Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell ; 185(15): 2756-2769, 2022 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-35868278

RESUMO

For decades, insight into fundamental principles of human biology and disease has been obtained primarily by experiments in animal models. While this has allowed researchers to understand many human biological processes in great detail, some developmental and disease mechanisms have proven difficult to study due to inherent species differences. The advent of organoid technology more than 10 years ago has established laboratory-grown organ tissues as an additional model system to recapitulate human-specific aspects of biology. The use of human 3D organoids, as well as other advances in single-cell technologies, has revealed unprecedented insights into human biology and disease mechanisms, especially those that distinguish humans from other species. This review highlights novel advances in organoid biology with a focus on how organoid technology has generated a better understanding of human-specific processes in development and disease.


Assuntos
Modelos Biológicos , Organoides , Animais , Humanos
2.
Nature ; 621(7978): 373-380, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37704762

RESUMO

The development of the human brain involves unique processes (not observed in many other species) that can contribute to neurodevelopmental disorders1-4. Cerebral organoids enable the study of neurodevelopmental disorders in a human context. We have developed the CRISPR-human organoids-single-cell RNA sequencing (CHOOSE) system, which uses verified pairs of guide RNAs, inducible CRISPR-Cas9-based genetic disruption and single-cell transcriptomics for pooled loss-of-function screening in mosaic organoids. Here we show that perturbation of 36 high-risk autism spectrum disorder genes related to transcriptional regulation uncovers their effects on cell fate determination. We find that dorsal intermediate progenitors, ventral progenitors and upper-layer excitatory neurons are among the most vulnerable cell types. We construct a developmental gene regulatory network of cerebral organoids from single-cell transcriptomes and chromatin modalities and identify autism spectrum disorder-associated and perturbation-enriched regulatory modules. Perturbing members of the BRG1/BRM-associated factor (BAF) chromatin remodelling complex leads to enrichment of ventral telencephalon progenitors. Specifically, mutating the BAF subunit ARID1B affects the fate transition of progenitors to oligodendrocyte and interneuron precursor cells, a phenotype that we confirmed in patient-specific induced pluripotent stem cell-derived organoids. Our study paves the way for high-throughput phenotypic characterization of disease susceptibility genes in organoid models with cell state, molecular pathway and gene regulatory network readouts.


Assuntos
Transtorno do Espectro Autista , Encéfalo , Deficiências do Desenvolvimento , Organoides , Análise da Expressão Gênica de Célula Única , Humanos , Transtorno do Espectro Autista/complicações , Transtorno do Espectro Autista/genética , Transtorno do Espectro Autista/patologia , Transtorno Autístico/complicações , Transtorno Autístico/genética , Transtorno Autístico/patologia , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem da Célula/genética , Cromatina/genética , Proteína 9 Associada à CRISPR/metabolismo , Sistemas CRISPR-Cas , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/genética , Deficiências do Desenvolvimento/patologia , Edição de Genes , Mutação com Perda de Função , Mosaicismo , Neurônios/metabolismo , Neurônios/patologia , Organoides/citologia , Organoides/metabolismo , RNA Guia de Sistemas CRISPR-Cas , Transcrição Gênica
3.
EMBO J ; 42(22): e113213, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37842725

RESUMO

The establishment and maintenance of apical-basal polarity is a fundamental step in brain development, instructing the organization of neural progenitor cells (NPCs) and the developing cerebral cortex. Particularly, basally located extracellular matrix (ECM) is crucial for this process. In vitro, epithelial polarization can be achieved via endogenous ECM production, or exogenous ECM supplementation. While neuroepithelial development is recapitulated in neural organoids, the effects of different ECM sources in tissue morphogenesis remain underexplored. Here, we show that exposure to a solubilized basement membrane matrix substrate, Matrigel, at early neuroepithelial stages causes rapid tissue polarization and rearrangement of neuroepithelial architecture. In cultures exposed to pure ECM components or unexposed to any exogenous ECM, polarity acquisition is slower and driven by endogenous ECM production. After the onset of neurogenesis, tissue architecture and neuronal differentiation are largely independent of the initial ECM source, but Matrigel exposure has long-lasting effects on tissue patterning. These results advance the knowledge on mechanisms of exogenously and endogenously guided morphogenesis, demonstrating the self-sustainability of neuroepithelial cultures by endogenous processes.


Assuntos
Matriz Extracelular , Organoides , Humanos , Morfogênese
5.
Immunity ; 32(2): 240-52, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20153221

RESUMO

Injury to the central nervous system initiates an uncontrolled inflammatory response that results in both tissue repair and destruction. Here, we showed that, in rodents and humans, injury to the spinal cord triggered surface expression of CD95 ligand (CD95L, FasL) on peripheral blood myeloid cells. CD95L stimulation of CD95 on these cells activated phosphoinositide 3-kinase (PI3K) and metalloproteinase-9 (MMP-9) via recruitment and activation of Syk kinase, ultimately leading to increased migration. Exclusive CD95L deletion in myeloid cells greatly decreased the number of neutrophils and macrophages infiltrating the injured spinal cord or the inflamed peritoneum after thioglycollate injection. Importantly, deletion of myeloid CD95L, but not of CD95 on neural cells, led to functional recovery of spinal injured animals. Our results indicate that CD95L acts on peripheral myeloid cells to induce tissue damage. Thus, neutralization of CD95L should be considered as a means to create a controlled beneficial inflammatory response.


Assuntos
Movimento Celular , Proteína Ligante Fas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Células Mieloides/metabolismo , Peritonite/imunologia , Proteínas Tirosina Quinases/metabolismo , Animais , Células Cultivadas , Proteína Ligante Fas/genética , Proteína Ligante Fas/imunologia , Humanos , Inflamação , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Mieloides/imunologia , Células Mieloides/patologia , Peritônio/imunologia , Peritônio/patologia , Peritonite/induzido quimicamente , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Medula Espinal/imunologia , Medula Espinal/patologia , Quinase Syk , Tioglicolatos/administração & dosagem
6.
Cell Stem Cell ; 31(6): 866-885.e14, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38718796

RESUMO

Mutations in ARID1B, a member of the mSWI/SNF complex, cause severe neurodevelopmental phenotypes with elusive mechanisms in humans. The most common structural abnormality in the brain of ARID1B patients is agenesis of the corpus callosum (ACC), characterized by the absence of an interhemispheric white matter tract that connects distant cortical regions. Here, we find that neurons expressing SATB2, a determinant of callosal projection neuron (CPN) identity, show impaired maturation in ARID1B+/- neural organoids. Molecularly, a reduction in chromatin accessibility of genomic regions targeted by TCF-like, NFI-like, and ARID-like transcription factors drives the differential expression of genes required for corpus callosum (CC) development. Through an in vitro model of the CC tract, we demonstrate that this transcriptional dysregulation impairs the formation of long-range axonal projections, causing structural underconnectivity. Our study uncovers new functions of the mSWI/SNF during human corticogenesis, identifying cell-autonomous axonogenesis defects in SATB2+ neurons as a cause of ACC in ARID1B patients.


Assuntos
Axônios , Corpo Caloso , Proteínas de Ligação a DNA , Organoides , Fatores de Transcrição , Humanos , Corpo Caloso/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Organoides/metabolismo , Axônios/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/genética , Transcrição Gênica , Neurônios/metabolismo
7.
Science ; 375(6579): eabf5546, 2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-35084981

RESUMO

Evolutionary development of the human brain is characterized by the expansion of various brain regions. Here, we show that developmental processes specific to humans are responsible for malformations of cortical development (MCDs), which result in developmental delay and epilepsy in children. We generated a human cerebral organoid model for tuberous sclerosis complex (TSC) and identified a specific neural stem cell type, caudal late interneuron progenitor (CLIP) cells. In TSC, CLIP cells over-proliferate, generating excessive interneurons, brain tumors, and cortical malformations. Epidermal growth factor receptor inhibition reduces tumor burden, identifying potential treatment options for TSC and related disorders. The identification of CLIP cells reveals the extended interneuron generation in the human brain as a vulnerability for disease. In addition, this work demonstrates that analyzing MCDs can reveal fundamental insights into human-specific aspects of brain development.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/patologia , Interneurônios/citologia , Células-Tronco Neurais/fisiologia , Esclerose Tuberosa/genética , Esclerose Tuberosa/patologia , Encéfalo/embriologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Carcinogênese , Linhagem da Célula , Proliferação de Células , Progressão da Doença , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Perfilação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas , Interneurônios/fisiologia , Perda de Heterozigosidade , Células-Tronco Neurais/citologia , Organoides , RNA-Seq , Serina-Treonina Quinases TOR/metabolismo , Esclerose Tuberosa/tratamento farmacológico , Esclerose Tuberosa/metabolismo , Proteína 1 do Complexo Esclerose Tuberosa/genética , Proteína 1 do Complexo Esclerose Tuberosa/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo
8.
Cell Stem Cell ; 22(2): 143-145, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29451854

RESUMO

Neural stem cells in the ventricular-subventricular zone of the adult brain continuously generate differentiated neurons without depleting the stem cell pool. In this issue of Cell Stem Cell, Obernier et al. (2018) present the surprising finding that this occurs through mostly symmetric divisions that either generate two differentiating or two self-renewing daughter cells.


Assuntos
Divisão Celular , Neurogênese , Diferenciação Celular , Autorrenovação Celular , Células-Tronco Neurais
10.
Cell Stem Cell ; 22(4): 543-558.e12, 2018 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-29625069

RESUMO

Stem cell-specific transcriptional networks are well known to control pluripotency, but constitutive cellular processes such as mRNA splicing and protein synthesis can add complex layers of regulation with poorly understood effects on cell-fate decisions. Here, we show that the RNA binding protein HTATSF1 controls embryonic stem cell differentiation by regulating multiple aspects of RNA processing during ribosome biogenesis. HTATSF1, in a complex with splicing factor SF3B1, controls intron removal from ribosomal protein transcripts and regulates ribosomal RNA transcription and processing, thereby controlling 60S ribosomal abundance and protein synthesis. HTATSF1-dependent protein synthesis is essential for naive pre-implantation epiblast to transition into post-implantation epiblast, a stage with transiently low protein synthesis, and further differentiation toward neuroectoderm. Together, these results identify coordinated regulation of ribosomal RNA and protein synthesis by HTATSF1 and show that this essential mechanism controls protein synthesis during early mammalian embryogenesis.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/metabolismo , RNA Ribossômico/metabolismo , Animais , Humanos , Camundongos , Transativadores/metabolismo
11.
Nat Biotechnol ; 35(7): 659-666, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28562594

RESUMO

Three-dimensional cell culture models have either relied on the self-organizing properties of mammalian cells or used bioengineered constructs to arrange cells in an organ-like configuration. While self-organizing organoids excel at recapitulating early developmental events, bioengineered constructs reproducibly generate desired tissue architectures. Here, we combine these two approaches to reproducibly generate human forebrain tissue while maintaining its self-organizing capacity. We use poly(lactide-co-glycolide) copolymer (PLGA) fiber microfilaments as a floating scaffold to generate elongated embryoid bodies. Microfilament-engineered cerebral organoids (enCORs) display enhanced neuroectoderm formation and improved cortical development. Furthermore, reconstitution of the basement membrane leads to characteristic cortical tissue architecture, including formation of a polarized cortical plate and radial units. Thus, enCORs model the distinctive radial organization of the cerebral cortex and allow for the study of neuronal migration. Our data demonstrate that combining 3D cell culture with bioengineering can increase reproducibility and improve tissue architecture.


Assuntos
Técnicas de Cultura Celular por Lotes/métodos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Organoides/crescimento & desenvolvimento , Prosencéfalo/crescimento & desenvolvimento , Engenharia Tecidual/métodos , Células Cultivadas , Regeneração Tecidual Guiada/métodos , Humanos , Células-Tronco Neurais/citologia , Técnicas de Cultura de Órgãos/métodos , Organoides/citologia , Prosencéfalo/citologia
12.
Cell Stem Cell ; 12(2): 204-14, 2013 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-23395445

RESUMO

Memory impairment has been associated with age-related decline in adult hippocampal neurogenesis. Although Notch, bone morphogenetic protein, and Wnt signaling pathways are known to regulate multiple aspects of adult neural stem cell function, the molecular basis of declining neurogenesis in the aging hippocampus remains unknown. Here, we show that expression of the Wnt antagonist Dickkopf-1 (Dkk1) increases with age and that its loss enhances neurogenesis in the hippocampus. Neural progenitors with inducible loss of Dkk1 increase their Wnt activity, which leads to enhanced self-renewal and increased generation of immature neurons. This Wnt-expanded progeny subsequently matures into glutamatergic granule neurons with increased dendritic complexity. As a result, mice deficient in Dkk1 exhibit enhanced spatial working memory and memory consolidation and also show improvements in affective behavior. Taken together, our findings show that upregulating Wnt signaling by reducing Dkk1 expression can counteract age-related decrease in neurogenesis and its associated cognitive decline.


Assuntos
Cognição/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neurogênese/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Hipocampo/citologia , Peptídeos e Proteínas de Sinalização Intercelular/genética , Memória de Curto Prazo/fisiologia , Camundongos , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia
13.
PLoS One ; 6(2): e15786, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21347250

RESUMO

Dickkopf1 (Dkk1) is a Wnt/ß-catenin inhibitor that participates in many processes during embryonic development. One of its roles during embryogenesis is to induce head formation, since Dkk1-null mice lack head structures anterior to midbrain. The Wnt/ß-catenin pathway is also known to regulate different aspects of ventral midbrain (VM) dopaminergic (DA) neuron development and, in vitro, Dkk1-mediated inhibition of the Wnt/ß-catenin pathway improves the DA differentiation in mouse embryonic stem cells (mESC). However, the in vivo function of Dkk1 on the development of midbrain DA neurons remains to be elucidated. Here we examined Dkk1(+/-) embryos and found that Dkk1 is required for the differentiation of DA precursors/neuroblasts into DA neurons at E13.5. This deficit persisted until E17.5, when a defect in the number and distribution of VM DA neurons was detected. Furthermore, analysis of the few Dkk1(-/-) embryos that survived until E17.5 revealed a more severe loss of midbrain DA neurons and morphogenesis defects. Our results thus show that Dkk1 is required for midbrain DA differentiation and morphogenesis.


Assuntos
Diferenciação Celular , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Mesencéfalo/citologia , Mesencéfalo/crescimento & desenvolvimento , Morfogênese , Animais , Contagem de Células , Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mesencéfalo/metabolismo , Camundongos , Mutação , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo
14.
Cell Stem Cell ; 6(5): 403-4, 2010 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-20452311

RESUMO

The laminin receptor Integrin alpha6beta1 anchors adult neural stem cells to the niche vasculature. In this issue of Cell Stem Cell, Lathia et al. (2010) show that glioblastoma stem cells highly express integrin alpha6 and that their interaction with laminin on endothelial cells directly regulates their tumorigenic capacity.

15.
Cell Stem Cell ; 5(2): 178-90, 2009 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-19664992

RESUMO

Adult neurogenesis persists in the subventricular zone and the dentate gyrus and can be induced upon central nervous system injury. However, the final contribution of newborn neurons to neuronal networks is limited. Here we show that in neural stem cells, stimulation of the "death receptor" CD95 does not trigger apoptosis but unexpectedly leads to increased stem cell survival and neuronal specification. These effects are mediated via activation of the Src/PI3K/AKT/mTOR signaling pathway, ultimately leading to a global increase in protein translation. Induction of neurogenesis by CD95 was further confirmed in the ischemic CA1 region, in the naive dentate gyrus, and after forced expression of CD95L in the adult subventricular zone. Lack of hippocampal CD95 resulted in a reduction in neurogenesis and working memory deficits. Following global ischemia, CD95-mediated brain repair rescued behavioral impairment. Thus, we identify the CD95/CD95L system as an instructive signal for ongoing and injury-induced neurogenesis.


Assuntos
Células-Tronco Adultas/metabolismo , Isquemia Encefálica/metabolismo , Encéfalo/metabolismo , Proteína Ligante Fas/metabolismo , Neurogênese/fisiologia , Receptor fas/metabolismo , Células-Tronco Adultas/transplante , Animais , Isquemia Encefálica/terapia , Feminino , Expressão Gênica/fisiologia , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Proteínas Quinases/metabolismo , Transdução de Sinais/fisiologia , Transplante de Células-Tronco , Serina-Treonina Quinases TOR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA