Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Vis Exp ; (200)2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37929987

RESUMO

The blood-brain barrier (BBB) is a key physiological component of the central nervous system (CNS), maintaining nutrients, clearing waste, and protecting the brain from pathogens. The inherent barrier properties of the BBB pose a challenge for therapeutic drug delivery into the CNS to treat neurological diseases. Impaired BBB function has been related to neurological disease. Cerebral amyloid angiopathy (CAA), the deposition of amyloid in the cerebral vasculature leading to a compromised BBB, is a co-morbidity in most cases of Alzheimer's disease (AD), suggesting that BBB dysfunction or breakdown may be involved in neurodegeneration. Due to limited access to human BBB tissue, the mechanisms that contribute to proper BBB function and BBB degeneration remain unknown. To address these limitations, we have developed a human pluripotent stem cell-derived BBB (iBBB) by incorporating endothelial cells, pericytes, and astrocytes in a 3D matrix. The iBBB self-assembles to recapitulate the anatomy and cellular interactions present in the BBB. Seeding iBBBs with amyloid captures key aspects of CAA. Additionally, the iBBB offers a flexible platform to modulate genetic and environmental factors implicated in cerebrovascular disease and neurodegeneration, to investigate how genetics and lifestyle affect disease risk. Finally, the iBBB can be used for drug screening and medicinal chemistry studies to optimize therapeutic delivery to the CNS. In this protocol, we describe the differentiation of the three types of cells (endothelial cells, pericytes, and astrocytes) arising from human pluripotent stem cells, how to assemble the differentiated cells into the iBBB, and how to model CAA in vitro using exogenous amyloid. This model overcomes the challenge of studying live human brain tissue with a system that has both biological fidelity and experimental flexibility, and enables the interrogation of the human BBB and its role in neurodegeneration.


Assuntos
Doença de Alzheimer , Angiopatia Amiloide Cerebral , Humanos , Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Encéfalo/metabolismo , Sistema Nervoso Central/metabolismo , Doença de Alzheimer/metabolismo
2.
bioRxiv ; 2023 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-37645757

RESUMO

Patient-specific, human-based cellular models that integrate biomimetic BBB, immune, and myelinated neuron components are critically needed to enable translationally relevant and accelerated discovery of neurological disease mechanisms and interventions. By engineering a brain-mimicking 3D hydrogel and co-culturing all six major brain cell types derived from patient iPSCs, we have constructed, characterized, and utilized a multicellular integrated brain (miBrain) immuno-glial-neurovascular model with in vivo- like hallmarks. As proof of principle, here we utilized the miBrain to model Alzheimer's Disease pathologies associated with APOE4 genetic risk. APOE4 miBrains differentially exhibit amyloid aggregation, tau phosphorylation, and astrocytic GFAP. Unlike the co-emergent fate specification of glia and neurons in organoids, miBrains integrate independently differentiated cell types in a modular system with unique utility for elucidating cell-type specific contributions to pathogenesis. We here harness this feature to identify that risk factor APOE4 in astrocytes promotes tau pathogenesis and neuronal dysregulation through crosstalk with microglia. One-Sentence Summary: A novel patient-specific brain model with BBB, neuronal, immune, and glial components was developed, characterized, and harnessed to model Alzheimer's Disease-associated pathologies and APOE4 genetic risk.

3.
Methods Mol Biol ; 2683: 135-151, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37300772

RESUMO

The blood-brain barrier (BBB) is a key physiological component of the brain, protecting the brain from peripheral processes and pathogens. The BBB is a dynamic structure that is heavily involved in cerebral blood flow, angiogenesis, and other neural functions. However, the BBB also creates a challenging barrier for the entry of therapeutics into the brain, blocking more than 98% of drugs from contact with the brain. Neurovascular comorbidities are common in several neurological diseases including Alzheimer's and Parkinson's Disease, suggesting that BBB dysfunction or break down likely has a causal role in neurodegeneration. However, the mechanisms by which the human BBB is formed, maintained, and degenerated in diseases remain largely unknown due to limited access to human BBB tissue. To address these limitations, we have developed an in vitro induced human BBB (iBBB) derived from pluripotent stem cells. The iBBB model can be used for discovery of disease mechanisms, drug targets, drug screening, and medicinal chemistry studies to optimize brain penetration of central nervous system therapeutics. In this chapter, we will explain the steps to differentiate the three cellular components (endothelial cells, pericytes, and astrocytes) from induced pluripotent stem cells, and how to assemble them into the iBBB.


Assuntos
Barreira Hematoencefálica , Células-Tronco Pluripotentes Induzidas , Humanos , Células Endoteliais , Astrócitos , Encéfalo
5.
Nature ; 611(7937): 769-779, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36385529

RESUMO

APOE4 is the strongest genetic risk factor for Alzheimer's disease1-3. However, the effects of APOE4 on the human brain are not fully understood, limiting opportunities to develop targeted therapeutics for individuals carrying APOE4 and other risk factors for Alzheimer's disease4-8. Here, to gain more comprehensive insights into the impact of APOE4 on the human brain, we performed single-cell transcriptomics profiling of post-mortem human brains from APOE4 carriers compared with non-carriers. This revealed that APOE4 is associated with widespread gene expression changes across all cell types of the human brain. Consistent with the biological function of APOE2-6, APOE4 significantly altered signalling pathways associated with cholesterol homeostasis and transport. Confirming these findings with histological and lipidomic analysis of the post-mortem human brain, induced pluripotent stem-cell-derived cells and targeted-replacement mice, we show that cholesterol is aberrantly deposited in oligodendrocytes-myelinating cells that are responsible for insulating and promoting the electrical activity of neurons. We show that altered cholesterol localization in the APOE4 brain coincides with reduced myelination. Pharmacologically facilitating cholesterol transport increases axonal myelination and improves learning and memory in APOE4 mice. We provide a single-cell atlas describing the transcriptional effects of APOE4 on the aging human brain and establish a functional link between APOE4, cholesterol, myelination and memory, offering therapeutic opportunities for Alzheimer's disease.


Assuntos
Apolipoproteína E4 , Encéfalo , Colesterol , Fibras Nervosas Mielinizadas , Oligodendroglia , Animais , Humanos , Camundongos , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Apolipoproteína E4/genética , Apolipoproteína E4/metabolismo , Encéfalo/metabolismo , Encéfalo/patologia , Colesterol/metabolismo , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Fibras Nervosas Mielinizadas/metabolismo , Fibras Nervosas Mielinizadas/patologia , Autopsia , Células-Tronco Pluripotentes Induzidas , Neurônios/metabolismo , Neurônios/patologia , Heterozigoto , Transporte Biológico , Homeostase , Análise de Célula Única , Memória , Envelhecimento/genética , Perfilação da Expressão Gênica , Bainha de Mielina/metabolismo , Bainha de Mielina/patologia
6.
Nat Rev Neurol ; 18(1): 25-39, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34750588

RESUMO

Alzheimer disease (AD) is the most prevalent type of dementia. It is marked by severe memory loss and cognitive decline, and currently has limited effective treatment options. Although individuals with AD have common neuropathological hallmarks, emerging data suggest that the disease has a complex polygenic aetiology, and more than 25 genetic loci have been linked to an elevated risk of AD and dementia. Nevertheless, our ability to decipher the cellular and molecular mechanisms that underlie genetic susceptibility to AD, and its progression and severity, remains limited. Here, we discuss ongoing efforts to leverage genomic data from patients using cellular reprogramming technologies to recapitulate complex brain systems and build in vitro discovery platforms. Much attention has already been given to methodologies to derive major brain cell types from pluripotent stem cells. We therefore focus on technologies that combine multiple cell types to recreate anatomical and physiological properties of human brain tissue in vitro. We discuss the advances in the field for modelling four domains that have come into view as key contributors to the pathogenesis of AD: the blood-brain barrier, myelination, neuroinflammation and neuronal circuits. We also highlight opportunities for the field to further interrogate the complex genetic and environmental factors of AD using in vitro models.


Assuntos
Doença de Alzheimer/patologia , Encéfalo/patologia , Modelos Anatômicos , Modelos Neurológicos , Doença de Alzheimer/genética , Doença de Alzheimer/fisiopatologia , Animais , Encéfalo/fisiologia , Encéfalo/fisiopatologia , Predisposição Genética para Doença , Humanos
8.
Cell Stem Cell ; 27(4): 508-510, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-33007233

RESUMO

COVID-19 has unfortunately halted lab work, conferences, and in-person networking, which is especially detrimental to researchers just starting their labs. Through social media and our reviewer networks, we met some early-career stem cell investigators impacted by the closures. Here, they introduce themselves and their research to our readers.

9.
Nat Med ; 26(6): 952-963, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32514169

RESUMO

In Alzheimer's disease, amyloid deposits along the brain vasculature lead to a condition known as cerebral amyloid angiopathy (CAA), which impairs blood-brain barrier (BBB) function and accelerates cognitive degeneration. Apolipoprotein (APOE4) is the strongest risk factor for CAA, yet the mechanisms underlying this genetic susceptibility are unknown. Here we developed an induced pluripotent stem cell-based three-dimensional model that recapitulates anatomical and physiological properties of the human BBB in vitro. Similarly to CAA, our in vitro BBB displayed significantly more amyloid accumulation in APOE4 compared to APOE3. Combinatorial experiments revealed that dysregulation of calcineurin-nuclear factor of activated T cells (NFAT) signaling and APOE in pericyte-like mural cells induces APOE4-associated CAA pathology. In the human brain, APOE and NFAT are selectively dysregulated in pericytes of APOE4 carriers, and inhibition of calcineurin-NFAT signaling reduces APOE4-associated CAA pathology in vitro and in vivo. Our study reveals the role of pericytes in APOE4-mediated CAA and highlights calcineurin-NFAT signaling as a therapeutic target in CAA and Alzheimer's disease.


Assuntos
Apolipoproteína E4/genética , Barreira Hematoencefálica/metabolismo , Calcineurina/metabolismo , Angiopatia Amiloide Cerebral/genética , Fatores de Transcrição NFATC/genética , Pericitos/metabolismo , Peptídeos beta-Amiloides/metabolismo , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Apolipoproteína E4/metabolismo , Barreira Hematoencefálica/citologia , Humanos , Técnicas In Vitro , Células-Tronco Pluripotentes Induzidas , Fatores de Transcrição NFATC/metabolismo , Permeabilidade , RNA-Seq , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
10.
Cell Stem Cell ; 24(3): 347-349, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30849361

RESUMO

Conflicting clinical studies have reported that statins both reduce and accelerate cognitive impairments in Alzheimer's disease. In this issue, Van der Kant et al. (2019) use iPSC-derived neurons to thoroughly dissect the link between cholesterol synthesis, phospho-Tau, and amyloid-ß, revealing new therapeutic opportunities in Alzheimer's disease and related dementias.


Assuntos
Doença de Alzheimer , Inibidores de Hidroximetilglutaril-CoA Redutases , Peptídeos beta-Amiloides , Colesterol , Humanos , Neurônios , Proteínas tau
11.
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
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.
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
14.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...