Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cereb Blood Flow Metab ; 43(10): 1656-1671, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37395346

RESUMO

Ischemic stroke (IS) induces neurological damage due to cerebrovascular occlusion. Restoring blood perfusion to the ischemic brain area in a timely fashion is the most effective treatment strategy. Hypoxia is an effective way of restoring blood perfusion by improving cerebrovascular microcirculation, while the effect varies greatly depending on hypoxic mode. This study aimed to screen for the optimal hypoxic mode to improve cerebrovascular microcirculation and prevent IS. Here, we found that compared with continuous hypoxia (CH), intermittent hypoxia (IH) significantly improved cerebral blood flow and oxygen saturation in mice without causing neurological impairment. By analyzing cerebrovascular microcirculation from mice, we found that the IH mode (13%, 5*10) with 13% O2, 5 min interval, and 10 cycles per day significantly improved the cerebrovascular microcirculation by promoting angiogenesis without affecting the integrity of the blood-brain barrier. In addition, IH (13%, 5*10) treatment of distal middle cerebral artery occlusion (dMCAO) mice significantly alleviated neurological dysfunction and reduced cerebral infarct volume by improving cerebrovascular microcirculation. CH had none of these positive effects. In summary, our study screened for an appropriate intermittent hypoxic mode that could improve cerebrovascular microcirculation, laying a theoretical foundation for the prevention and treatment of IS in clinical practice.


Assuntos
Hipóxia-Isquemia Encefálica , Hipóxia , Camundongos , Animais , Hipóxia-Isquemia Encefálica/prevenção & controle , Encéfalo/irrigação sanguínea , Barreira Hematoencefálica , Infarto da Artéria Cerebral Média , Circulação Cerebrovascular/fisiologia
2.
Aging Dis ; 14(2): 370-385, 2023 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-37008044

RESUMO

Cerebrovascular microcirculation is essential for maintaining the physiological functions of the brain. The brain can be protected from stress injury by remodeling the microcirculation network. Angiogenesis is a type of cerebral vascular remodeling. It is an effective approach to improve the blood flow of the cerebral microcirculation, which is necessary for preventing and treating various neurological disorders. Hypoxia is one of the most important regulators of angiogenesis, affecting the sprouting, proliferation, and maturation stages of angiogenesis. Moreover, hypoxia negatively affects cerebral vascular tissue by impairing the structural and functional integrity of the blood-brain barrier and vascular-nerve decoupling. Therefore, hypoxia has a dual effect on blood vessels and is affected by confounding factors including oxygen concentration, hypoxia duration, and hypoxia frequency and extent. Establishing an optimal model that promotes cerebral microvasculogenesis without causing vascular injury is essential. In this review, we first elaborate on the effects of hypoxia on blood vessels from two different perspectives: (1) the promotion of angiogenesis and (2) cerebral microcirculation damage. We further discuss the factors influencing the dual role of hypoxia and emphasize the benefits of moderate hypoxic irritation and its potential application as an easy, safe, and effective treatment for multiple nervous system disorders.

3.
CNS Neurosci Ther ; 29(2): 544-558, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36514210

RESUMO

AIMS: The etiology of Parkinson's disease (PD) is complex and the mechanism is unclear. It has become a top priority to find common factors that induce and affect PD pathology. We explored the key role of hypoxia in promoting the pathological propagation of α-synuclein (α-syn) and the progression of PD. METHODS: We performed PD modeling by conducting intracranial stereotaxic surgery in the unilateral striatum of mice. We then measured protein aggregation in vitro. The rotarod and pole tests were employed next to measure the damage of the phenotype. Pathological deposition and autophagy were also observed by immunofluorescence staining and protein levels measured by western blotting. RESULTS: We demonstrated that short-term hypoxia activated phosphorylated (p)-α-syn in mice. We confirmed that p-α-syn was more readily formed aggregates than α-syn in vitro. Furthermore, we found that hypoxia promoted the activation and propagation of endogenous α-syn, contributing to the earlier degeneration of dopaminergic neurons in the substantia nigra and the deposition of p-α-syn in our animal model. Finally, autophagy inhibition contributed to the above pathologies. CONCLUSION: Hypoxia was shown to accelerate the pathological progression and damage phenotype in PD model mice. The results provided a promising research target for determining common interventions for PD in the future.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Camundongos , Animais , alfa-Sinucleína/metabolismo , Neurônios Dopaminérgicos/metabolismo , Doença de Parkinson/genética , Substância Negra , Corpo Estriado/metabolismo
4.
CNS Neurosci Ther ; 29(1): 202-215, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36401601

RESUMO

BACKGROUND: Central nervous system diseases are associated with hypoxia, which usually cause irreversible nerve damage, but the underlying mechanism is unclear and effective intervention strategies are lacking. This study was designed to explore the mechanism and treatment strategy of hypoxia-induced nerve injury. METHODS: In this study, 13% O2 was used to treat mice for 0, 1, 3 7, and 14 days, Morris water maze and other animal behavior experiments were used to evaluate the neurological function of mice. TUNEL, BrdU, PCNA, DCX, and SOX2 staining were used to observe the apoptosis and proliferation of mouse neurons. RT-PCR and Iba1 staining were used to evaluate the release of inflammatory factors IL-1ß, IL-6, and TNF-α and the activation of microglia. RESULTS: Short-term hypoxia promotes neurogenesis, while long-term hypoxia inhibits neurogenesis. The changes in hypoxia-induced neurogenesis were positively correlated with neurological functions, but negatively correlated with apoptosis. Moreover, intermittent hypoxic conditioning restored long-term hypoxia-induced neurological dysfunction by promoting neural stem cell generation and inhibiting the release of inflammatory factors IL-1ß, IL-6, and TNF-α and the activation of microglia. CONCLUSION: Hypoxia promoted neurogenesis in a time-dependent manner, and intermittent hypoxic conditioning exerted a neuroprotective effect through promoting neural stem cell generation and suppressing inflammation induced by long-term hypoxia stress, which provided a novel concept to develop a treatment for hypoxia-related brain injury.


Assuntos
Hipóxia Encefálica , Fator de Necrose Tumoral alfa , Camundongos , Animais , Interleucina-6 , Hipóxia , Neurônios , Microglia
5.
Int J Mol Sci ; 23(22)2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36430571

RESUMO

Hypoxic stress occurs in various physiological and pathological states, such as aging, disease, or high-altitude exposure, all of which pose a challenge to many organs in the body, necessitating adaptation. However, the exact mechanisms by which hypoxia affects advanced brain function (learning and memory skills in particular) remain unclear. In this study, we investigated the effects of hypoxic stress on hippocampal function. Specifically, we studied the effects of the dysfunction of mitochondrial oxidative phosphorylation using global proteomics. First, we found that hypoxic stress impaired cognitive and motor abilities, whereas it caused no substantial changes in the brain morphology or structure of mice. Second, bioinformatics analysis indicated that hypoxia affected the expression of 516 proteins, of which 71.1% were upregulated and 28.5% were downregulated. We demonstrated that mitochondrial function was altered and manifested as a decrease in NADH dehydrogenase (ubiquinone) 1 alpha subcomplex 4 expression, accompanied by increased reactive oxygen species generation, resulting in further neuronal injury. These results may provide some new insights into how hypoxic stress alters hippocampal function via the dysfunction of mitochondrial oxidative phosphorylation.


Assuntos
Mitocôndrias , Proteômica , Camundongos , Animais , Mitocôndrias/metabolismo , Hipocampo/metabolismo , Hipóxia/metabolismo , Neurônios/metabolismo
6.
Cell Death Discov ; 8(1): 473, 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36450714

RESUMO

Chronic hypoxia leads to irreversible cognitive impairment, primarily due to hippocampal neurodegeneration, for which the underlying mechanism remains poorly understood. We administered hypoxia (13%) to C57BL mice for 1-14 days in this study. Chronic hypoxia for 7 or 14 d, but not 1 or 3 d, resulted in alpha-synuclein hyperphosphorylation at serine129 (α-Syn p-S129) and protein aggregation, hippocampal neurodegeneration, and cognitive deficits, whereas the latter could be prevented by alpha-synuclein knockdown or an administered short peptide competing at α-Syn S129. These results suggest that α-Syn p-S129 mediates hippocampal degeneration and cognitive impairment following chronic hypoxia. Furthermore, we found that chronic hypoxia enhanced ceramide catabolism by inducing hypoxia-inducible factor (HIF)-2α and HIF-2α-dependent transcriptional activation of alkaline ceramidase 2 (Acer2). Thus, the enzymatic activity of protein phosphatase 2A (PP2A), a specific phosphatase for α-syn, is inhibited, leading to the sustained induction of α-Syn p-S129. Finally, we found that intermittent hypoxic preconditioning protected against subsequent chronic hypoxia-induced hippocampal neurodegeneration and cognitive impairment by preventing α-Syn p-S129. These results proved the critical role of α-syn pathology in chronic hypoxia-afforded cognitive impairment and revealed a novel mechanism underlying α-syn hyperphosphorylation during chronic hypoxia. The findings bear implications in developing novel therapeutic interventions for chronic hypoxia-related brain disorders.

7.
CNS Neurosci Ther ; 27(8): 869-882, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34237192

RESUMO

As the organ with the highest demand for oxygen, the brain has a poor tolerance to ischemia and hypoxia. Despite severe ischemia/hypoxia induces the occurrence and development of various central nervous system (CNS) diseases, sublethal insult may induce strong protection against subsequent fatal injuries by improving tolerance. Searching for potential measures to improve brain ischemic/hypoxic is of great significance for treatment of ischemia/hypoxia related CNS diseases. Ischemic/hypoxic preconditioning (I/HPC) refers to the approach to give the body a short period of mild ischemic/hypoxic stimulus which can significantly improve the body's tolerance to subsequent more severe ischemia/hypoxia event. It has been extensively studied and been considered as an effective therapeutic strategy in CNS diseases. Its protective mechanisms involved multiple processes, such as activation of hypoxia signaling pathways, anti-inflammation, antioxidant stress, and autophagy induction, etc. As a strategy to induce endogenous neuroprotection, I/HPC has attracted extensive attention and become one of the research frontiers and hotspots in the field of neurotherapy. In this review, we discuss the basic and clinical research progress of I/HPC on CNS diseases, and summarize its mechanisms. Furthermore, we highlight the limitations and challenges of their translation from basic research to clinical application.


Assuntos
Encéfalo/irrigação sanguínea , Hipóxia-Isquemia Encefálica/terapia , Precondicionamento Isquêmico/métodos , Doenças do Sistema Nervoso/terapia , Neuroproteção/fisiologia , Animais , Autofagia/fisiologia , Encéfalo/metabolismo , Humanos , Hipóxia-Isquemia Encefálica/metabolismo , Doenças do Sistema Nervoso/metabolismo , Resultado do Tratamento
8.
Cell Death Dis ; 10(9): 646, 2019 09 09.
Artigo em Inglês | MEDLINE | ID: mdl-31501413

RESUMO

Direct conversion of readily available non-neural cells from patients into induced neurons holds great promise for neurological disease modeling and cell-based therapy. Olfactory ensheathing cells (OECs) is a unique population of glia in olfactory nervous system. Based on the regeneration-promoting properties and the relative clinical accessibility, OECs are attracting increasing attention from neuroscientists as potential therapeutic agents for use in neural repair. Here, we report that OECs can be directly, rapidly and efficiently reprogrammed into neuronal cells by the single transcription factor Neurogenin 2 (NGN2). These induced cells exhibit typical neuronal morphologies, express multiple neuron-specific markers, produce action potentials, and form functional synapses. Genome-wide RNA-sequencing analysis shows that the transcriptome profile of OECs is effectively reprogrammed towards that of neuronal lineage. Importantly, these OEC-derived induced neurons survive and mature after transplantation into adult mouse spinal cords. Taken together, our study provides a direct and efficient strategy to quickly obtain neuronal cells from adult OECs, suggestive of promising potential for personalized disease modeling and cell replacement-mediated therapeutic approaches to neurological disorders.


Assuntos
Regeneração Nervosa/fisiologia , Bulbo Olfatório/fisiopatologia , Células Cultivadas , Humanos , Neurônios
9.
Cells ; 8(6)2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31212628

RESUMO

Direct conversion of non-neural cells into induced neurons holds great promise for brain repair. As the most common malignant tumor in the central nervous system, glioma is currently incurable due to its exponential growth and invasive behavior. Given that neurons are irreversible postmitotic cells, reprogramming glioma cells into terminally differentiated neuron-like cells represents a potential approach to inhibit brain tumor development. We here show that human glioma cells can be directly, rapidly and efficiently reprogrammed into terminally differentiated neuron-like cells by the single transcription factor ASCL1 (Achaete-scute complex-like 1, also known as MASH1). These induced cells exhibit typical neuron-like morphology and express multiple neuron-specific markers. Importantly, ASCL1-mediated neuronal reprogramming drives human glioma cells to exit the cell cycle and results in dramatic inhibition of proliferation, both in vitro and in vivo. Taken together, this proof-of-principle study demonstrates a potential strategy for impeding brain tumor development by ASCL1-induced direct neuronal reprogramming.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Reprogramação Celular , Glioma/patologia , Neurônios/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Linhagem Celular Tumoral , Proliferação de Células , Proteínas do Domínio Duplacortina , Regulação da Expressão Gênica , Glioma/metabolismo , Glioma/mortalidade , Humanos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurogênese , Neurônios/citologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Sinapsinas/metabolismo , Transplante Heterólogo , Tubulina (Proteína)/metabolismo
10.
Brain Behav Immun ; 80: 394-405, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30959174

RESUMO

Astrocytes become reactive in response to spinal cord injury (SCI) and ultimately form a histologically apparent glial scar at the lesion site. It is controversial whether astrocytic scar is detrimental or beneficial to the axonal regeneration and SCI repair. Therefore, much effort has focused on understanding the functions of reactive astrocytes. Here, we used a lentivirus-mediated herpes simplex thymidine kinase/ganciclovir (HSVtk/GCV) system to selectively kill scar-forming reactive proliferating astrocytes. The suicide gene expression was regulated by human glial fibrillary acidic protein (hGFAP) promoter, which is active primarily in astrocytes. Conditional ablation of reactive astrocytes in a mouse SCI model with crush injury impeded glial scar formation and resulted in widespread infiltration of inflammatory cells, increased neuronal loss, and severe tissue degeneration, which ultimately led to the failure of spontaneous functional recovery. These results suggest that reactive proliferating astrocytes play key roles in the healing process after SCI, shedding light on the potential benefit for the repair after central nervous system (CNS) injury.


Assuntos
Astrócitos/fisiologia , Mielite/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia , Regeneração da Medula Espinal/fisiologia , Animais , Cicatriz/etiologia , Cicatriz/fisiopatologia , Feminino , Camundongos Endogâmicos C57BL , Mielite/etiologia , Mielite/patologia , Neurônios/patologia , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/patologia
11.
Stem Cell Reports ; 12(2): 290-304, 2019 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-30713039

RESUMO

The adult CNS has poor ability to replace degenerated neurons following injury or disease. Recently, direct reprogramming of astrocytes into induced neurons has been proposed as an innovative strategy toward CNS repair. As a cell population that shows high diversity on physiological properties and functions depending on their spatiotemporal distribution, however, whether the astrocyte heterogeneity affect neuronal reprogramming is not clear. Here, we show that astrocytes derived from cortex, cerebellum, and spinal cord exhibit biological heterogeneity and possess distinct susceptibility to transcription factor-induced neuronal reprogramming. The heterogeneous expression level of NOTCH1 signaling in the different CNS regions-derived astrocytes is shown to be responsible for the neuronal reprogramming diversity. Taken together, our findings demonstrate that region-restricted astrocytes reveal different intrinsic limitation of the response to neuronal reprogramming.


Assuntos
Astrócitos/fisiologia , Reprogramação Celular/fisiologia , Neurônios/fisiologia , Animais , Astrócitos/metabolismo , Células Cultivadas , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais/fisiologia , Medula Espinal/metabolismo , Medula Espinal/fisiologia
12.
Gene ; 681: 26-35, 2019 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-30266499

RESUMO

BACKGROUND: Neural stem cells (NSCs) have unique biological characteristics such as continuous proliferation and multipotential differentiation, providing a possible method for restoration of central nervous system (CNS) function after injury or disease. NSCs and astrocytes share many similar biological properties including cell morphology and molecular expression and can trans-differentiate into each other under certain conditions. However, characteristic genes specifically expressed by NSCs have not been well described. METHODS: To provide insights into the characteristic expression of NSCs, bioinformatics analysis of two microarrays of mouse NSCs and astrocytes was performed. Compared to astrocytes, the differentially expressed genes (DEGs) in NSCs were identified and annotated by GO, KEGG and GSEA analysis, respectively. Then key genes were screened by protein-protein interaction (PPI) network and modules analysis, and were verified using multiple high-throughput sequencing resources. Finally, the expression difference between the two cell types was confirmed by Real-time Quantitative PCR (qPCR), western blotting and immunochemical analysis. RESULTS: In the present study, 282 and 250 NSC-enriched genes from two microarrays were identified and annotated respectively, and the 77 overlapping DEGs were then selected. From the PPI network 24 key genes in three modules were screened out. Importantly, sequencing data of tissues showed that these 24 key genes tended to be highly expressed in NSCs compared with astrocytes. Furthermore, qPCR and western blot analysis of cultured NSCs and astrocytes showed two genes (KIF2C and TOP2A) were not only differentially expressed in RNA level but also at the protein level. Importantly, the NSC-specific genes KIF2C and TOP2A were validated by immunohistochemistry in vivo. CONCLUSION: In present study, we identified 2 hub genes (KIF2C and TOP2A) that might serve as potential biomarkers for distinguishing NSCs from astrocytes, contributing to our comprehensive understanding of the biological properties and functions of NSCs.


Assuntos
Astrócitos/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Separação Celular/métodos , Células-Tronco Neurais/metabolismo , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Biomarcadores/análise , Células Cultivadas , Embrião de Mamíferos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes Controladores do Desenvolvimento , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia
13.
Sheng Li Xue Bao ; 69(6): 794-804, 2017 Dec 25.
Artigo em Chinês | MEDLINE | ID: mdl-29270596

RESUMO

Spinal cord injury (SCI) is a challenging medical problem in the field of neurology, showing high incidence rate, disability rate, treatment cost and low-aged trend. Despite the clinical application of drug intervention, surgical treatment and modern rehabilitation training, no ideal curative effect has been achieved. Therefore, future study is necessary to clarify detailed pathological mechanism of SCI and identify the potential target cells for therapeutic intervention. In the central nervous system (CNS), astrocytes are the most abundant and widely distributed glial cells which play multiple key roles in maintaining homeostasis of the CNS in physiological and pathological conditions. Increasing evidence indicates that astrocytes are ideal therapeutic target cells for SCI. Here, we review current knowledge of the roles of astrocytes in the pathological reaction after SCI, astroglial transplantation and astrocyte reprogramming.


Assuntos
Astrócitos/fisiologia , Traumatismos da Medula Espinal/terapia , Animais , Astrócitos/efeitos dos fármacos , Reprogramação Celular , Neuroglia/transplante , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/fisiopatologia
14.
Brain Res Bull ; 132: 10-19, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28499803

RESUMO

As a major class of glial cells, astrocytes have been indicated to play multi-roles in physiological and pathological brain. Astrocyte cultures derived from postnatal mouse brains have been extensively used to characterize their biological properties. However, the inability to culture adult mouse primary astrocytes has long stymied studies of function in adult brain. Here, we developed a protocol to successfully establish highly enriched astrocyte cultures from the brains of adult mouse. Cortical tissues were collected to prepare cell suspension by enzymatic digestion and mechanical dissociation, and then plated onto vessels pre-coated with gelatin and matrigel and cultured in DMEM medium containing 20% fetal bovine serum (FBS). Forskolin (FSK) and glial-derived neurotrophic factor (GDNF) were use to promote astrocyte proliferation and survival respectively. These adult astrocyte cultures were identified by immunocytochemical, immunobloting and PCR analysis. Furthermore, biological and functional analysis indicated that they possess the biochemical and physiological properties of astrocytes, suggestive of a useful cell model for astroglial studies in adult brain.


Assuntos
Astrócitos , Encéfalo , Técnicas de Cultura de Células/métodos , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Astrócitos/citologia , Astrócitos/patologia , Astrócitos/fisiologia , Western Blotting , Encéfalo/citologia , Encéfalo/fisiologia , Bromodesoxiuridina , Proliferação de Células/fisiologia , Células Cultivadas , Técnicas de Reprogramação Celular , Feminino , Imunofluorescência , Gliose/patologia , Gliose/fisiopatologia , Masculino , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Reação em Cadeia da Polimerase em Tempo Real
15.
Sheng Li Xue Bao ; 69(2): 207-217, 2017 Apr 25.
Artigo em Chinês | MEDLINE | ID: mdl-28435980

RESUMO

Glial cells, including astrocytes, oligodendrocyte progenitor cells (OPCs), NG2-glia, etc, are broadly distributed throughout the central nervous system (CNS). Also, it has been well known that glial cells play multi-roles in physiological and pathological processes in the CNS, such as maintaining homeostasis, providing neurotrophins for neurons and regulating neural signal transmission. Recently, increasing evidence showed that glial cells may also function as neural stem/progenitor cells and contribute to adult neurogenesis or neuroregeneration. In pathological conditions, for instance, astrocytes and OPCs could be activated to proliferate and differentiate. When cultured in vitro, they could form neurospheres which possess the ability to differentiate into astrocytes, oligodendrocytes and neurons. Additionally, forced expression of exogenous genes in astrocytes and NG2-glia can successfully reprogram them into neurons, which may also be suggestive of their stem/progenitor cell features. Here, we review current knowledge of the stem cell-like properties of glial cells, including what types of glial cells can function as stem/progenitor cells, how they can acquire the stem/progenitor potential and what progenies can be produced. These insights may foster a better understanding of glial cell biology and function in physiological or pathological processes in the CNS and lead to the idea of using the stem/progenitor-like glial cells as endogenous cell source for neural repair.


Assuntos
Células-Tronco Neurais/citologia , Neuroglia/citologia , Animais , Astrócitos/citologia , Diferenciação Celular , Sistema Nervoso Central , Humanos , Neurogênese , Neurônios/citologia , Oligodendroglia/citologia
16.
Org Biomol Chem ; 12(18): 2850-3, 2014 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-24643770

RESUMO

Triptycene-derived macrotricyclic polyether containing an anthracene unit is a powerful host for 1,2-bis(pyridium)ethane, diquat and 2,7-diazapyrenium salt with association constants of the 1 : 1 complexes at >10(5) M(-1). Crystal structures showed that π-π stacking interactions between the host and the guests play an important role in the formation of the stable complexes.

17.
Chem Commun (Camb) ; 48(90): 11076-8, 2012 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-23044486

RESUMO

Triptycene-based macrotricyclic host containing two dibenzo-30-crown-10 moieties could form ladder-like supramolecular poly[3]pseudorotaxanes with π-extended viologens in both high concentration solution and solid state.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...