Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cell ; 187(9): 2143-2157.e15, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38670072

RESUMO

A central question for regenerative neuroscience is whether synthetic neural circuits, such as those built from two species, can function in an intact brain. Here, we apply blastocyst complementation to selectively build and test interspecies neural circuits. Despite approximately 10-20 million years of evolution, and prominent species differences in brain size, rat pluripotent stem cells injected into mouse blastocysts develop and persist throughout the mouse brain. Unexpectedly, the mouse niche reprograms the birth dates of rat neurons in the cortex and hippocampus, supporting rat-mouse synaptic activity. When mouse olfactory neurons are genetically silenced or killed, rat neurons restore information flow to odor processing circuits. Moreover, they rescue the primal behavior of food seeking, although less well than mouse neurons. By revealing that a mouse can sense the world using neurons from another species, we establish neural blastocyst complementation as a powerful tool to identify conserved mechanisms of brain development, plasticity, and repair.


Assuntos
Neurônios , Animais , Camundongos , Ratos , Neurônios/metabolismo , Neurônios/citologia , Neurônios/fisiologia , Blastocisto/metabolismo , Blastocisto/citologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , Encéfalo/citologia , Encéfalo/fisiologia , Feminino , Hipocampo/citologia , Hipocampo/fisiologia , Especificidade da Espécie , Camundongos Endogâmicos C57BL , Masculino
2.
Cell ; 175(7): 1796-1810.e20, 2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30528432

RESUMO

The 9p21.3 cardiovascular disease locus is the most influential common genetic risk factor for coronary artery disease (CAD), accounting for ∼10%-15% of disease in non-African populations. The ∼60 kb risk haplotype is human-specific and lacks coding genes, hindering efforts to decipher its function. Here, we produce induced pluripotent stem cells (iPSCs) from risk and non-risk individuals, delete each haplotype using genome editing, and generate vascular smooth muscle cells (VSMCs). Risk VSMCs exhibit globally altered transcriptional networks that intersect with previously identified CAD risk genes and pathways, concomitant with aberrant adhesion, contraction, and proliferation. Unexpectedly, deleting the risk haplotype rescues VSMC stability, while expressing the 9p21.3-associated long non-coding RNA ANRIL induces risk phenotypes in non-risk VSMCs. This study shows that the risk haplotype selectively predisposes VSMCs to adopt a cell state associated with CAD phenotypes, defines new VSMC-based networks of CAD risk genes, and establishes haplotype-edited iPSCs as powerful tools for functionally annotating the human genome.


Assuntos
Cromossomos Humanos Par 9 , Doença da Artéria Coronariana , Edição de Genes , Haplótipos , Células-Tronco Pluripotentes Induzidas , Polimorfismo de Nucleotídeo Único , Idoso , Idoso de 80 Anos ou mais , Cromossomos Humanos Par 9/genética , Cromossomos Humanos Par 9/metabolismo , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/metabolismo , Doença da Artéria Coronariana/patologia , Feminino , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Pessoa de Meia-Idade , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transcrição Gênica
3.
Nature ; 557(7705): 375-380, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29743677

RESUMO

The transcriptional programs that establish neuronal identity evolved to produce the rich diversity of neuronal cell types that arise sequentially during development. Remarkably, transient expression of certain transcription factors can also endow non-neural cells with neuronal properties. The relationship between reprogramming factors and the transcriptional networks that produce neuronal identity and diversity remains largely unknown. Here, from a screen of 598 pairs of transcription factors, we identify 76 pairs of transcription factors that induce mouse fibroblasts to differentiate into cells with neuronal features. By comparing the transcriptomes of these induced neuronal cells (iN cells) with those of endogenous neurons, we define a 'core' cell-autonomous neuronal signature. The iN cells also exhibit diversity; each transcription factor pair produces iN cells with unique transcriptional patterns that can predict their pharmacological responses. By linking distinct transcription factor input 'codes' to defined transcriptional outputs, this study delineates cell-autonomous features of neuronal identity and diversity and expands the reprogramming toolbox to facilitate engineering of induced neurons with desired patterns of gene expression and related functional properties.


Assuntos
Reprogramação Celular/genética , Neurônios/citologia , Neurônios/metabolismo , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Neurônios/efeitos dos fármacos , Análise de Sequência de RNA , Análise de Célula Única , Fatores de Transcrição/metabolismo , Transcriptoma/genética
4.
J Biol Chem ; 294(6): 1846-1859, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30552117

RESUMO

Friedreich ataxia (FRDA) is a neurodegenerative disorder caused by transcriptional silencing of the frataxin (FXN) gene, resulting in loss of the essential mitochondrial protein frataxin. Based on the knowledge that a GAA·TTC repeat expansion in the first intron of FXN induces heterochromatin, we previously showed that 2-aminobenzamide-type histone deacetylase inhibitors (HDACi) increase FXN mRNA levels in induced pluripotent stem cell (iPSC)-derived FRDA neurons and in circulating lymphocytes from patients after HDACi oral administration. How the reduced expression of frataxin leads to neurological and other systemic symptoms in FRDA patients remains unclear. Similar to other triplet-repeat disorders, it is unknown why FRDA affects only specific cell types, primarily the large sensory neurons of the dorsal root ganglia and cardiomyocytes. The combination of iPSC technology and genome-editing techniques offers the unique possibility to address these questions in a relevant cell model of FRDA, obviating confounding effects of variable genetic backgrounds. Here, using "scarless" gene-editing methods, we created isogenic iPSC lines that differ only in the length of the GAA·TTC repeats. To uncover the gene expression signatures due to the GAA·TTC repeat expansion in FRDA neuronal cells and the effect of HDACi on these changes, we performed RNA-seq-based transcriptomic analysis of iPSC-derived central nervous system (CNS) and isogenic sensory neurons. We found that cellular pathways related to neuronal function, regulation of transcription, extracellular matrix organization, and apoptosis are affected by frataxin loss in neurons of the CNS and peripheral nervous system and that these changes are partially restored by HDACi treatment.


Assuntos
Ataxia de Friedreich/genética , Inibidores de Histona Desacetilases/farmacologia , Neurônios/patologia , Transcriptoma , Células Cultivadas , Ataxia de Friedreich/patologia , Edição de Genes/métodos , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/química , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Neurônios/química , Expansão das Repetições de Trinucleotídeos/genética , Frataxina
5.
EMBO J ; 34(11): 1445-55, 2015 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-25908841

RESUMO

The nervous system is comprised of a vast diversity of distinct neural cell types. Differences between neuronal subtypes drive the assembly of neuronal circuits and underlie the subtype specificity of many neurological diseases. Yet, because neurons are irreversibly post-mitotic and not readily available from patients, it has not been feasible to study specific subtypes of human neurons in larger numbers. A powerful means to study neuronal diversity and neurological disease is to establish methods to produce desired neuronal subtypes in vitro. Traditionally this has been accomplished by treating pluripotent or neural stem cells with growth factors and morphogens that recapitulate exogenous developmental signals. These approaches often require extended periods of culture, which can limit their utility. However, more recently, it has become possible to produce neurons directly from fibroblasts using transcription factors and/or microRNAs. This technique referred to as direct reprogramming or transdifferentiation has proven to be a rapid, robust, and reproducible method to generate mature neurons of many different subtypes from multiple cell sources. Here, we highlight recent advances in generating neurons of specific subtypes using direct reprogramming and outline various scenarios in which induced neurons may be applied to studies of neuronal function and neurological disease.


Assuntos
Engenharia Celular/métodos , Reprogramação Celular , Fibroblastos , MicroRNAs , Células-Tronco Neurais , Neurônios , Fatores de Transcrição , Animais , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , MicroRNAs/biossíntese , MicroRNAs/genética , Doenças do Sistema Nervoso/metabolismo , Doenças do Sistema Nervoso/terapia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
6.
Nature ; 472(7342): 217-20, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21451523

RESUMO

Sensory information may be represented in the brain by stereotyped mapping of axonal inputs or by patterning that varies between individuals. In olfaction, a stereotyped map is evident in the first sensory processing centre, the olfactory bulb (OB), where different odours elicit activity in unique combinatorial patterns of spatially invariant glomeruli. Activation of each glomerulus is relayed to higher cortical processing centres by a set of ∼20-50 'homotypic' mitral and tufted (MT) neurons. In the cortex, target neurons integrate information from multiple glomeruli to detect distinct features of chemically diverse odours. How this is accomplished remains unclear, perhaps because the cortical mapping of glomerular information by individual MT neurons has not been described. Here we use new viral tracing and three-dimensional brain reconstruction methods to compare the cortical projections of defined sets of MT neurons. We show that the gross-scale organization of the OB is preserved in the patterns of axonal projections to one processing centre yet reordered in another, suggesting that distinct coding strategies may operate in different targets. However, at the level of individual neurons neither glomerular order nor stereotypy is preserved in either region. Rather, homotypic MT neurons from the same glomerulus innervate broad regions that differ between individuals. Strikingly, even in the same animal, MT neurons exhibit extensive diversity in wiring; axons of homotypic MT pairs diverge from each other, emit primary branches at distinct locations and 70-90% of branches of homotypic and heterotypic pairs are non-overlapping. This pronounced reorganization of sensory maps in the cortex offers an anatomic substrate for expanded combinatorial integration of information from spatially distinct glomeruli and predicts an unanticipated role for diversification of otherwise similar output neurons.


Assuntos
Mapeamento Encefálico , Condutos Olfatórios/citologia , Condutos Olfatórios/fisiologia , Percepção Olfatória/fisiologia , Neurônios Receptores Olfatórios/citologia , Neurônios Receptores Olfatórios/fisiologia , Animais , Feminino , Masculino , Camundongos , Técnicas de Rastreamento Neuroanatômico , Odorantes/análise , Bulbo Olfatório/anatomia & histologia , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Condutos Olfatórios/anatomia & histologia , Sindbis virus/genética , Sindbis virus/fisiologia , Olfato/fisiologia
7.
Nature ; 461(7260): 91-4, 2009 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-19672243

RESUMO

Recent landmark experiments have shown that transient overexpression of a small number of transcription factors can reprogram differentiated cells into induced pluripotent stem (iPS) cells that resemble embryonic stem (ES) cells. These iPS cells hold great promise for medicine because they have the potential to generate patient-specific cell types for cell replacement therapy and produce in vitro models of disease, without requiring embryonic tissues or oocytes. Although current iPS cell lines resemble ES cells, they have not passed the most stringent test of pluripotency by generating full-term or adult mice in tetraploid complementation assays, raising questions as to whether they are sufficiently potent to generate all of the cell types in an organism. Whether this difference between iPS and ES cells reflects intrinsic limitations of direct reprogramming is not known. Here we report fertile adult mice derived entirely from iPS cells that we generated by inducible genetic reprogramming of mouse embryonic fibroblasts. Producing adult mice derived entirely from a reprogrammed fibroblast shows that all features of a differentiated cell can be restored to an embryonic level of pluripotency without exposure to unknown ooplasmic factors. Comparing these fully pluripotent iPS cell lines to less developmentally potent lines may reveal molecular markers of different pluripotent states. Furthermore, mice derived entirely from iPS cells will provide a new resource to assess the functional and genomic stability of cells and tissues derived from iPS cells, which is important to validate their utility in cell replacement therapy and research applications.


Assuntos
Envelhecimento , Células-Tronco Pluripotentes/fisiologia , Técnicas Reprodutivas , Animais , Desdiferenciação Celular , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Feminino , Fibroblastos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/citologia , Gravidez , Taxa de Sobrevida
8.
Commun Biol ; 4(1): 1026, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34471188

RESUMO

Autism arises in high and low-risk families. De novo mutation contributes to autism incidence in low-risk families as there is a higher incidence in the affected of the simplex families than in their unaffected siblings. But the extent of contribution in low-risk families cannot be determined solely from simplex families as they are a mixture of low and high-risk. The rate of de novo mutation in nearly pure populations of high-risk families, the multiplex families, has not previously been rigorously determined. Moreover, rates of de novo mutation have been underestimated from studies based on low resolution microarrays and whole exome sequencing. Here we report on findings from whole genome sequence (WGS) of both simplex families from the Simons Simplex Collection (SSC) and multiplex families from the Autism Genetic Resource Exchange (AGRE). After removing the multiplex samples with excessive cell-line genetic drift, we find that the contribution of de novo mutation in multiplex is significantly smaller than the contribution in simplex. We use WGS to provide high resolution CNV profiles and to analyze more than coding regions, and revise upward the rate in simplex autism due to an excess of de novo events targeting introns. Based on this study, we now estimate that de novo events contribute to 52-67% of cases of autism arising from low risk families, and 30-39% of cases of all autism.


Assuntos
Transtorno Autístico/epidemiologia , Predisposição Genética para Doença/genética , Mutação , Adulto , Transtorno do Espectro Autista , Transtorno Autístico/genética , Feminino , Humanos , Incidência , Masculino , Pessoa de Meia-Idade , New York/epidemiologia , Fatores de Risco , Adulto Jovem
9.
Nature ; 428(6978): 44-9, 2004 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-14990966

RESUMO

Cloning by nuclear transplantation has been successfully carried out in various mammals, including mice. Until now mice have not been cloned from post-mitotic cells such as neurons. Here, we have generated fertile mouse clones derived by transferring the nuclei of post-mitotic, olfactory sensory neurons into oocytes. These results indicate that the genome of a post-mitotic, terminally differentiated neuron can re-enter the cell cycle and be reprogrammed to a state of totipotency after nuclear transfer. Moreover, the pattern of odorant receptor gene expression and the organization of odorant receptor genes in cloned mice was indistinguishable from wild-type animals, indicating that irreversible changes to the DNA of olfactory neurons do not accompany receptor gene choice.


Assuntos
Clonagem de Organismos , Embrião de Mamíferos/citologia , Desenvolvimento Embrionário e Fetal , Neurônios Receptores Olfatórios/citologia , Células-Tronco Totipotentes/citologia , Animais , Núcleo Celular/genética , Embrião de Mamíferos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Técnicas de Transferência Nuclear , Neurônios Receptores Olfatórios/metabolismo , Oócitos/citologia , Poliploidia , Receptores Odorantes/metabolismo , Células-Tronco Totipotentes/metabolismo
10.
Stem Cell Res ; 46: 101803, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32442913

RESUMO

Human induced pluripotent stem cell (hiPSC) lines have previously been generated through the NHLBI sponsored NextGen program at nine individual study sites. Here, we examined the structural integrity of 506 hiPSC lines as determined by copy number variations (CNVs). We observed that 149 hiPSC lines acquired 258 CNVs relative to donor DNA. We identified six recurrent regions of CNVs on chromosomes 1, 2, 3, 16 and 20 that overlapped with cancer associated genes. Furthermore, the genes mapping to regions of acquired CNVs show an enrichment in cancer related biological processes (IL6 production) and signaling cascades (JNK cascade & NFκB cascade). The genomic region of instability on chr20 (chr20q11.2) includes transcriptomic signatures for cancer associated genes such as ID1, BCL2L1, TPX2, PDRG1 and HCK. Of these HCK shows statistically significant differential expression between carrier and non-carrier hiPSC lines. Overall, while a low level of genomic instability was observed in the NextGen generated hiPSC lines, the observation of structural instability in regions with known cancer associated genes substantiates the importance of systematic evaluation of genetic variations in hiPSCs before using them as disease/research models.


Assuntos
Células-Tronco Pluripotentes Induzidas , Diferenciação Celular , Variações do Número de Cópias de DNA/genética , Proteínas de Ligação a DNA , Instabilidade Genômica , Genômica , Humanos , National Heart, Lung, and Blood Institute (U.S.) , Estados Unidos
11.
Nat Biomed Eng ; 3(2): 137-146, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30911429

RESUMO

How common polymorphisms in noncoding genome regions can regulate cellular function remains largely unknown. Here we show that cardiac fibrosis, mimicked using a hydrogel with controllable stiffness, affects the regulation of the phenotypes of human cardiomyocytes by a portion of the long noncoding RNA ANRIL, the gene of which is located in the disease-associated 9p21 locus. In a physiological environment, cultured cardiomyocytes derived from induced pluripotent stem cells obtained from patients who are homozygous for cardiovascular-risk alleles (R/R cardiomyocytes) or from healthy individuals who are homozygous for nonrisk alleles contracted synchronously, independently of genotype. After hydrogel stiffening to mimic fibrosis, only the R/R cardiomyocytes exhibited asynchronous contractions. These effects were associated with increased expression of the short ANRIL isoform in R/R cardiomyocytes, which induced a c-Jun N-terminal kinase (JNK) phosphorylation-based mechanism that impaired gap junctions (particularly, loss of connexin-43 expression) following stiffening. Deletion of the risk locus or treatment with a JNK antagonist was sufficient to maintain gap junctions and prevent asynchronous contraction of cardiomyocytes. Our findings suggest that mechanical changes in the microenvironment of cardiomyocytes can activate the regulation of their function by noncoding loci.

12.
Nat Biotechnol ; 35(10): 960-968, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28892074

RESUMO

The reprogramming of differentiated cells into induced pluripotent stem cells (iPSCs) is usually achieved by exogenous induction of transcription by factors acting in the nucleus. In contrast, during development, signaling pathways initiated at the membrane induce differentiation. The central idea of this study is to identify antibodies that can catalyze cellular de-differentiation and nuclear reprogramming by acting at the cell surface. We screen a lentiviral library encoding ∼100 million secreted and membrane-bound single-chain antibodies and identify antibodies that can replace either Sox2 and Myc (c-Myc) or Oct4 during reprogramming of mouse embryonic fibroblasts into iPSCs. We show that one Sox2-replacing antibody antagonizes the membrane-associated protein Basp1, thereby de-repressing nuclear factors WT1, Esrrb and Lin28a (Lin28) independent of Sox2. By manipulating this pathway, we identify three methods to generate iPSCs. Our results establish unbiased selection from autocrine combinatorial antibody libraries as a robust method to discover new biologics and uncover membrane-to-nucleus signaling pathways that regulate pluripotency and cell fate.


Assuntos
Anticorpos/metabolismo , Reprogramação Celular , Técnicas de Química Combinatória , Animais , Comunicação Autócrina , Blastocisto/citologia , Proteínas de Ligação a Calmodulina/metabolismo , Reprogramação Celular/efeitos dos fármacos , Células Clonais , Proteínas do Citoesqueleto/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Fosforilação/efeitos dos fármacos , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Reprodutibilidade dos Testes , Fatores de Transcrição SOXB1/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Regulação para Cima/efeitos dos fármacos
13.
Nat Biotechnol ; 35(1): 69-74, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27941802

RESUMO

Induced pluripotent stem cells (iPSCs) are being pursued as a source of cells for autologous therapies, many of which will be aimed at aged patients. To explore the impact of age on iPSC quality, we produced iPSCs from blood cells of 16 donors aged 21-100. We find that iPSCs from older donors retain an epigenetic signature of age, which can be reduced through passaging. Clonal expansion via reprogramming also enables the discovery of somatic mutations present in individual donor cells, which are missed by bulk sequencing methods. We show that exomic mutations in iPSCs increase linearly with age, and all iPSC lines analyzed carry at least one gene-disrupting mutation, several of which have been associated with cancer or dysfunction. Unexpectedly, elderly donors (>90 yrs) harbor fewer mutations than predicted, likely due to a contracted blood progenitor pool. These studies establish that donor age is associated with an increased risk of abnormalities in iPSCs and will inform clinical development of reprogramming technology.


Assuntos
Envelhecimento/genética , Envelhecimento/patologia , Aberrações Cromossômicas , Epigênese Genética/genética , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Adulto , Idoso , Diferenciação Celular/genética , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transplante de Células-Tronco , Doadores de Tecidos , Adulto Jovem
14.
Neuron ; 93(6): 1359-1374.e6, 2017 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-28262416

RESUMO

Neural circuit wiring relies on selective synapse formation whereby a presynaptic release apparatus is matched with its cognate postsynaptic machinery. At metabotropic synapses, the molecular mechanisms underlying this process are poorly understood. In the mammalian retina, rod photoreceptors form selective contacts with rod ON-bipolar cells by aligning the presynaptic voltage-gated Ca2+ channel directing glutamate release (CaV1.4) with postsynaptic mGluR6 receptors. We show this coordination requires an extracellular protein, α2δ4, which complexes with CaV1.4 and the rod synaptogenic mediator, ELFN1, for trans-synaptic alignment with mGluR6. Eliminating α2δ4 in mice abolishes rod synaptogenesis and synaptic transmission to rod ON-bipolar cells, and disrupts postsynaptic mGluR6 clustering. We further find that in rods, α2δ4 is crucial for organizing synaptic ribbons and setting CaV1.4 voltage sensitivity. In cones, α2δ4 is essential for CaV1.4 function, but is not required for ribbon organization, synaptogenesis, or synaptic transmission. These findings offer insights into retinal pathologies associated with α2δ4 dysfunction.


Assuntos
Axônios/fisiologia , Canais de Cálcio Tipo L/fisiologia , Canais de Cálcio/fisiologia , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Transmissão Sináptica/fisiologia , Animais , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Proteínas do Tecido Nervoso/metabolismo , Receptores de Glutamato Metabotrópico/fisiologia , Células Bipolares da Retina/fisiologia , Células Fotorreceptoras Retinianas Cones/fisiologia , Sinapses/metabolismo
15.
Neuron ; 89(6): 1223-1236, 2016 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-26948891

RESUMO

Somatic mutation in neurons is linked to neurologic disease and implicated in cell-type diversification. However, the origin, extent, and patterns of genomic mutation in neurons remain unknown. We established a nuclear transfer method to clonally amplify the genomes of neurons from adult mice for whole-genome sequencing. Comprehensive mutation detection and independent validation revealed that individual neurons harbor ∼100 unique mutations from all classes but lack recurrent rearrangements. Most neurons contain at least one gene-disrupting mutation and rare (0-2) mobile element insertions. The frequency and gene bias of neuronal mutations differ from other lineages, potentially due to novel mechanisms governing postmitotic mutation. Fertile mice were cloned from several neurons, establishing the compatibility of mutated adult neuronal genomes with reprogramming to pluripotency and development.


Assuntos
Clonagem Molecular , Mutação/genética , Neurônios/fisiologia , Análise de Sequência de DNA , Fatores Etários , Animais , Animais Recém-Nascidos , Proteínas Relacionadas a Caderinas , Caderinas/genética , Caderinas/metabolismo , Divisão Celular/genética , Elementos de DNA Transponíveis/genética , Embrião de Mamíferos , Feminino , Humanos , Antígeno Ki-67/metabolismo , Camundongos , Camundongos Transgênicos , Repetições de Microssatélites/genética , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Transferência Nuclear , Bulbo Olfatório/citologia , Bulbo Olfatório/embriologia , Bulbo Olfatório/crescimento & desenvolvimento , Oócitos/fisiologia
16.
Stem Cells Dev ; 24(11): 1265-83, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25749371

RESUMO

The technology to convert adult human non-neural cells into neural lineages, through induced pluripotent stem cells (iPSCs), somatic cell nuclear transfer, and direct lineage reprogramming or transdifferentiation has progressed tremendously in recent years. Reprogramming-based approaches aimed at manipulating cellular identity have enormous potential for disease modeling, high-throughput drug screening, cell therapy, and personalized medicine. Human iPSC (hiPSC)-based cellular disease models have provided proof of principle evidence of the validity of this system. However, several challenges remain before patient-specific neurons produced by reprogramming can provide reliable insights into disease mechanisms or be efficiently applied to drug discovery and transplantation therapy. This review will first discuss limitations of currently available reprogramming-based methods in faithfully and reproducibly recapitulating disease pathology. Specifically, we will address issues such as culture heterogeneity, interline and inter-individual variability, and limitations of two-dimensional differentiation paradigms. Second, we will assess recent progress and the future prospects of reprogramming-based neurologic disease modeling. This includes three-dimensional disease modeling, advances in reprogramming technology, prescreening of hiPSCs and creating isogenic disease models using gene editing.


Assuntos
Reprogramação Celular , Terapia Genética/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Doenças Neurodegenerativas/terapia , Medicina Regenerativa/métodos , Animais , Transplante de Células/métodos , Humanos , Doenças Neurodegenerativas/patologia
17.
Nat Neurosci ; 18(1): 25-35, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25420069

RESUMO

Humans and mice detect pain, itch, temperature, pressure, stretch and limb position via signaling from peripheral sensory neurons. These neurons are divided into three functional classes (nociceptors/pruritoceptors, mechanoreceptors and proprioceptors) that are distinguished by their selective expression of TrkA, TrkB or TrkC receptors, respectively. We found that transiently coexpressing Brn3a with either Ngn1 or Ngn2 selectively reprogrammed human and mouse fibroblasts to acquire key properties of these three classes of sensory neurons. These induced sensory neurons (iSNs) were electrically active, exhibited distinct sensory neuron morphologies and matched the characteristic gene expression patterns of endogenous sensory neurons, including selective expression of Trk receptors. In addition, we found that calcium-imaging assays could identify subsets of iSNs that selectively responded to diverse ligands known to activate itch- and pain-sensing neurons. These results offer a simple and rapid means for producing genetically diverse human sensory neurons suitable for drug screening and mechanistic studies.


Assuntos
Fibroblastos/fisiologia , Sistema Nervoso Periférico/citologia , Células Receptoras Sensoriais/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Feminino , Fibroblastos/ultraestrutura , Humanos , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Nociceptores/ultraestrutura , Técnicas de Patch-Clamp , Sistema Nervoso Periférico/ultraestrutura , Gravidez , Receptor trkC/genética , Células Receptoras Sensoriais/ultraestrutura , Fator de Transcrição Brn-3A/genética , Fator de Transcrição Brn-3A/fisiologia
18.
Neuron ; 87(6): 1248-1260, 2015 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-26402607

RESUMO

In the retina, rod and cone photoreceptors form distinct connections with different classes of downstream bipolar cells. However, the molecular mechanisms responsible for their selective connectivity are unknown. Here we identify a cell-adhesion protein, ELFN1, to be essential for the formation of synapses between rods and rod ON-bipolar cells in the primary rod pathway. ELFN1 is expressed selectively in rods where it is targeted to the axonal terminals by the synaptic release machinery. At the synapse, ELFN1 binds in trans to mGluR6, the postsynaptic receptor on rod ON-bipolar cells. Elimination of ELFN1 in mice prevents the formation of synaptic contacts involving rods, but not cones, allowing a dissection of the contributions of primary and secondary rod pathways to retinal circuit function and vision. We conclude that ELFN1 is necessary for the selective wiring of rods into the primary rod pathway and is required for high sensitivity of vision.


Assuntos
Rede Nervosa/fisiologia , Estimulação Luminosa/métodos , Células Fotorreceptoras Retinianas Bastonetes/fisiologia , Sinapses/fisiologia , Visão Ocular/fisiologia , Animais , Células HEK293 , Humanos , Camundongos , Camundongos Knockout , Rede Nervosa/ultraestrutura , Ratos , Retina/fisiologia , Retina/ultraestrutura , Células Fotorreceptoras Retinianas Bastonetes/ultraestrutura , Sinapses/ultraestrutura
19.
Cell Stem Cell ; 17(1): 101-15, 2015 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-26140606

RESUMO

Cell-based therapies to treat retinal degeneration are now being tested in clinical trials. However, it is not known whether the source of stem cells is important for the production of differentiated cells suitable for transplantation. To test this, we generated induced pluripotent stem cells (iPSCs) from murine rod photoreceptors (r-iPSCs) and scored their ability to make retinae by using a standardized quantitative protocol called STEM-RET. We discovered that r-iPSCs more efficiently produced differentiated retinae than did embryonic stem cells (ESCs) or fibroblast-derived iPSCs (f-iPSCs). Retinae derived from f-iPSCs had fewer amacrine cells and other inner nuclear layer cells. Integrated epigenetic analysis showed that DNA methylation contributes to the defects in f-iPSC retinogenesis and that rod-specific CTCF insulator protein-binding sites may promote r-iPSC retinogenesis. Together, our data suggest that the source of stem cells is important for producing retinal neurons in three-dimensional (3D) organ cultures.


Assuntos
Epigênese Genética , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Retina/crescimento & desenvolvimento , Células Fotorreceptoras Retinianas Bastonetes/citologia , Células Fotorreceptoras Retinianas Bastonetes/metabolismo , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Linhagem Celular , Reprogramação Celular , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Retina/citologia , Retina/metabolismo , Degeneração Retiniana/genética , Degeneração Retiniana/patologia , Degeneração Retiniana/terapia
20.
J Vis Exp ; (69): e4003, 2012 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-23222420

RESUMO

The production of induced pluripotent stem cells (iPSCs) from somatic cells provides a means to create valuable tools for basic research and may also produce a source of patient-matched cells for regenerative therapies. iPSCs may be generated using multiple protocols and derived from multiple cell sources. Once generated, iPSCs are tested using a variety of assays including immunostaining for pluripotency markers, generation of three germ layers in embryoid bodies and teratomas, comparisons of gene expression with embryonic stem cells (ESCs) and production of chimeric mice with or without germline contribution(2). Importantly, iPSC lines that pass these tests still vary in their capacity to produce different differentiated cell types(2). This has made it difficult to establish which iPSC derivation protocols, donor cell sources or selection methods are most useful for different applications. The most stringent test of whether a stem cell line has sufficient developmental potential to generate all tissues required for survival of an organism (termed full pluripotency) is tetraploid embryo complementation (TEC)(3-5). Technically, TEC involves electrofusion of two-cell embryos to generate tetraploid (4n) one-cell embryos that can be cultured in vitro to the blastocyst stage(6). Diploid (2n) pluripotent stem cells (e.g. ESCs or iPSCs) are then injected into the blastocoel cavity of the tetraploid blastocyst and transferred to a recipient female for gestation (see Figure 1). The tetraploid component of the complemented embryo contributes almost exclusively to the extraembryonic tissues (placenta, yolk sac), whereas the diploid cells constitute the embryo proper, resulting in a fetus derived entirely from the injected stem cell line. Recently, we reported the derivation of iPSC lines that reproducibly generate adult mice via TEC(1). These iPSC lines give rise to viable pups with efficiencies of 5-13%, which is comparable to ESCs(3,4,7) and higher than that reported for most other iPSC lines(8-12). These reports show that direct reprogramming can produce fully pluripotent iPSCs that match ESCs in their developmental potential and efficiency of generating pups in TEC tests. At present, it is not clear what distinguishes between fully pluripotent iPSCs and less potent lines(13-15). Nor is it clear which reprogramming methods will produce these lines with the highest efficiency. Here we describe one method that produces fully pluripotent iPSCs and "all- iPSC" mice, which may be helpful for investigators wishing to compare the pluripotency of iPSC lines or establish the equivalence of different reprogramming methods.


Assuntos
Células-Tronco Pluripotentes/citologia , Animais , Técnicas Citológicas/métodos , Feminino , Camundongos , Gravidez , Pesquisa com Células-Tronco
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa