Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Methods Mol Biol ; 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38801498

RESUMO

Recent findings from studies involving astronauts and animal models indicate that microgravity increases immune cell activity and potentially alters the white and gray matter of the central nervous system (CNS). To further investigate the impact of microgravity on CNS cells, we established cultures of three-dimensional neural organoids containing isogenic microglia, the brain's resident immune cells, and sent them onboard the International Space Station. When using induced pluripotent stem cell (iPSC) lines from individuals affected by neuroinflammatory and neurodegenerative diseases such as multiple sclerosis (MS) and Parkinson's disease (PD), these cultures can provide novel insights into pathogenic pathways that may be exacerbated by microgravity. We have devised a cryovial culture strategy that enables organoids to be maintained through space travel and onboard the International Space Station (ISS) without the need for medium or carbon dioxide exchange. Here, we provide a comprehensive description of all the steps involved: generating various types of neural organoids, establishing long-term cultures, arranging plans for shipment to the Kennedy Space Center (KSC), and ultimately preparing organoids for launch into low-Earth orbit (LEO) and return to Earth for post-flight analyses.

2.
iScience ; 26(7): 107001, 2023 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-37534130

RESUMO

Population-based genome-wide association studies (GWAS) normally require a large sample size, which can be labor intensive and costly. Recently, we reported a human induced pluripotent stem cell (hiPSC) array-based GWAS method, identifying NDUFA4 as a host factor for Zika virus (ZIKV) infection. In this study, we extended our analysis to trophectoderm cells, which constitute one of the major routes of mother-to-fetus transmission of ZIKV during pregnancy. We differentiated hiPSCs from various donors into trophectoderm cells. We then infected cells carrying loss of function mutations in NDUFA4, harboring risk versus non-risk alleles of SNPs (rs917172 and rs12386620) or having deletions in the NDUFA4 cis-regulatory region with ZIKV. We found that loss/reduction of NDUFA4 suppressed ZIKV infection in trophectoderm cells. This study validated our published hiPSC array-based system as a useful platform for GWAS and confirmed the role of NDUFA4 as a susceptibility locus for ZIKV in disease-relevant trophectoderm cells.

3.
bioRxiv ; 2023 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-38234801

RESUMO

To explain why individuals exposed to identical stressors experience divergent clinical outcomes, we determine how molecular encoding of stress modifies genetic risk for brain disorders. Analysis of post-mortem brain (n=304) revealed 8557 stress-interactive expression quantitative trait loci (eQTLs) that dysregulate expression of 915 eGenes in response to stress, and lie in stress-related transcription factor binding sites. Response to stress is robust across experimental paradigms: up to 50% of stress-interactive eGenes validate in glucocorticoid treated hiPSC-derived neurons (n=39 donors). Stress-interactive eGenes show brain region- and cell type-specificity, and, in post-mortem brain, implicate glial and endothelial mechanisms. Stress dysregulates long-term expression of disorder risk genes in a genotype-dependent manner; stress-interactive transcriptomic imputation uncovered 139 novel genes conferring brain disorder risk only in the context of traumatic stress. Molecular stress-encoding explains individualized responses to traumatic stress; incorporating trauma into genomic studies of brain disorders is likely to improve diagnosis, prognosis, and drug discovery.

4.
Nat Neurosci ; 25(11): 1434-1445, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36266471

RESUMO

Post-traumatic stress disorder (PTSD) can develop following severe trauma, but the extent to which genetic and environmental risk factors contribute to individual clinical outcomes is unknown. Here, we compared transcriptional responses to hydrocortisone exposure in human induced pluripotent stem cell (hiPSC)-derived glutamatergic neurons and peripheral blood mononuclear cells (PBMCs) from combat veterans with PTSD (n = 19 hiPSC and n = 20 PBMC donors) and controls (n = 20 hiPSC and n = 20 PBMC donors). In neurons only, we observed diagnosis-specific glucocorticoid-induced changes in gene expression corresponding with PTSD-specific transcriptomic patterns found in human postmortem brains. We observed glucocorticoid hypersensitivity in PTSD neurons, and identified genes that contribute to this PTSD-dependent glucocorticoid response. We find evidence of a coregulated network of transcription factors that mediates glucocorticoid hyper-responsivity in PTSD. These findings suggest that induced neurons represent a platform for examining the molecular mechanisms underlying PTSD, identifying biomarkers of stress response, and conducting drug screening to identify new therapeutics.


Assuntos
Células-Tronco Pluripotentes Induzidas , Transtornos de Estresse Pós-Traumáticos , Humanos , Transtornos de Estresse Pós-Traumáticos/genética , Glucocorticoides/farmacologia , Leucócitos Mononucleares , Interação Gene-Ambiente , Células-Tronco Pluripotentes Induzidas/metabolismo , Expressão Gênica , Neurônios/metabolismo
5.
Cell Stem Cell ; 29(10): 1475-1490.e6, 2022 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-36206731

RESUMO

Population-based studies to identify disease-associated risk alleles typically require samples from a large number of individuals. Here, we report a human-induced pluripotent stem cell (hiPSC)-based screening strategy to link human genetics with viral infectivity. A genome-wide association study (GWAS) identified a cluster of single-nucleotide polymorphisms (SNPs) in a cis-regulatory region of the NDUFA4 gene, which was associated with susceptibility to Zika virus (ZIKV) infection. Loss of NDUFA4 led to decreased sensitivity to ZIKV, dengue virus, and SARS-CoV-2 infection. Isogenic hiPSC lines carrying non-risk alleles of SNPs or deletion of the cis-regulatory region lower sensitivity to viral infection. Mechanistic studies indicated that loss/reduction of NDUFA4 causes mitochondrial stress, which leads to the leakage of mtDNA and thereby upregulation of type I interferon signaling. This study provides proof-of-principle for the application of iPSC arrays in GWAS and identifies NDUFA4 as a previously unknown susceptibility locus for viral infection.


Assuntos
COVID-19 , Dengue , Complexo IV da Cadeia de Transporte de Elétrons , Infecção por Zika virus , Humanos , Alelos , COVID-19/genética , DNA Mitocondrial/metabolismo , Complexo IV da Cadeia de Transporte de Elétrons/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla , Células-Tronco Pluripotentes Induzidas/metabolismo , Interferon Tipo I/metabolismo , Polimorfismo de Nucleotídeo Único , SARS-CoV-2 , Zika virus , Infecção por Zika virus/genética , Dengue/genética
6.
iScience ; 25(4): 104153, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35434558

RESUMO

The sinoatrial node (SAN) is the primary pacemaker of the heart. The human SAN is poorly understood due to limited primary tissue access and limitations in robust in vitro derivation methods. We developed a dual SHOX2:GFP; MYH6:mCherry knockin human embryonic stem cell (hESC) reporter line, which allows the identification and purification of SAN-like cells. Using this line, we performed several rounds of chemical screens and developed an efficient strategy to generate and purify hESC-derived SAN-like cells (hESC-SAN). The derived hESC-SAN cells display molecular and electrophysiological characteristics of bona fide nodal cells, which allowed exploration of their transcriptional profile at single-cell level. In sum, our dual reporter system facilitated an effective strategy for deriving human SAN-like cells, which can potentially be used for future disease modeling and drug discovery.

7.
Nat Commun ; 13(1): 1590, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35338121

RESUMO

Drug discovery for diseases such as Parkinson's disease are impeded by the lack of screenable cellular phenotypes. We present an unbiased phenotypic profiling platform that combines automated cell culture, high-content imaging, Cell Painting, and deep learning. We applied this platform to primary fibroblasts from 91 Parkinson's disease patients and matched healthy controls, creating the largest publicly available Cell Painting image dataset to date at 48 terabytes. We use fixed weights from a convolutional deep neural network trained on ImageNet to generate deep embeddings from each image and train machine learning models to detect morphological disease phenotypes. Our platform's robustness and sensitivity allow the detection of individual-specific variation with high fidelity across batches and plate layouts. Lastly, our models confidently separate LRRK2 and sporadic Parkinson's disease lines from healthy controls (receiver operating characteristic area under curve 0.79 (0.08 standard deviation)), supporting the capacity of this platform for complex disease modeling and drug screening applications.


Assuntos
Aprendizado Profundo , Doença de Parkinson , Fibroblastos , Humanos , Aprendizado de Máquina , Redes Neurais de Computação
8.
Cell Mol Life Sci ; 79(2): 78, 2022 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-35044538

RESUMO

Three-dimensional (3D) in vitro culture systems using human induced pluripotent stem cells (hiPSCs) are useful tools to model neurodegenerative disease biology in physiologically relevant microenvironments. Though many successful biomaterials-based 3D model systems have been established for other neurogenerative diseases, such as Alzheimer's disease, relatively few exist for Parkinson's disease (PD) research. We employed tissue engineering approaches to construct a 3D silk scaffold-based platform for the culture of hiPSC-dopaminergic (DA) neurons derived from healthy individuals and PD patients harboring LRRK2 G2019S or GBA N370S mutations. We then compared results from protein, gene expression, and metabolic analyses obtained from two-dimensional (2D) and 3D culture systems. The 3D platform enabled the formation of dense dopamine neuronal network architectures and developed biological profiles both similar and distinct from 2D culture systems in healthy and PD disease lines. PD cultures developed in 3D platforms showed elevated levels of α-synuclein and alterations in purine metabolite profiles. Furthermore, computational network analysis of transcriptomic networks nominated several novel molecular interactions occurring in neurons from patients with mutations in LRRK2 and GBA. We conclude that the brain-like 3D system presented here is a realistic platform to interrogate molecular mechanisms underlying PD biology.


Assuntos
Neurônios Dopaminérgicos/patologia , Doença de Parkinson/patologia , Bioengenharia , Técnicas de Cultura de Células em Três Dimensões , Células Cultivadas , Neurônios Dopaminérgicos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/patologia , Neurogênese , Seda/química , Alicerces Teciduais/química
9.
J Neurochem ; 156(6): 867-879, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32865230

RESUMO

Mitochondria and releasable endoplasmic reticulum (ER) calcium modulate neuronal calcium signaling, and both change in Alzheimer's disease (AD). The releasable calcium stores in the ER are exaggerated in fibroblasts from AD patients and in multiple models of AD. The activity of the alpha-ketoglutarate dehydrogenase complex (KGDHC), a key mitochondrial enzyme complex, is diminished in brains from AD patients, and can be plausibly linked to plaques and tangles. Our previous studies in cell lines and mouse neurons demonstrate that reductions in KGDHC increase the ER releasable calcium stores. The goal of these studies was to test whether the relationship was true in human iPSC-derived neurons. Inhibition of KGDHC for one or 24 hr increased the ER releasable calcium store in human neurons by 69% and 144%, respectively. The effect was mitochondrial enzyme specific because inhibiting the pyruvate dehydrogenase complex, another key mitochondrial enzyme complex, diminished the ER releasable calcium stores. The link of KGDHC to ER releasable calcium stores was cell type specific as the interaction was not present in iPSC or neural stem cells. Thus, these studies in human neurons verify a link between KGDHC and releasable ER calcium stores, and support the use of human neurons to examine mechanisms and potential therapies for AD.


Assuntos
Cálcio/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Mitocôndrias/enzimologia , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Linhagem Celular , Retículo Endoplasmático/metabolismo , Humanos , Imuno-Histoquímica , Complexo Cetoglutarato Desidrogenase/metabolismo , Potássio/metabolismo , Complexo Piruvato Desidrogenase/metabolismo
10.
Nat Commun ; 11(1): 319, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31949159

RESUMO

Optimal functioning of neuronal networks is critical to the complex cognitive processes of memory and executive function that deteriorate in Alzheimer's disease (AD). Here we use cellular and animal models as well as human biospecimens to show that AD-related stressors mediate global disturbances in dynamic intra- and inter-neuronal networks through pathologic rewiring of the chaperome system into epichaperomes. These structures provide the backbone upon which proteome-wide connectivity, and in turn, protein networks become disturbed and ultimately dysfunctional. We introduce the term protein connectivity-based dysfunction (PCBD) to define this mechanism. Among most sensitive to PCBD are pathways with key roles in synaptic plasticity. We show at cellular and target organ levels that network connectivity and functional imbalances revert to normal levels upon epichaperome inhibition. In conclusion, we provide proof-of-principle to propose AD is a PCBDopathy, a disease of proteome-wide connectivity defects mediated by maladaptive epichaperomes.


Assuntos
Doença de Alzheimer/metabolismo , Hipocampo/metabolismo , Plasticidade Neuronal/fisiologia , Proteoma/metabolismo , Doença de Alzheimer/patologia , Animais , Encéfalo/patologia , Mapeamento Encefálico , Disfunção Cognitiva/metabolismo , Função Executiva/fisiologia , Feminino , Hipocampo/patologia , Humanos , Masculino , Memória/fisiologia , Camundongos , Vias Neurais
11.
Cell Stem Cell ; 25(1): 120-136.e10, 2019 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-31155483

RESUMO

Current challenges in capturing naive human pluripotent stem cells (hPSCs) suggest that the factors regulating human naive versus primed pluripotency remain incompletely defined. Here we demonstrate that the widely used Essential 8 minimal medium (E8) captures hPSCs at a naive-to-primed intermediate state of pluripotency expressing several naive-like developmental, bioenergetic, and epigenomic features despite providing primed-state-sustaining growth factor conditions. Transcriptionally, E8 hPSCs are marked by activated lipid biosynthesis and suppressed MAPK/TGF-ß gene expression, resulting in endogenous ERK inhibition. These features are dependent on lipid-free culture conditions and are lost upon lipid exposure, whereas short-term pharmacological ERK inhibition restores naive-to-primed intermediate traits even in the presence of lipids. Finally, we identify de novo lipogenesis as a common transcriptional signature of E8 hPSCs and the pre-implantation human epiblast in vivo. These findings implicate exogenous lipid availability in regulating human pluripotency and define E8 hPSCs as a stable, naive-to-primed intermediate (NPI) pluripotent state.


Assuntos
Blastocisto/citologia , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/fisiologia , Diferenciação Celular , Células Cultivadas , Meios de Cultura Livres de Soro , Células-Tronco Embrionárias , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Metabolismo dos Lipídeos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
12.
Cell Rep ; 14(9): 2166-2179, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26923592

RESUMO

Attenuated auto-lysosomal system has been associated with Alzheimer disease (AD), yet all underlying molecular mechanisms leading to this impairment are unknown. We show that the amino acid sensing of mechanistic target of rapamycin complex 1 (mTORC1) is dysregulated in cells deficient in presenilin, a protein associated with AD. In these cells, mTORC1 is constitutively tethered to lysosomal membranes, unresponsive to starvation, and inhibitory to TFEB-mediated clearance due to a reduction in Sestrin2 expression. Normalization of Sestrin2 levels through overexpression or elevation of nuclear calcium rescued mTORC1 tethering and initiated clearance. While CLEAR network attenuation in vivo results in buildup of amyloid, phospho-Tau, and neurodegeneration, presenilin-knockout fibroblasts and iPSC-derived AD human neurons fail to effectively initiate autophagy. These results propose an altered mechanism for nutrient sensing in presenilin deficiency and underline an importance of clearance pathways in the onset of AD.


Assuntos
Presenilinas/genética , Doença de Alzheimer/metabolismo , Aminoácidos/metabolismo , Animais , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Cálcio/metabolismo , Núcleo Celular/metabolismo , Células Cultivadas , Redes Reguladoras de Genes , Humanos , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Fenômenos Fisiológicos da Nutrição , Peroxidases , Presenilinas/metabolismo , Serina-Treonina Quinases TOR/metabolismo
13.
Nat Cell Biol ; 18(3): 236-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26911906

RESUMO

Yaffe and colleagues discuss the issues surrounding the authentication and quality of induced pluripotent stem cells.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes/citologia , Transplante de Células-Tronco , Animais , Linhagem Celular , Humanos
14.
Nat Methods ; 12(9): 885-92, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26237226

RESUMO

Induced pluripotent stem cells (iPSCs) are an essential tool for modeling how causal genetic variants impact cellular function in disease, as well as an emerging source of tissue for regenerative medicine. The preparation of somatic cells, their reprogramming and the subsequent verification of iPSC pluripotency are laborious, manual processes limiting the scale and reproducibility of this technology. Here we describe a modular, robotic platform for iPSC reprogramming enabling automated, high-throughput conversion of skin biopsies into iPSCs and differentiated cells with minimal manual intervention. We demonstrate that automated reprogramming and the pooled selection of polyclonal pluripotent cells results in high-quality, stable iPSCs. These lines display less line-to-line variation than either manually produced lines or lines produced through automation followed by single-colony subcloning. The robotic platform we describe will enable the application of iPSCs to population-scale biomedical problems including the study of complex genetic diseases and the development of personalized medicines.


Assuntos
Técnicas de Cultura Celular por Lotes/instrumentação , Separação Celular/instrumentação , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Técnicas Analíticas Microfluídicas/instrumentação , Robótica/instrumentação , Diferenciação Celular/fisiologia , Células Cultivadas , Desenho de Equipamento , Análise de Falha de Equipamento , Fibroblastos/citologia , Fibroblastos/fisiologia , Humanos
15.
Am J Physiol Cell Physiol ; 308(3): C209-19, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25394470

RESUMO

Production and isolation of forebrain interneuron progenitors are essential for understanding cortical development and developing cell-based therapies for developmental and neurodegenerative disorders. We demonstrate production of a population of putative calretinin-positive bipolar interneurons that express markers consistent with caudal ganglionic eminence identities. Using serum-free embryoid bodies (SFEBs) generated from human inducible pluripotent stem cells (iPSCs), we demonstrate that these interneuron progenitors exhibit morphological, immunocytochemical, and electrophysiological hallmarks of developing cortical interneurons. Finally, we develop a fluorescence-activated cell-sorting strategy to isolate interneuron progenitors from SFEBs to allow development of a purified population of these cells. Identification of this critical neuronal cell type within iPSC-derived SFEBs is an important and novel step in describing cortical development in this iPSC preparation.


Assuntos
Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Corpos Embrioides/fisiologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Interneurônios/fisiologia , Animais , Células Cultivadas , Fibroblastos/fisiologia , Humanos , Camundongos , Camundongos Knockout
16.
Hum Mol Genet ; 24(5): 1420-31, 2015 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-25398950

RESUMO

Giant axonal neuropathy (GAN) is a progressive neurodegenerative disease caused by autosomal recessive mutations in the GAN gene resulting in a loss of a ubiquitously expressed protein, gigaxonin. Gene replacement therapy is a promising strategy for treatment of the disease; however, the effectiveness and safety of gigaxonin reintroduction have not been tested in human GAN nerve cells. Here we report the derivation of induced pluripotent stem cells (iPSCs) from three GAN patients with different GAN mutations. Motor neurons differentiated from GAN iPSCs exhibit accumulation of neurofilament (NF-L) and peripherin (PRPH) protein and formation of PRPH aggregates, the key pathological phenotypes observed in patients. Introduction of gigaxonin either using a lentiviral vector or as a stable transgene resulted in normalization of NEFL and PRPH levels in GAN neurons and disappearance of PRPH aggregates. Importantly, overexpression of gigaxonin had no adverse effect on survival of GAN neurons, supporting the feasibility of gene replacement therapy. Our findings demonstrate that GAN iPSCs provide a novel model for studying human GAN neuropathologies and for the development and testing of new therapies in relevant cell types.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Neuropatia Axonal Gigante/genética , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas de Filamentos Intermediários/genética , Neurônios Motores/metabolismo , Axônios , Diferenciação Celular , Células Cultivadas , Proteínas do Citoesqueleto/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Terapia Genética/métodos , Vetores Genéticos/genética , Neuropatia Axonal Gigante/terapia , Humanos , Filamentos Intermediários/genética , Filamentos Intermediários/metabolismo , Cariotipagem , Lentivirus/genética , Neurônios Motores/citologia , Mutação , Fenótipo
17.
Cell Rep ; 9(4): 1173-82, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25456120

RESUMO

Parkinson's disease (PD) has been attributed to a combination of genetic and nongenetic factors. We studied a set of monozygotic twins harboring the heterozygous glucocerebrosidase mutation (GBA N370S) but clinically discordant for PD. We applied induced pluripotent stem cell (iPSC) technology for PD disease modeling using the twins' fibroblasts to evaluate and dissect the genetic and nongenetic contributions. Utilizing fluorescence-activated cell sorting, we obtained a homogenous population of "footprint-free" iPSC-derived midbrain dopaminergic (mDA) neurons. The mDA neurons from both twins had ∼50% GBA enzymatic activity, ∼3-fold elevated α-synuclein protein levels, and a reduced capacity to synthesize and release dopamine. Interestingly, the affected twin's neurons showed an even lower dopamine level, increased monoamine oxidase B (MAO-B) expression, and impaired intrinsic network activity. Overexpression of wild-type GBA and treatment with MAO-B inhibitors normalized α-synuclein and dopamine levels, suggesting a combination therapy for the affected twin.


Assuntos
Neurônios Dopaminérgicos/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Doença de Parkinson/patologia , Gêmeos Monozigóticos , Biomarcadores/metabolismo , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/enzimologia , Citometria de Fluxo , Glucosilceramidase/genética , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Masculino , Monoaminoxidase/metabolismo , Inibidores da Monoaminoxidase/farmacologia , Mutação/genética , Doença de Parkinson/enzimologia , Fenótipo , Análise de Sequência de RNA , alfa-Sinucleína/metabolismo
18.
PLoS One ; 9(7): e103418, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25072157

RESUMO

Many protocols have been designed to differentiate human embryonic stem cells (ESCs) and human induced pluripotent stem cells (iPSCs) into neurons. Despite the relevance of electrophysiological properties for proper neuronal function, little is known about the evolution over time of important neuronal electrophysiological parameters in iPSC-derived neurons. Yet, understanding the development of basic electrophysiological characteristics of iPSC-derived neurons is critical for evaluating their usefulness in basic and translational research. Therefore, we analyzed the basic electrophysiological parameters of forebrain neurons differentiated from human iPSCs, from day 31 to day 55 after the initiation of neuronal differentiation. We assayed the developmental progression of various properties, including resting membrane potential, action potential, sodium and potassium channel currents, somatic calcium transients and synaptic activity. During the maturation of iPSC-derived neurons, the resting membrane potential became more negative, the expression of voltage-gated sodium channels increased, the membrane became capable of generating action potentials following adequate depolarization and, at day 48-55, 50% of the cells were capable of firing action potentials in response to a prolonged depolarizing current step, of which 30% produced multiple action potentials. The percentage of cells exhibiting miniature excitatory post-synaptic currents increased over time with a significant increase in their frequency and amplitude. These changes were associated with an increase of Ca2+ transient frequency. Co-culturing iPSC-derived neurons with mouse glial cells enhanced the development of electrophysiological parameters as compared to pure iPSC-derived neuronal cultures. This study demonstrates the importance of properly evaluating the electrophysiological status of the newly generated neurons when using stem cell technology, as electrophysiological properties of iPSC-derived neurons mature over time.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Neurônios/citologia , Neurônios/fisiologia , Animais , Cálcio/metabolismo , Técnicas de Cocultura , Fenômenos Eletrofisiológicos , Humanos , Imunofenotipagem , Camundongos , Neuroglia , Técnicas de Patch-Clamp , Potenciais Sinápticos , Transmissão Sináptica , Fatores de Tempo
19.
Acta Neuropathol Commun ; 2: 4, 2014 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-24398250

RESUMO

BACKGROUND: Induced pluripotent stem cells (iPSCs) derived from patients with neurodegenerative disease generally lack neuropathological confirmation, the gold standard for disease classification and grading of severity. The use of tissue with a definitive neuropathological diagnosis would be an ideal source for iPSCs. The challenge to this approach is that the majority of biobanked brain tissue was not meant for growing live cells, and thus was not frozen in the presence of cryoprotectants such as DMSO. RESULTS: We report the generation of iPSCs from frozen non-cryoprotected dural tissue stored at -80°C for up to 11 years. This autopsy cohort included subjects with Alzheimer's disease and four other neurodegenerative diseases. CONCLUSIONS: Disease-specific iPSCs can be generated from readily available, archival biobanked tissue. This allows for rapid expansion of generating iPSCs with confirmed pathology as well as allowing access to rare patient variants that have been banked.


Assuntos
Dura-Máter/patologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Doença de Alzheimer/patologia , Animais , Antígenos de Superfície/metabolismo , Diferenciação Celular , Linhagem Celular Transformada/patologia , Proliferação de Células , Bases de Dados como Assunto , Fibroblastos/metabolismo , Fibroblastos/virologia , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Proteína Homeobox Nanog , Doenças Neurodegenerativas/patologia , Fator 3 de Transcrição de Octâmero/metabolismo , Mudanças Depois da Morte , Proteoglicanas/metabolismo , Pele/citologia , Antígenos Embrionários Estágio-Específicos/metabolismo
20.
PLoS One ; 9(1): e84547, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24416243

RESUMO

Presenilin 1 (PSEN1) encodes the catalytic subunit of γ-secretase, and PSEN1 mutations are the most common cause of early onset familial Alzheimer's disease (FAD). In order to elucidate pathways downstream of PSEN1, we characterized neural progenitor cells (NPCs) derived from FAD mutant PSEN1 subjects. Thus, we generated induced pluripotent stem cells (iPSCs) from affected and unaffected individuals from two families carrying PSEN1 mutations. PSEN1 mutant fibroblasts, and NPCs produced greater ratios of Aß42 to Aß40 relative to their control counterparts, with the elevated ratio even more apparent in PSEN1 NPCs than in fibroblasts. Molecular profiling identified 14 genes differentially-regulated in PSEN1 NPCs relative to control NPCs. Five of these targets showed differential expression in late onset AD/Intermediate AD pathology brains. Therefore, in our PSEN1 iPSC model, we have reconstituted an essential feature in the molecular pathogenesis of FAD, increased generation of Aß42/40, and have characterized novel expression changes.


Assuntos
Doença de Alzheimer/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/metabolismo , Presenilina-1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Peptídeos beta-Amiloides/biossíntese , Animais , Apolipoproteínas E/genética , Proteínas Reguladoras de Apoptose , Sequência de Bases , Encéfalo/citologia , Encéfalo/patologia , Diferenciação Celular , Linhagem Celular , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Genótipo , Humanos , Mutação , Proteínas do Tecido Nervoso/genética , Neurônios/citologia , Neurônios/patologia , Fragmentos de Peptídeos/biossíntese , Presenilina-1/genética , Ratos , Proteínas Supressoras da Sinalização de Citocina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA