Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(8)2021 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-33921461

RESUMEN

Astrocytes play central roles in normal brain function and are critical components of synaptic networks that oversee behavioral outputs. Despite their close affiliation with neurons, how neuronal-derived signals influence astrocyte function at the gene expression level remains poorly characterized, largely due to difficulties associated with dissecting neuron- versus astrocyte-specific effects. Here, we use an in vitro system of stem cell-derived astrocytes to identify gene expression profiles in astrocytes that are influenced by neurons and regulate astrocyte development. Furthermore, we show that neurotransmitters and neuromodulators induce distinct transcriptomic and chromatin accessibility changes in astrocytes that are unique to each of these neuroactive compounds. These findings are highlighted by the observation that noradrenaline has a more profound effect on transcriptional profiles of astrocytes compared to glutamate, gamma-aminobutyric acid (GABA), acetylcholine, and serotonin. This is demonstrated through enhanced noradrenaline-induced transcriptomic and chromatin accessibility changes in vitro and through enhanced calcium signaling in vivo. Taken together, our study reveals distinct transcriptomic and chromatin architecture signatures in astrocytes in response to neuronal-derived neuroactive compounds. Since astrocyte function is affected in all neurological disorders, this study provides a new entry point for exploring genetic mechanisms of astrocyte-neuron communication that may be dysregulated in disease.


Asunto(s)
Astrocitos/metabolismo , Encéfalo/metabolismo , Neuronas/metabolismo , Transcriptoma/genética , Acetilcolina/genética , Animales , Astrocitos/efectos de los fármacos , Encéfalo/efectos de los fármacos , Comunicación Celular/efectos de los fármacos , Ácido Glutámico/genética , Ratones , Células Madre Embrionarias de Ratones/efectos de los fármacos , Neuronas/efectos de los fármacos , Neurotransmisores/farmacología , Norepinefrina/genética , Serotonina/genética , Transducción de Señal/efectos de los fármacos , Ácido gamma-Aminobutírico/genética
2.
Genes Dev ; 26(9): 891-907, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22549954

RESUMEN

Astrocytes are no longer seen as a homogenous population of cells. In fact, recent studies indicate that astrocytes are morphologically and functionally diverse and play critical roles in neurodevelopmental diseases such as Rett syndrome and fragile X mental retardation. This review summarizes recent advances in astrocyte development, including the role of neural tube patterning in specification and developmental functions of astrocytes during synaptogenesis. We propose here that a precise understanding of astrocyte development is critical to defining heterogeneity and could lead advances in understanding and treating a variety of neuropsychiatric diseases.


Asunto(s)
Astrocitos/fisiología , Trastornos Heredodegenerativos del Sistema Nervioso/etiología , Trastornos Mentales/etiología , Neurogénesis , Astrocitos/patología , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Humanos , Trastornos Mentales/patología , Células-Madre Neurales/patología , Células-Madre Neurales/fisiología
3.
Dev Dyn ; 248(1): 21-33, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30016584

RESUMEN

Astrocytes exhibit dynamic and complex reactions to various insults. Recently, investigations into the transitions that occur during cellular specification, differentiation, maturation, and disease responses have provided insights into understanding the mechanisms that underlie these altered states of reactivity and function. Here we summarize current concepts in how astrocyte state transitions, termed astroplasticity, are regulated, as well as how this affects neural circuit function through extracellular signaling. We postulate that a promising future approach toward enhancing functional repair after injury and disease would be to steer astrocytes away from an inhibitory response and toward one that is beneficial to neuroplasticity and neuroregeneration. Toward this goal, we discuss emerging biotechnological advancements, with a focus on human pluripotent stem cell bioengineering, which has high potential for effective manipulation and control of astroplasticity. Highlights include innovations in cellular transdifferentiation techniques, nanomedicine, organoid and three-dimensional (3D) spheroid microcircuit development, and the use of biomaterials to influence the extracellular environment. Current barriers and future applications are also summarized in order to augment the design of future preclinical trials aimed toward astrocyte-targeted neuroregeneration with a concept termed astrocellular therapeutics. Developmental Dynamics 248:21-33, 2019. © 2018 Wiley Periodicals, Inc.


Asunto(s)
Astrocitos/citología , Bioingeniería/tendencias , Plasticidad de la Célula , Regeneración Nerviosa , Animales , Bioingeniería/métodos , Transdiferenciación Celular , Humanos , Células Madre Pluripotentes , Terapéutica/métodos , Terapéutica/tendencias
4.
Proc Natl Acad Sci U S A ; 113(50): 14408-14413, 2016 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-27911847

RESUMEN

The rapid spread of Zika virus (ZIKV) and its association with abnormal brain development constitute a global health emergency. Congenital ZIKV infection produces a range of mild to severe pathologies, including microcephaly. To understand the pathophysiology of ZIKV infection, we used models of the developing brain that faithfully recapitulate the tissue architecture in early to midgestation. We identify the brain cell populations that are most susceptible to ZIKV infection in primary human tissue, provide evidence for a mechanism of viral entry, and show that a commonly used antibiotic protects cultured brain cells by reducing viral proliferation. In the brain, ZIKV preferentially infected neural stem cells, astrocytes, oligodendrocyte precursor cells, and microglia, whereas neurons were less susceptible to infection. These findings suggest mechanisms for microcephaly and other pathologic features of infants with congenital ZIKV infection that are not explained by neural stem cell infection alone, such as calcifications in the cortical plate. Furthermore, we find that blocking the glia-enriched putative viral entry receptor AXL reduced ZIKV infection of astrocytes in vitro, and genetic knockdown of AXL in a glial cell line nearly abolished infection. Finally, we evaluate 2,177 compounds, focusing on drugs safe in pregnancy. We show that the macrolide antibiotic azithromycin reduced viral proliferation and virus-induced cytopathic effects in glial cell lines and human astrocytes. Our characterization of infection in the developing human brain clarifies the pathogenesis of congenital ZIKV infection and provides the basis for investigating possible therapeutic strategies to safely alleviate or prevent the most severe consequences of the epidemic.


Asunto(s)
Azitromicina/farmacología , Encéfalo/embriología , Encéfalo/virología , Tropismo Viral/efectos de los fármacos , Infección por el Virus Zika/tratamiento farmacológico , Virus Zika/efectos de los fármacos , Virus Zika/fisiología , Encéfalo/patología , Línea Celular , Efecto Citopatogénico Viral/efectos de los fármacos , Femenino , Humanos , Recién Nacido , Pruebas de Sensibilidad Microbiana , Microcefalia/tratamiento farmacológico , Microcefalia/embriología , Microcefalia/patología , Neuroglía/efectos de los fármacos , Neuroglía/patología , Neuroglía/virología , Embarazo , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/fisiología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/fisiología , Tropismo Viral/fisiología , Internalización del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Virus Zika/patogenicidad , Infección por el Virus Zika/embriología , Infección por el Virus Zika/patología , Tirosina Quinasa del Receptor Axl
5.
Neuron ; 111(6): 857-873.e8, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36640767

RESUMEN

Using machine learning (ML), we interrogated the function of all human-chimpanzee variants in 2,645 human accelerated regions (HARs), finding 43% of HARs have variants with large opposing effects on chromatin state and 14% on neurodevelopmental enhancer activity. This pattern, consistent with compensatory evolution, was confirmed using massively parallel reporter assays in chimpanzee and human neural progenitor cells. The species-specific enhancer activity of HARs was accurately predicted from the presence and absence of transcription factor footprints in each species. Despite these striking cis effects, activity of a given HAR sequence was nearly identical in human and chimpanzee cells. This suggests that HARs did not evolve to compensate for changes in the trans environment but instead altered their ability to bind factors present in both species. Thus, ML prioritized variants with functional effects on human neurodevelopment and revealed an unexpected reason why HARs may have evolved so rapidly.


Asunto(s)
Encéfalo , Elementos de Facilitación Genéticos , Pan troglodytes , Animales , Humanos , Cromatina , Aprendizaje Automático , Pan troglodytes/metabolismo , Factores de Transcripción/genética , Encéfalo/crecimiento & desarrollo
6.
J Cell Biol ; 221(4)2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35139144

RESUMEN

Astrocyte reactivity can directly modulate nervous system function and immune responses during disease and injury. However, the consequence of human astrocyte reactivity in response to specific contexts and within neural networks is obscure. Here, we devised a straightforward bioengineered neural organoid culture approach entailing transcription factor-driven direct differentiation of neurons and astrocytes from human pluripotent stem cells combined with genetically encoded tools for dual cell-selective activation. This strategy revealed that Gq-GPCR activation via chemogenetics in astrocytes promotes a rise in intracellular calcium followed by induction of immediate early genes and thrombospondin 1. However, astrocytes also undergo NF-κB nuclear translocation and secretion of inflammatory proteins, correlating with a decreased evoked firing rate of cocultured optogenetic neurons in suboptimal conditions, without overt neurotoxicity. Altogether, this study clarifies the intrinsic reactivity of human astrocytes in response to targeting GPCRs and delivers a bioengineered approach for organoid-based disease modeling and preclinical drug testing.


Asunto(s)
Astrocitos/metabolismo , Bioingeniería , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Neuronas/metabolismo , Organoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adenosina Trifosfato/farmacología , Astrocitos/patología , Calcio/metabolismo , Línea Celular , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Inflamación/patología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Células Madre Pluripotentes/metabolismo , Reproducibilidad de los Resultados , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Sinaptofisina/metabolismo
7.
Drug Deliv Transl Res ; 11(6): 2394-2413, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34176099

RESUMEN

Host immune cells interact bi-directionally with their extracellular matrix (ECM) to receive and deposit molecular signals, which orchestrate cellular activation, proliferation, differentiation, and function to maintain healthy tissue homeostasis. In response to pathogens or damage, immune cells infiltrate diseased sites and synthesize critical ECM molecules such as glycoproteins, proteoglycans, and glycosaminoglycans to promote healing. When the immune system misidentifies pathogens or fails to survey damaged cells effectively, maladies such as chronic inflammation, autoimmune diseases, and cancer can develop. In these conditions, it is essential to restore balance to the body through modulation of the immune system and the ECM. This review details the components of dysregulated ECM implicated in pathogenic environments and therapeutic approaches to restore tissue homeostasis. We evaluate emerging strategies to overcome inflamed, immune inhibitory, and otherwise diseased microenvironments, including mechanical stimulation, targeted proteases, adoptive cell therapy, mechanomedicine, and biomaterial-based cell therapeutics. We highlight various strategies that have produced efficacious responses in both pre-clinical and human trials and identify additional opportunities to develop next-generation interventions. Significantly, we identify a need for therapies to address dense or fibrotic tissue for the treatment of organ tissue damage and various cancer subtypes. Finally, we conclude that therapeutic techniques that disrupt, evade, or specifically target the pathogenic microenvironment have a high potential for improving therapeutic outcomes and should be considered a priority for immediate exploration. A schematic showing the various methods of extracellular matrix disruption/targeting in both fibrotic and cancerous environments. a Biomaterial-based cell therapy can be used to deliver anti-inflammatory cytokines, chemotherapeutics, or other factors for localized, slow release of therapeutics. b Mechanotherapeutics can be used to inhibit the deposition of molecules such as collagen that affect stiffness. c Ablation of the ECM and target tissue can be accomplished via mechanical degradation such as focused ultrasound. d Proteases can be used to improve the distribution of therapies such as oncolytic virus. e Localization of therapeutics such as checkpoint inhibitors can be improved with the targeting of specific ECM components, reducing off-target effects and toxicity.


Asunto(s)
Matriz Extracelular , Inmunomodulación , Tratamiento Basado en Trasplante de Células y Tejidos , Colágeno/metabolismo , Humanos , Inflamación
8.
Neurotherapeutics ; 18(3): 1710-1728, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34160773

RESUMEN

CAG repeat expansion is the genetic cause of nine incurable polyglutamine (polyQ) diseases with neurodegenerative features. Silencing repeat RNA holds great therapeutic value. Here, we developed a repeat-based RNA-cleaving DNAzyme that catalyzes the destruction of expanded CAG repeat RNA of six polyQ diseases with high potency. DNAzyme preferentially cleaved the expanded allele in spinocerebellar ataxia type 1 (SCA1) cells. While cleavage was non-allele-specific for spinocerebellar ataxia type 3 (SCA3) cells, treatment of DNAzyme leads to improved cell viability without affecting mitochondrial metabolism or p62-dependent aggresome formation. DNAzyme appears to be stable in mouse brain for at least 1 month, and an intermediate dosage of DNAzyme in a SCA3 mouse model leads to a significant reduction of high molecular weight ATXN3 proteins. Our data suggest that DNAzyme is an effective RNA silencing molecule for potential treatment of multiple polyQ diseases.


Asunto(s)
ADN Catalítico/administración & dosificación , ADN Catalítico/genética , Enfermedad de Machado-Joseph/genética , Péptidos/genética , ARN/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Ataxina-3/genética , Línea Celular Tumoral , Silenciador del Gen/fisiología , Células HEK293 , Humanos , Enfermedad de Machado-Joseph/terapia , Ratones , Péptidos/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/terapia , Técnicas Estereotáxicas
9.
Adv Sci (Weinh) ; 8(19): e2101437, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34382379

RESUMEN

Nanovesicles (NVs) are emerging as innovative, theranostic tools for cargo delivery. Recently, surface engineering of NVs with membrane proteins from specific cell types has been shown to improve the biocompatibility of NVs and enable the integration of functional attributes. However, this type of biomimetic approach has not yet been explored using human neural cells for applications within the nervous system. Here, this paper optimizes and validates the scalable and reproducible production of two types of neuron-targeting NVs, each with a distinct lipid formulation backbone suited to potential therapeutic cargo, by integrating membrane proteins that are unbiasedly sourced from human pluripotent stem-cell-derived neurons. The results establish that both endogenous and genetically engineered cell-derived proteins effectively transfer to NVs without disruption of their physicochemical properties. NVs with neuron-derived membrane proteins exhibit enhanced neuronal association and uptake compared to bare NVs. Viability of 3D neural sphere cultures is not disrupted by treatment, which verifies the utility of organoid-based approaches as NV testing platforms. Finally, these results confirm cellular association and uptake of the biomimetic humanized NVs to neurons within rodent cranial nerves. In summary, the customizable NVs reported here enable next-generation functionalized theranostics aimed to promote neuroregeneration.


Asunto(s)
Materiales Biomiméticos/metabolismo , Biomimética/métodos , Vesículas Extracelulares/metabolismo , Nanoestructuras/química , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Animales , Comunicación Celular , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL
10.
Ann Clin Transl Neurol ; 8(11): 2211-2221, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34647437

RESUMEN

Intracerebral hemorrhage (ICH) remains a common and debilitating form of stroke. This neurological emergency must be diagnosed and treated rapidly yet effectively. In this article, we review the medical, surgical, repair, and regenerative treatment options for managing ICH. Topics of focus include the management of blood pressure, intracranial pressure, coagulopathy, and intraventricular hemorrhage, as well as the role of surgery, regeneration, rehabilitation, and secondary prevention. Results of various phase II and III trials are incorporated. In summary, ICH patients should undergo rapid evaluation with neuroimaging, and early interventions should include systolic blood pressure control in the range of 140 mmHg, correction of coagulopathy if indicated, and assessment for surgical intervention. ICH patients should be managed in dedicated neurosurgical intensive care or stroke units where continuous monitoring of neurological status and evaluation for neurological deterioration is rapidly possible. Extravasation of hematoma may be helpful in patients with intraventricular extension of ICH. The goal of care is to reduce mortality and enable multimodal rehabilitative therapy.


Asunto(s)
Hemorragia Cerebral/terapia , Fármacos Hematológicos , Rehabilitación Neurológica , Procedimientos Neuroquirúrgicos , Prevención Secundaria , Trasplante de Células Madre , Hemorragia Cerebral/tratamiento farmacológico , Hemorragia Cerebral/cirugía , Humanos
11.
J Vis Exp ; (138)2018 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-30176009

RESUMEN

A barrier to our understanding of how various cell types and signals contribute to synaptic circuit function is the lack of relevant models for studying the human brain. One emerging technology to address this issue is the use of three dimensional (3D) neural cell cultures, termed 'organoids' or 'spheroids', for long term preservation of intercellular interactions including extracellular adhesion molecules. However, these culture systems are time consuming and not systematically generated. Here, we detail a method to rapidly and consistently produce 3D cocultures of neurons and astrocytes from human pluripotent stem cells. First, pre-differentiated astrocytes and neuronal progenitors are dissociated and counted. Next, cells are combined in sphere-forming dishes with a Rho-Kinase inhibitor and at specific ratios to produce spheres of reproducible size. After several weeks of culture as floating spheres, cocultures ('asteroids') are finally sectioned for immunostaining or plated upon multielectrode arrays to measure synaptic density and strength. In general, it is expected that this protocol will yield 3D neural spheres that display mature cell-type restricted markers, form functional synapses, and exhibit spontaneous synaptic network burst activity. Together, this system permits drug screening and investigations into mechanisms of disease in a more suitable model compared to monolayer cultures.


Asunto(s)
Astrocitos/citología , Emparejamiento Cromosómico/fisiología , Técnicas de Cocultivo/métodos , Neuronas/citología , Células Madre Pluripotentes/citología , Astrocitos/metabolismo , Diferenciación Celular/fisiología , Humanos , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo
12.
Cell Rep ; 25(4): 947-958.e4, 2018 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-30355500

RESUMEN

How mutations in glial fibrillary acidic protein (GFAP) cause Alexander disease (AxD) remains elusive. We generated iPSCs from two AxD patients and corrected the GFAP mutations to examine the effects of mutant GFAP on human astrocytes. AxD astrocytes displayed GFAP aggregates, recapitulating the pathological hallmark of AxD. RNA sequencing implicated the endoplasmic reticulum, vesicle regulation, and cellular metabolism. Corroborating this analysis, we observed enlarged and heterogeneous morphology coupled with perinuclear localization of endoplasmic reticulum and lysosomes in AxD astrocytes. Functionally, AxD astrocytes showed impaired extracellular ATP release, which is responsible for attenuated calcium wave propagation. These results reveal that AxD-causing mutations in GFAP disrupt intracellular vesicle regulation and impair astrocyte secretion, resulting in astrocyte dysfunction and AxD pathogenesis.


Asunto(s)
Astrocitos/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Mutación/genética , Orgánulos/metabolismo , Adenosina Trifosfato/metabolismo , Enfermedad de Alexander/metabolismo , Enfermedad de Alexander/patología , Animales , Astrocitos/citología , Señalización del Calcio , Diferenciación Celular , Retículo Endoplásmico/metabolismo , Humanos , Lisosomas/metabolismo , Ratones , Agregado de Proteínas , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
Curr Opin Chem Biol ; 10(6): 592-7, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17046316

RESUMEN

Stem cells can produce progenies that constitute an organism or a tissue while replenishing (renewing) themselves. The ability to produce large quantities of stage-specific cells from self-renewing stem cells in a precisely controlled manner makes it possible to dissect out complex interactions among macromolecules along development, such as early brain development at the global level. These cellular differentiation pathways also serve as templates for identifying biological effects of novel or existing chemical compounds. Thus, stem cells find their most powerful use in chemical biology, which may ultimately lead to applications in regenerative medicine.


Asunto(s)
Diferenciación Celular , Neuronas/citología , Células Madre/citología
14.
Brain Res Bull ; 129: 66-73, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27570101

RESUMEN

Cellular components of synaptic circuits have been adjusted for increased human brain size, neural cell density, energy consumption and developmental duration. How does the human brain make these accommodations? There is evidence that astrocytes are one of the most divergent neural cell types in primate brain evolution and it is now becoming clear that they have critical roles in controlling synaptic development, function and plasticity. Yet, we still do not know how the precise developmental appearance of these cells and subsequent astrocyte-derived signals modulate diverse neuronal circuit subtypes. Here, we discuss what is currently known about the influence of glial factors on synaptic maturation and focus on unique features of human astrocytes including their potential roles in regenerative and translational medicine. Human astrocyte distinctiveness may be a major contributor to high level neuronal processing of the human brain and act in novel ways during various neuropathies ranging from autism spectrum disorders, viral infection, injury and neurodegenerative conditions.


Asunto(s)
Astrocitos/fisiología , Sinapsis/fisiología , Animales , Encéfalo/fisiología , Humanos , Neurogénesis/fisiología
15.
J Exp Med ; 214(12): 3481-3495, 2017 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-29141869

RESUMEN

Prions are infectious agents that cause neurodegenerative diseases such as Creutzfeldt-Jakob disease (CJD). The absence of a human cell culture model that replicates human prions has hampered prion disease research for decades. In this paper, we show that astrocytes derived from human induced pluripotent stem cells (iPSCs) support the replication of prions from brain samples of CJD patients. For experimental exposure of astrocytes to variant CJD (vCJD), the kinetics of prion replication occur in a prion protein codon 129 genotype-dependent manner, reflecting the genotype-dependent susceptibility to clinical vCJD found in patients. Furthermore, iPSC-derived astrocytes can replicate prions associated with the major sporadic CJD strains found in human patients. Lastly, we demonstrate the subpassage of prions from infected to naive astrocyte cultures, indicating the generation of prion infectivity in vitro. Our study addresses a long-standing gap in the repertoire of human prion disease research, providing a new in vitro system for accelerated mechanistic studies and drug discovery.


Asunto(s)
Astrocitos/metabolismo , Células Madre Pluripotentes Inducidas/citología , Proteínas Priónicas/genética , Priones/metabolismo , Adulto , Células Cultivadas , Codón/genética , Síndrome de Creutzfeldt-Jakob/patología , Femenino , Genotipo , Humanos , Cinética , Masculino , Persona de Mediana Edad , Adulto Joven
16.
Stem Cell Reports ; 9(6): 1745-1753, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29198827

RESUMEN

Human astrocytes network with neurons in dynamic ways that are still poorly defined. Our ability to model this relationship is hampered by the lack of relevant and convenient tools to recapitulate this complex interaction. To address this barrier, we have devised efficient coculture systems utilizing 3D organoid-like spheres, termed asteroids, containing pre-differentiated human pluripotent stem cell (hPSC)-derived astrocytes (hAstros) combined with neurons generated from hPSC-derived neural stem cells (hNeurons) or directly induced via Neurogenin 2 overexpression (iNeurons). Our systematic methods rapidly produce structurally complex hAstros and synapses in high-density coculture with iNeurons in precise numbers, allowing for improved studies of neural circuit function, disease modeling, and drug screening. We conclude that these bioengineered neural circuit model systems are reliable and scalable tools to accurately study aspects of human astrocyte-neuron functional properties while being easily accessible for cell-type-specific manipulations and observations.


Asunto(s)
Astrocitos/citología , Diferenciación Celular/genética , Técnicas de Cocultivo , Neuronas/citología , Astrocitos/metabolismo , Linaje de la Célula/genética , Linaje de la Célula/fisiología , Células Cultivadas , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Sinapsis/metabolismo , Sinapsis/fisiología
17.
J Neurosci Methods ; 266: 42-9, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27036311

RESUMEN

BACKGROUND: Basal forebrain cholinergic neurons (BFCNs) play critical roles in learning, memory and cognition. Dysfunction or degeneration of BFCNs may connect to neuropathology, such as Alzheimer's disease, Down's syndrome and dementia. Generation of functional BFCNs may contribute to the studies of cell-based therapy and pathogenesis that is related to learning and memory deficits. NEW METHOD: Here we describe a detail method for robust generation of BFCNs from human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). In this method, BFCN progenitors are patterned from hESC or hiPSC-derived primitive neuroepithelial cells, with the treatment of sonic hedgehog (SHH) or combination with its agonist Purmorphamine, and by co-culturing with human astrocytes. RESULTS: At day 20, ∼90% hPSC-derived progenitors expressed NKX2.1, which is a transcriptional marker for MGE. Moreover, around 40% of NKX2.1+ cells co-expressed OLIG2 and ∼15% of NKX2.1+ cells co-expressed ISLET1, which are ventral markers. At day 35, ∼40% neurons robustly express ChAT, most of which are co-labeled with NKX2.1, ISLET1 and FOXG1, indicating the basal forebrain-like identity. At day 45, these neurons express mature neuronal markers MAP2, Synapsin, and VAChT. COMPARISON WITH EXISTING METHOD(S): In this method, undefined conditions including genetic modification or cell-sorting are avoided. As a choice, feeder free conditions are used to avoid ingredients of animal origin. Moreover, Purmorphamine can be substituted for SHH to induce ventral progenitors effectively and economically. CONCLUSION: We provide an efficient method to generate BFCNs from multiple hPSC lines, which offers the potential application for disease modeling and pharmacological studies.


Asunto(s)
Prosencéfalo Basal/fisiología , Técnicas de Cultivo de Célula/métodos , Células Madre Embrionarias/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Astrocitos/citología , Astrocitos/fisiología , Prosencéfalo Basal/citología , Técnicas de Cultivo de Célula/instrumentación , Línea Celular , Colina O-Acetiltransferasa/metabolismo , Técnicas de Cocultivo/instrumentación , Técnicas de Cocultivo/métodos , Células Madre Embrionarias/citología , Factores de Transcripción Forkhead/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Proteínas del Tejido Nervioso/metabolismo , Nestina/metabolismo , Neuronas/citología , Factor de Transcripción PAX6/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factor Nuclear Tiroideo 1/metabolismo
18.
Sci Transl Med ; 7(286): 286ra66, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25947161

RESUMEN

Astrocytes produce an assortment of signals that promote neuronal maturation according to a precise developmental timeline. Is this orchestrated timing and signaling altered in human neurodevelopmental disorders? To address this question, the astroglial lineage was investigated in two model systems of a developmental disorder with intellectual disability caused by mutant Harvey rat sarcoma viral oncogene homolog (HRAS) termed Costello syndrome: mutant HRAS human induced pluripotent stem cells (iPSCs) and transgenic mice. Human iPSCs derived from patients with Costello syndrome differentiated to astroglia more rapidly in vitro than those derived from wild-type cell lines with normal HRAS, exhibited hyperplasia, and also generated an abundance of extracellular matrix remodeling factors and proteoglycans. Acute treatment with a farnesyl transferase inhibitor and knockdown of the transcription factor SNAI2 reduced expression of several proteoglycans in Costello syndrome iPSC-derived astrocytes. Similarly, mice in which mutant HRAS was expressed selectively in astrocytes exhibited experience-independent increased accumulation of perineuronal net proteoglycans in cortex, as well as increased parvalbumin expression in interneurons, when compared to wild-type mice. Our data indicate that astrocytes expressing mutant HRAS dysregulate cortical maturation during development as shown by abnormal extracellular matrix remodeling and implicate excessive astrocyte-to-neuron signaling as a possible drug target for treating mental impairment and enhancing neuroplasticity.


Asunto(s)
Astrocitos/citología , Síndrome de Costello/metabolismo , Matriz Extracelular/metabolismo , Células Madre Pluripotentes Inducidas/citología , Transducción de Señal , Animales , Astrocitos/metabolismo , Diferenciación Celular , Línea Celular , Regulación de la Expresión Génica , Genes ras , Genotipo , Hipocampo/metabolismo , Humanos , Espectrometría de Masas , Ratones , Ratones Transgénicos , Mutación , Plasticidad Neuronal , Neuronas/citología , Neuronas/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Proteoglicanos/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas ras/metabolismo
19.
Sci Rep ; 5: 18550, 2015 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-26670131

RESUMEN

Human pluripotent stem cells (hPSCs) have potential to differentiate to unlimited number of neural cells, which provide powerful tools for neural regeneration. To date, most reported protocols were established with an animal feeder system. However, cells derived on this system are inappropriate for the translation to clinical applications because of the introduction of xenogenetic factors. In this study, we provided an optimized paradigm to generate region-specific forebrain neurons from hPSCs under a defined system. We assessed five conditions and found that a vitronectin-coated substrate was the most efficient method to differentiate hPSCs to neurons and astrocytes. More importantly, by applying different doses of purmorphamine, a small-molecule agonist of sonic hedgehog signaling, hPSCs were differentiated to different region-specific forebrain neuron subtypes, including glutamatergic neurons, striatal medium spiny neurons, and GABA interneurons. Our study offers a highly defined system without exogenetic factors to produce human neurons and astrocytes for translational medical studies, including cell therapy and stem cell-based drug discovery.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Neuronas/citología , Células Madre Pluripotentes/citología , Prosencéfalo/citología , Línea Celular , Humanos , Neuronas/metabolismo , Células Madre Pluripotentes/metabolismo , Prosencéfalo/metabolismo
20.
Front Cell Neurosci ; 7: 25, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23503583

RESUMEN

What roles do astrocytes play in human disease?This question remains unanswered for nearly every human neurological disorder. Yet, because of their abundance and complexity astrocytes can impact neurological function in many ways. The differentiation of human pluripotent stem cells (hPSCs) into neuronal and glial subtypes, including astrocytes, is becoming routine, thus their use as tools for modeling neurodevelopment and disease will provide one important approach to answer this question. When designing experiments, careful consideration must be given to choosing paradigms for differentiation, maturation, and functional analysis of these temporally asynchronous cellular populations in culture. In the case of astrocytes, they display heterogeneous characteristics depending upon species of origin, brain region, developmental stage, environmental factors, and disease states, all of which may render experimental results highly variable. In this review, challenges and future directions are discussed for using hPSC-derived astroglial progenitors and mature astrocytes for neurodevelopmental studies with a focus on exploring human astrocyte effects upon neuronal function. As new technologies emerge to measure the functions of astrocytes in vitro and in vivo, there is also a need for a standardized source of human astrocytes that are most relevant to the diseases of interest.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA