Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Stem Cell Res Ther ; 15(1): 356, 2024 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-39385216

RESUMO

BACKGROUND: Parkinson´s disease (PD), the second most common neurodegenerative disease in the world, is characterized by the death or impairment of dopaminergic neurons (DAn) in the substantia nigra pars compacta and dopamine depletion in the striatum. Currently, there is no cure for PD, and treatments only help to reduce the symptoms of the disease, and do not repair or replace the DAn damaged or lost in PD. Cell replacement therapy (CRT) seeks to relieve both pathological and symptomatic PD manifestations and has been shown to have beneficial effects in experimental PD models as well as in PD patients, but an apt cell line to be used in the treatment of PD has yet to be established. The purpose of this study was to examine the effects of the transplantation of hVM1 clone 32 cells, a bankable line of human neural stem cells (hNSCs), in a PD mouse model at four months post-transplant. METHODS: Adult (five month-old) C57BL/6JRccHsd male mice were injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and subsequently transplanted with hVM1 clone 32 cells, or buffer, in the left striatum. Four months post-transplant, behavioral effects were explored using the open field and paw print tests, and histological analyses were performed. RESULTS: Transplantation of hVM1 clone 32 cells rescued dopaminergic nigrostriatal populations in adult Parkinsonian mice. Motor and neurological deterioration were observed in buffer-treated mice, the latter of which had a tendency to improve in hNSC-transplanted mice. Detection of mast cell migration to the superficial cervical lymph nodes in cell-transplanted mice denoted a peripheral effect. Transplantation of hNSCs also rescued neuroblast neurogenesis in the subgranular zone, which was correlated with dopaminergic recovery and is indicative of local recovery mechanisms. CONCLUSIONS: In this proof-of-concept study, the transplantation of hVM1 clone 32 cells provided neuroprotection in adult Parkinsonian mice by restoring the dopaminergic nigrostriatal pathway and hippocampal neurogenesis, demonstrating the efficacy of cell replacement therapy as a treatment for PD.


Assuntos
Modelos Animais de Doenças , Neurônios Dopaminérgicos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais , Doença de Parkinson , Animais , Células-Tronco Neurais/transplante , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Camundongos , Masculino , Doença de Parkinson/terapia , Neurônios Dopaminérgicos/metabolismo , Humanos , Substância Negra , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina
2.
Int J Mol Med ; 53(1)2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38038161

RESUMO

Schizophrenia (SZ) is a multifactorial disorder characterized by volume reduction in gray and white matter, oxidative stress, neuroinflammation, altered neurotransmission, as well as molecular deficiencies such as punctual mutation in Disrupted­in­Schizophrenia 1 protein. In this regard, it is essential to understand the underlying molecular disturbances to determine the pathophysiological mechanisms of the disease. The signaling pathways activated by G protein­coupled receptors (GPCRs) are key molecular signaling pathways altered in SZ. Convenient models need to be designed and validated to study these processes and mechanisms at the cellular level. Cultured olfactory stem cells are used to investigate neural molecular and cellular alterations related to the pathophysiology of SZ. Multipotent human olfactory stem cells are undifferentiated and express GPCRs involved in numerous physiological functions such as proliferation, differentiation and bioenergetics. The use of olfactory stem cells obtained from patients with SZ may identify alterations in GPCR signaling that underlie dysfunctional processes in both undifferentiated and specialized neurons or derived neuroglia. The present review aimed to analyze the role of GPCRs and their signaling in the pathophysiology of SZ. Culture of olfactory epithelial cells constitutes a suitable model to study SZ and other psychiatric disorders at the cellular level.


Assuntos
Esquizofrenia , Humanos , Esquizofrenia/genética , Esquizofrenia/metabolismo , Células Neuroepiteliais/metabolismo , Neurônios/metabolismo , Receptores Acoplados a Proteínas G , Células-Tronco/metabolismo
3.
Cells ; 12(23)2023 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-38067139

RESUMO

Major features of aging might be progressive decreases in cognitive function and physical activity, in addition to withered appearance. Previously, we reported that the intracerebroventricular injection of human neural stem cells (NSCs named F3) encoded the choline acetyltransferase gene (F3.ChAT). The cells secreted acetylcholine and growth factors (GFs) and neurotrophic factors (NFs), thereby improving learning and memory function as well as the physical activity of aged animals. In this study, F344 rats (10 months old) were intravenously transplanted with F3 or F3.ChAT NSCs (1 × 106 cells) once a month to the 21st month of age. Their physical activity and cognitive function were investigated, and brain acetylcholine (ACh) and cholinergic and dopaminergic system markers were analyzed. Neuroprotective and neuroregenerative activities of stem cells were also confirmed by analyzing oxidative damages, neuronal skeletal protein, angiogenesis, brain and muscle weights, and proliferating host stem cells. Stem cells markedly improved both cognitive and physical functions, in parallel with the elevation in ACh levels in cerebrospinal fluid and muscles, in which F3.ChAT cells were more effective than F3 parental cells. Stem cell transplantation downregulated CCL11 and recovered GFs and NFs in the brain, leading to restoration of microtubule-associated protein 2 as well as functional markers of cholinergic and dopaminergic systems, along with neovascularization. Stem cells also restored muscular GFs and NFs, resulting in increased angiogenesis and muscle mass. In addition, stem cells enhanced antioxidative capacity, attenuating oxidative damage to the brain and muscles. The results indicate that NSCs encoding ChAT improve cognitive function and physical activity of aging animals by protecting and recovering functions of multiple organs, including cholinergic and dopaminergic systems, as well as muscles from oxidative injuries through secretion of ACh and GFs/NFs, increased antioxidant elements, and enhanced blood flow.


Assuntos
Acetilcolina , Células-Tronco Neurais , Ratos , Animais , Humanos , Masculino , Idoso , Lactente , Ratos Endogâmicos F344 , Acetilcolina/metabolismo , Colina O-Acetiltransferase/genética , Colina O-Acetiltransferase/metabolismo , Colina O-Acetiltransferase/farmacologia , Aprendizagem em Labirinto/fisiologia , Envelhecimento/fisiologia , Células-Tronco Neurais/metabolismo , Administração Intravenosa , Colinérgicos
4.
Stem Cell Rev Rep ; 19(4): 1001-1018, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36652144

RESUMO

Traumatic brain injury (TBI) causes a variety of complex pathological changes in brain parenchymal tissue by increasing neuroinflammatory and apoptosis responses. Currently, there is no treatment to resolve the consequences related to TBI. Recently, an extensive literature has grown up around the theme of bystander effects of stem cells, a mechanism of stem cells without the need for cell transplantation, which is called cell-free therapy. The purpose of this investigation was to determine the efficacy of a cell-free-based therapy strategy using exosomes derived from human neural stem cells (hNSCs) and a novel nano-scaffold in rats subjected to TBI. In this study, a series of in vitro and in vivo experiments from behavior tests to gene expression was performed to define the effect of exosomes in combination with a three-dimensional (3D) nano-scaffold containing a bio-motif of SDF1α (Nano-SDF). Application of exosomes with Nano-SDF significantly decreased oxidative stress in serum and brain samples. Moreover, treatment with exosomes and Nano-SDF significantly reduced the expression of Toll-like receptor 4 and its downstream signaling pathway, including NF-kß and interleukin-1ß. We also found that the cell-free-based therapy strategy could decrease reactive gliosis at the injury site. Interestingly, we showed that exosomes with Nano-SDF increased neurogenesis in the sub-ventricular zone of the lateral ventricle, indicating a bio-bridge mechanism. To sum up, the most obvious finding to emerge from this study is that a cell-free-based therapy strategy can be an effective option for future practice in the course of TBI.


Assuntos
Lesões Encefálicas Traumáticas , Exossomos , Células-Tronco Neurais , Ratos , Humanos , Animais , Exossomos/metabolismo , Doenças Neuroinflamatórias , Lesões Encefálicas Traumáticas/terapia , Lesões Encefálicas Traumáticas/patologia , Neurogênese/fisiologia , Células-Tronco Neurais/metabolismo
5.
Bioelectrochemistry ; 149: 108306, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36345111

RESUMO

The pathophysiological progress of Parkinson's disease leads through degeneration of dopaminergic neurons in the substantia nigra to complete cell death and lack of dopamine in the striatum where it modulates motor functions. Transplantation of dopaminergic stem cell-derived neurons is a possible therapy to restore dopamine levels. We have previously presented multifunctional pyrolytic carbon coated leaky optoelectrical fibers (LOEFs) with laser ablated micro-optical windows (µOWs) as carriers for channelrhodopsin-2 modified optogenetically active neurons for light-induced on-demand dopamine release and amperometric real-time detection. To increase the dopamine release by stimulating a larger neuronal population with light, we present here a novel approach to generate µOWs through laser ablation around the entire circumference of optical fibers to obtain Omni-LOEFs. Cyclic voltammetric characterization of the pyrolytic carbon showed that despite the increased number of µOWs, the electrochemical properties were not deteriorated. Finally, we demonstrate that the current recorded during real-time detection of dopamine upon light-induced stimulation of neurons differentiated on Omni-LOEFs is significantly higher compared to recordings from the same number of cells seeded on LOEFs with µOWs only on one side. Moreover, by varying the cell seeding density, we show that the recorded current is proportional to the dimension of the cell population.


Assuntos
Dopamina , Optogenética , Neurônios/fisiologia , Carbono/metabolismo
6.
Pathogens ; 11(10)2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36297275

RESUMO

Powassan virus (POWV) is a tick-borne flavivirus (TBFV) that can cause severe encephalitis in humans with a case-fatality rate as high as 11%. Patients who survive severe encephalitic disease can develop long-term neurological sequelae that can be debilitating and life-long. In this study, we have sought to characterize a primary human fetal brain neural stem cell system (hNSC), which can be differentiated into neuron and astrocyte co-cultures, to serve as a translational in vitro system for infection with POWV and a comparative mosquito-borne flavivirus (MBFV), West Nile virus (WNV). We found that both viruses are able to infect both cell types in the co-culture and that WNV elicits a strong inflammatory response characterized by increased cytokines IL-4, IL-6, IL-8, TNF-α and IL-1ß and activation of apoptosis pathways. POWV infection resulted in fewer cytokine responses, as well as less detectable apoptosis, while neurons infected with POWV exhibited structural aberrations forming in the dendrites. These anomalies are consistent with previous findings in which tick-borne encephalitis virus (TBEV) infected murine primary neurons formed laminal membrane structures (LMS). Furthermore, these structural aberrations are also recapitulated in brain tissue from infected mice. Our findings indicate that POWV is capable of infecting human primary neurons and astrocytes without causing apparent widespread apoptosis, while forming punctate structures reminiscent with LMS in primary human neurons and in vivo.

7.
Exp Ther Med ; 23(2): 164, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35069845

RESUMO

Bisphenol A (BPA) is a common industrial chemical widely used to produce various plastics and is known to impair neural stem cells (NSCs). However, the effects of low-dose BPA exposure on the stemness maintenance and differentiation fate of NSCs remain unclear in the infant brain. The present study demonstrated that 1 µM BPA promoted human NSC proliferation and stemness, without significantly increasing apoptosis. The Chip-seq experiments demonstrated that both the cell cycle and the TGF-ß signaling pathway were accelerated after treatment with 1 µM BPA. Subsequently, estrogen-related receptor α (ERRα) gene knockout cell lines were constructed using CRISPR/Cas9. Further western blotting and chromatin immunoprecipitation-PCR experiments demonstrated that BPA maintained cell stemness by binding to an EERα receptor and activating the TGF-ß1 signaling pathway, including the downstream factors Aurora kinases B and Id2. In conclusion, the stemness of NSCs could be maintained by BPA at 1 µM through the activation of the ERRα and TGF-ß1 signaling pathways and could restrain the differentiation of NSCs into neurons. The present research further clarified the mechanism of BPA toxicity on NSCs from the novel perspective of ERRα and TGF-ß1 signaling pathways regulated by BPA and provided insights into potential novel methods of prevention and therapy for neurogenic diseases.

8.
Stem Cell Reports ; 16(1): 75-88, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33357407

RESUMO

Serotonin receptor 6 (5-HT6R), a typical G protein-coupled receptor (GPCR) mainly expressed in the neurogenic area with constitutive activity, is of particular interest as a promising target for emotional impairment. Here, we found that 5-HT6R was highly expressed in human NSCs and activation of the receptor promoted self-renewal of human NSCs, and thus induced the expansion and folding of human cerebral organoids; dysfunction of receptor or inhibition of its constitutive activity resulted in the premature differentiation of NSCs, which ultimately depleted the NSC pool. The following mechanistic study revealed that EPAC-CREB signaling was involved in 5-HT6R regulation. Furthermore, we showed that mice with genetic deletion of 5-HT6R or knockin A268R mutant presented depression-like behaviors and impaired hippocampal neurogenesis for progressive decrease of the NSC pool. Thus, this study indicates that the modulation of 5-HT6R and its constitutive activity may provide a therapeutic alternative to alleviate depression.


Assuntos
Encéfalo/metabolismo , Depressão/patologia , Organoides/metabolismo , Receptores de Serotonina/metabolismo , Animais , Encéfalo/citologia , Proteína de Ligação a CREB/metabolismo , Diferenciação Celular , Autorrenovação Celular/efeitos dos fármacos , Etilaminas/farmacologia , Edição de Genes , Humanos , Indóis/farmacologia , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese , Organoides/citologia , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores de Serotonina/química , Receptores de Serotonina/genética , Transdução de Sinais , Proteínas rab1 de Ligação ao GTP/metabolismo
9.
Int J Mol Sci ; 21(24)2020 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-33348804

RESUMO

Stemness and apoptosis may highlight the dichotomy between regeneration and demise in the complex pathway proceeding from ontogenesis to the end of life. In the last few years, the concept has emerged that the same microRNAs (miRNAs) can be concurrently implicated in both apoptosis-related mechanisms and cell differentiation. Whether the differentiation process gives rise to the architecture of brain areas, any long-lasting perturbation of miRNA expression can be related to the occurrence of neurodevelopmental/neuropathological conditions. Moreover, as a consequence of neural stem cell (NSC) transformation to cancer stem cells (CSCs), the fine modulation of distinct miRNAs becomes necessary. This event implies controlling the expression of pro/anti-apoptotic target genes, which is crucial for the management of neural/neural crest-derived CSCs in brain tumors, neuroblastoma, and melanoma. From a translational point of view, the current progress on the emerging miRNA-based neuropathology therapeutic applications and antitumor strategies will be disclosed and their advantages and shortcomings discussed.


Assuntos
Apoptose , Diferenciação Celular , MicroRNAs/genética , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neurais/patologia , Animais , Humanos , Neoplasias/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neurais/metabolismo
10.
Int J Mol Sci ; 21(11)2020 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-32486466

RESUMO

In Alzheimer disease (AD) patients, degeneration of the cholinergic system utilizing acetylcholine for memory acquisition is observed. Since AD therapy using acetylcholinesterase (AChE) inhibitors are only palliative for memory deficits without slowing or reversing disease progress, there is a need for effective therapies, and stem cell-based therapeutic approaches targeting AD should fulfill this requirement. We established a human neural stem cell (NSC) line encoding choline acetyltransferase (ChAT) gene, an acetylcholine-synthesizing enzyme. APPswe/PS1dE9 AD model mice transplanted with the F3.ChAT NSCs exhibited improved cognitive function and physical activity. Transplanted F3.ChAT NSCs in the AD mice differentiated into neurons and astrocytes, produced ChAT protein, increased the ACh level, and improved the learning and memory function. F3.ChAT cell transplantation reduced Aß deposits by recovering microglial function; i.e., the down-regulation of ß-secretase and inflammatory cytokines and up-regulation of Aß-degrading enzyme neprilysin. F3.ChAT cells restored growth factors (GFs) and neurotrophic factors (NFs), and they induced the proliferation of NSCs in the host brain. These findings indicate that NSCs overexpressing ChAT can ameliorate complex cognitive and physical deficits of AD animals by releasing ACh, reducing Aß deposit, and promoting neuroregeneration by the production of GFs/NFs. It is suggested that NSCs overexpressing ChAT could be a candidate for cell therapy in advanced AD therapy.


Assuntos
Acetilcolina/biossíntese , Peptídeos beta-Amiloides/metabolismo , Colina O-Acetiltransferase/metabolismo , Transtornos Cognitivos/terapia , Células-Tronco Neurais/metabolismo , Regeneração , Precursor de Proteína beta-Amiloide/genética , Animais , Astrócitos/metabolismo , Encéfalo/metabolismo , Proliferação de Células , Terapia Baseada em Transplante de Células e Tecidos , Cognição , Hipocampo/metabolismo , Humanos , Transtornos da Memória/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/metabolismo , Fatores de Crescimento Neural/metabolismo , Células-Tronco Neurais/citologia , Neurônios/metabolismo , Presenilina-1/genética , Receptores Colinérgicos/metabolismo
11.
Mol Cells ; 43(6): 551-571, 2020 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-32522891

RESUMO

Nuclear receptor-related 1 (Nurr1) protein has been identified as an obligatory transcription factor in midbrain dopaminergic neurogenesis, but the global set of human NURR1 target genes remains unexplored. Here, we identified direct gene targets of NURR1 by analyzing genome-wide differential expression of NURR1 together with NURR1 consensus sites in three human neural stem cell (hNSC) lines. Microarray data were validated by quantitative PCR in hNSCs and mouse embryonic brains and through comparison to published human data, including genome-wide association study hits and the BioGPS gene expression atlas. Our analysis identified ~40 NURR1 direct target genes, many of them involved in essential protein modules such as synapse formation, neuronal cell migration during brain development, and cell cycle progression and DNA replication. Specifically, expression of genes related to synapse formation and neuronal cell migration correlated tightly with NURR1 expression, whereas cell cycle progression correlated negatively with it, precisely recapitulating midbrain dopaminergic development. Overall, this systematic examination of NURR1-controlled regulatory networks provides important insights into this protein's biological functions in dopamine-based neurogenesis.


Assuntos
Pontos de Checagem do Ciclo Celular/genética , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla , Células-Tronco Neurais/metabolismo , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo , Sinapses/metabolismo , Animais , Sequência de Bases , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Linhagem da Célula/genética , Movimento Celular/genética , Ontologia Genética , Humanos , Mesencéfalo/embriologia , Camundongos , Células-Tronco Neurais/citologia , Neurogênese/genética , Membro 2 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Doença de Parkinson/genética , Regiões Promotoras Genéticas/genética , Reprodutibilidade dos Testes , Ativação Transcricional/genética
12.
Curr Alzheimer Res ; 16(10): 934-949, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31642778

RESUMO

BACKGROUND: Amyloid-ß42 oligomers (Aß42O), the proximate effectors of neurotoxicity observed in Alzheimer's disease (AD), can induce mitochondrial oxidative stress and impair mitochondrial function besides causing mitochondrial DNA (mtDNA) damage. Aß42O also regulate the proliferative and differentiative properties of stem cells. OBJECTIVE: We aimed to study whether Aß42O-induced mtDNA damage is involved in the regulation of stem cell differentiation. METHOD: Human iPSCs-derived neural stem cell (NSC) was applied to investigate the effect of Aß42O on reactive oxygen species (ROS) production and DNA damage using mitoSOX staining and long-range PCR lesion assay, respectively. mtDNA repair activity was measured by non-homologous end joining (NHEJ) in vitro assay using mitochondria isolates and the expression and localization of NHEJ components were determined by Western blot and immunofluorescence assay. The expressions of Tuj-1 and GFAP, detected by immunofluorescence and qPCR, respectively, were examined as an index of neurons and astrocytes production. RESULTS: We show that in NSC Aß42O treatment induces ROS production and mtDNA damage and impairs DNA end joining activity. NHEJ components, such as Ku70/80, DNA-PKcs, and XRCC4, are localized in mitochondria and silencing of XRCC4 significantly exacerbates the effect of Aß42O on mtDNA integrity. On the contrary, pre-treatment with Phytic Acid (IP6), which specifically stimulates DNA-PK-dependent end-joining, inhibits Aß42O-induced mtDNA damage and neuronal differentiation alteration. CONCLUSION: Aß42O-induced mtDNA repair impairment may change cell fate thus shifting human NSC differentiation toward an astrocytic lineage. Repair stimulation counteracts Aß42O neurotoxicity, suggesting mtDNA repair pathway as a potential target for the treatment of neurodegenerative disorders like AD.


Assuntos
Peptídeos beta-Amiloides/toxicidade , Diferenciação Celular/fisiologia , Reparo do DNA/fisiologia , DNA Mitocondrial/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Neurais/metabolismo , Fragmentos de Peptídeos/toxicidade , Diferenciação Celular/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos
13.
Biomolecules ; 9(10)2019 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-31547190

RESUMO

Hyaluronic acid (HA) is a polysaccharide polymer frequently used as a starting material to fabricate hydrogels, especially for recapitulating the brain's extracellular matrix (ECM) for in vitro neural stem cell (NSC) cultures. Here, we report the successful synthesis of a methacrylated HA (MeHA) polymer from an inexpensive cosmetic-grade hyaluronan starting material. The MeHA polymers synthesized from cosmetic-grade HA yielded similar chemical purity to those from pharmaceutical/research-grade HA reported in the literature. Crosslinked MeHA (x-MeHA) hydrogels were formed using radical polymerization which resulted in mechanical properties matching previously reported mechanical property ranges for enhanced neuronal differentiation of NSCs. We assessed cellular adhesion, spreading, proliferation, and stiffness-dependent neuronal differentiation properties of ReNcell VM human neural stem cells (hNSCs) and compared our results to studies reported in the literature (that utilized non-human and human pluripotent cell-derived NSCs).


Assuntos
Cosméticos/química , Ácido Hialurônico/química , Células-Tronco Neurais/citologia , Adesão Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cosméticos/farmacologia , Análise Custo-Benefício , Humanos , Ácido Hialurônico/farmacologia , Hidrogéis/química
14.
ACS Appl Mater Interfaces ; 11(19): 17247-17255, 2019 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-31009192

RESUMO

As neural stem cells (NSCs) interact with biophysical cues from their niche during development, it is important to understand the biomolecular mechanism of how the NSCs process these biophysical cues to regulate their behaviors. In particular, anisotropic geometric cues in micro-/nanoscale have been utilized to investigate the biophysical effect of the structure on NSCs behaviors. Here, a series of new nanoscale anisotropic wrinkle structures with the a range of wavelength scales (from 50 nm to 37 µm) was developed to demonstrate the effect of the anisotropic nanostructure on the fate commitment of NSCs. Intriguingly, two distinct characteristic length scales promoted the neurogenesis. Each wavelength scale showed a striking variation in terms of dependency on the directionality of the structures, suggesting the existence of at least two different ways in the processing of anisotropic geometries for neurogenesis. Furthermore, the combined effect of the two distinctive length scales was observed by employing hierarchical multiscale wrinkle structures with two characteristic neurogenesis-promoting wavelengths. Taken together, the wrinkle structure system developed in this study can serve as an effective platform to advance the understanding of how cells sense anisotropic geometries for their specific cellular behaviors. Furthermore, this could provide clues for improving nerve regeneration system of stem cell therapies.


Assuntos
Nanoestruturas/química , Regeneração Nervosa , Células-Tronco Neurais/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Anisotropia , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem da Célula/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Quinase 1 de Adesão Focal/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Células-Tronco Neurais/metabolismo , Transplante de Células-Tronco
15.
Dev Cell ; 46(1): 85-101.e8, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29974866

RESUMO

Neural stem cells (NSCs) constitute an endogenous reservoir for neurons that could potentially be harnessed for regenerative therapies in disease contexts such as neurodegeneration. However, in Alzheimer's disease (AD), NSCs lose plasticity and thus possible regenerative capacity. We investigate how NSCs lose their plasticity in AD by using starPEG-heparin-based hydrogels to establish a reductionist 3D cell-instructive neuro-microenvironment that promotes the proliferative and neurogenic ability of primary and induced human NSCs. We find that administration of AD-associated Amyloid-ß42 causes classical neuropathology and hampers NSC plasticity by inducing kynurenic acid (KYNA) production. Interleukin-4 restores NSC proliferative and neurogenic ability by suppressing the KYNA-producing enzyme Kynurenine aminotransferase (KAT2), which is upregulated in APP/PS1dE9 mouse model of AD and in postmortem human AD brains. Thus, our culture system enables a reductionist investigation of regulation of human NSC plasticity for the identification of potential therapeutic targets for intervention in AD.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Plasticidade Celular/fisiologia , Interleucina-4/metabolismo , Células-Tronco Neurais/citologia , Neurogênese/fisiologia , Adulto , Idoso de 80 Anos ou mais , Doença de Alzheimer , Animais , Encéfalo/metabolismo , Proliferação de Células/fisiologia , Células Cultivadas , Modelos Animais de Doenças , Feminino , Humanos , Ácido Cinurênico/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Pessoa de Meia-Idade , Células-Tronco Neurais/fisiologia , Neurônios/citologia , Transaminases/metabolismo , Ativação Transcricional/genética , Adulto Jovem
16.
Exp Cell Res ; 370(2): 680-691, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30048616

RESUMO

Numerous chemicals including environmental toxicants and drugs have not been fully evaluated for developmental neurotoxicity. A key gap exists in the ability to predict accurately and robustly in vivo outcomes based on in vitro assays. This is particularly the case for predicting the toxicity of chemicals on the developing human brain. A critical need for such in vitro assays is choice of a suitable model cell type. To that end, we have performed high-throughput in vitro assessment of proliferation and differentiation of human neural stem cells (hNSCs). Conventional in vitro assays typically use immunofluorescence staining to quantify changes in cell morphology and expression of neural cell-specific biomarkers, which is often time-consuming and subject to variable specificities of available antibodies. To alleviate these limitations, we developed a miniaturized, three-dimensional (3D) hNSC culture with ReNcell VM on microarray chip platforms and established a high-throughput promoter-reporter assay system using recombinant lentiviruses on hNSC spheroids to assess cell viability, self-renewal, and differentiation. Optimum cell viability and spheroid formation of 3D ReNcell VM culture were observed on a micropillar chip over a period of 9 days in a mixture of 0.75% (w/v) alginate and 1 mg/mL growth factor reduced (GFR) Matrigel with 25 mM CaCl2 as a crosslinker for alginate. In addition, 3D ReNcell VM culture exhibited self-renewal and differentiation on the microarray chip platform, which was efficiently monitored by enhanced green fluorescent protein (EGFP) expression of four NSC-specific biomarkers including sex determining region Y-box 2 (SOX2), glial fibrillary acidic protein (GFAP), synapsin1, and myelin basic protein (MBP) with the promoter-reporter assay system.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Técnicas de Cultura de Células/métodos , Sobrevivência Celular/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Humanos , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Análise Serial de Proteínas/métodos
17.
Mol Autism ; 9: 38, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29951184

RESUMO

Background: MicroRNAs (miRNAs) are small, non-coding RNAs that regulate gene expression at the post-transcriptional level. miRNAs have emerged as important modulators of brain development and neuronal function and are implicated in several neurological diseases. Previous studies found miR-146a upregulation is the most common miRNA deregulation event in neurodevelopmental disorders such as autism spectrum disorder (ASD), epilepsy, and intellectual disability (ID). Yet, how miR-146a upregulation affects the developing fetal brain remains unclear. Methods: We analyzed the expression of miR-146a in the temporal lobe of ASD children using Taqman assay. To assess the role of miR-146a in early brain development, we generated and characterized stably induced H9 human neural stem cell (H9 hNSC) overexpressing miR-146a using various cell and molecular biology techniques. Results: We first showed that miR-146a upregulation occurs early during childhood in the ASD brain. In H9 hNSC, miR-146a overexpression enhances neurite outgrowth and branching and favors differentiation into neuronal like cells. Expression analyses revealed that 10% of the transcriptome was deregulated and organized into two modules critical for cell cycle control and neuronal differentiation. Twenty known or predicted targets of miR-146a were significantly deregulated in the modules, acting as potential drivers. The two modules also display distinct transcription profiles during human brain development, affecting regions relevant for ASD including the neocortex, amygdala, and hippocampus. Cell type analyses indicate markers for pyramidal, and interneurons are highly enriched in the deregulated gene list. Up to 40% of known markers of newly defined neuronal lineages were deregulated, suggesting that miR-146a could participate also in the acquisition of neuronal identities. Conclusion: Our results demonstrate the dynamic roles of miR-146a in early neuronal development and provide new insight into the molecular events that link miR-146a overexpression to impaired neurodevelopment. This, in turn, may yield new therapeutic targets and strategies.


Assuntos
Transtorno do Espectro Autista/genética , MicroRNAs/genética , Células-Tronco Neurais/metabolismo , Neurogênese , Transtorno do Espectro Autista/metabolismo , Linhagem Celular , Linhagem da Célula , Criança , Feminino , Humanos , Masculino , MicroRNAs/metabolismo , Células-Tronco Neurais/citologia , Lobo Temporal/citologia , Lobo Temporal/metabolismo , Regulação para Cima
18.
Methods ; 133: 81-90, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29050826

RESUMO

Neural stem cell (NSC) cultures have been considered technically challenging for time-lapse analysis due to high motility, photosensitivity, and growth at confluent densities. We have tested feasibility of long-term live-cell time-lapse analysis for NSC migration and differentiation studies. Here, we describe a method to study the dynamics of cell cycle, migration, and lineage selection in cultured multipotent mouse or human NSCs using single-cell tracking during a long-term, 7-14 day live-cell time-lapse analysis. We used in-house made PDMS inserts with five microwells on a glass coverslip petri-dish to constrain NSC into the area of acquisition during long-term live-cell imaging. In parallel, we have defined image acquisition settings for single-cell tracking of cell cycle dynamics using Fucci-reporter mouse NSC for 7 days as well as lineage selection and migration using human NSC for 14 days. Overall, we show that adjustments of live-cell analysis settings can extend the time period of single-cell tracking in mouse or human NSC from 24-72 h up to 7-14 days and potentially longer. However, we emphasize that experimental use of repeated fluorescence imaging will require careful consideration of controls during acquisition and analysis.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Neurais/citologia , Análise de Célula Única/métodos , Imagem com Lapso de Tempo/métodos , Linhagem da Célula/fisiologia , Movimento Celular/fisiologia , Rastreamento de Células/métodos , Humanos , Células-Tronco Neurais/fisiologia
19.
Cell Transplant ; 26(7): 1247-1261, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28933218

RESUMO

Traumatic brain injury (TBI) is one of the leading causes of death and disability in the population worldwide, with a broad spectrum of symptoms and disabilities. Posttraumatic hyperexcitability is one of the most common neurological disorders that affect people after a head injury. A reliable animal model of posttraumatic hyperexcitability induced by TBI which allows one to test effective treatment strategies is yet to be developed. To address these issues, in the present study, we tested human embryonic stem cell-derived neural stem cell (NSC) transplantation in an animal model of posttraumatic hyperexcitability in which the brain injury was produced in one hemisphere of immunodeficient athymic nude rats by controlled cortical impact, and spontaneous seizures were produced by repeated electrical stimulation (kindling) in the contralateral hemisphere. At 14 wk posttransplantation, we report human NSC (hNSC) survival and differentiation into all 3 neural lineages in both sham and injured animals. We observed twice as many surviving hNSCs in the injured versus sham brain, and worse survival on the kindled side in both groups, indicating that kindling/seizures are detrimental to survival or proliferation of hNSCs. We also replicated our previous finding that hNSCs can ameliorate deficits on the novel place recognition task,33 but such improvements are abolished following kindling. We found no significant differences pre- or post-kindling on the elevated plus maze. No significant correlations were observed between hNSC survival and cognitive performance on either task. Together these findings suggest that Shef6-derived hNSCs may be beneficial as a therapy for TBI, but not in animals or patients with posttraumatic hyperexcitability.


Assuntos
Lesões Encefálicas Traumáticas/fisiopatologia , Lesões Encefálicas Traumáticas/terapia , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Neurais/transplante , Transplante de Células-Tronco , Animais , Lesões Encefálicas Traumáticas/patologia , Contagem de Células , Diferenciação Celular , Linhagem da Célula , Sobrevivência Celular , Cognição , Modelos Animais de Doenças , Humanos , Excitação Neurológica , Masculino , Aprendizagem em Labirinto , Células-Tronco Neurais/citologia , Ratos Nus , Análise e Desempenho de Tarefas
20.
Stem Cell Reports ; 8(6): 1534-1548, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28479305

RESUMO

Multipotent human central nervous system-derived neural stem cells transplanted at doses ranging from 10,000 (low) to 500,000 (very high) cells differentiated predominantly into the oligodendroglial lineage. However, while the number of engrafted cells increased linearly in relationship to increasing dose, the proportion of oligodendrocytic cells declined. Increasing dose resulted in a plateau of engraftment, enhanced neuronal differentiation, and increased distal migration caudal to the transplantation sites. Dose had no effect on terminal sensory recovery or open-field locomotor scores. However, total human cell number and decreased oligodendroglial proportion were correlated with hindlimb girdle coupling errors. Conversely, greater oligodendroglial proportion was correlated with increased Ab step pattern, decreased swing speed, and increased paw intensity, consistent with improved recovery. These data suggest that transplant dose, and/or target niche parameters can regulate donor cell engraftment, differentiation/maturation, and lineage-specific migration profiles.


Assuntos
Diferenciação Celular , Células-Tronco Neurais/transplante , Neurônios/citologia , Oligodendroglia/citologia , Traumatismos da Medula Espinal/terapia , Animais , Antígenos Nucleares/metabolismo , Linhagem da Célula , Movimento Celular , Células Cultivadas , Microambiente Celular , Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Oligodendroglia/metabolismo , Recuperação de Função Fisiológica , Nicho de Células-Tronco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA