus canicula) and eel (Anguilla anguilla) is characterized by a negative off-response, changing in sign under photopic condition. It increased under the effect of increased background illumination, but its amplitude never exceeded that of the b-wave. On the other hand, dark-adapted electroretinograms of two perch-like species, perch (Perca fluviatilis) and painted comber (Serranus scriba), exhibited a positive off-wave, exceeding the b-wave amplitude under bright photopic conditions.


Asunto(s)
Adaptación Ocular/fisiología , Cazón/fisiología , Anguilas/fisiología , Electrorretinografía , Estimulación Luminosa , Animales , Adaptación a la Oscuridad/fisiología , Peces , Iluminación , Umbral Sensorial/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA