Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Front Chem ; 8: 679, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33134242

RESUMO

Diketopyrrolopyrrole (DPP) and its derivatives have been widely studied in the past few years due to its intrinsic physical and chemical properties, such as strong electron-withdrawing, deep color, high charge carrier mobility, strong aggregation, good thermal-/photo-stability. In the 1970s, DPP was developed and used only in inks, paints, and plastics. Later, DPP containing materials were found to have potential other applications, typically in electronic devices, which attracted the attention of scientists. In this feature article, the synthesis pathway of DPP-based materials and their applications in organic field-effect transistors, photovoltaic devices, sensors, two photo-absorption materials, and others are reviewed, and possible future applications are discussed. The review outlines a theoretical scaffold for the development of conjugated DPP-based materials, which have multiple potential applications.

2.
Front Chem ; 8: 231, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32351932

RESUMO

High selectivity and sensitivity detection of fluoride anions (F-) in an organic solution by the naked eye has always been a challenge. In this investigation, a simple compound based on aminobenzodifuranone (ABDF) was designed and synthesized. Deprotonation of the amino moiety caused by F- is responsible for a color change from dark blue to various colors (colorless, yellow, orange, and red) in different common organic solvents due to a blue shift over 200 nm in the UV/Vis spectrum. The color change is quite visible to the naked eye under ambient light. The detection limit for F- can reach a concentration of as low as 5.0 × 10-7 M with high selectivity, even in a solution containing multiple anions.

3.
Biosci Rep ; 39(3)2019 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-30808715

RESUMO

Background: Intracranial aneurysm (IA) is a critical acquired cerebrovascular disease that may cause subarachnoid hemorrhage, and nuclear factor-κB (NF-κB)-mediated inflammation is involved in the pathogenesis of IA. Adenomatous polyposis coli (Apc) gene is a tumor suppressor gene associated with both familial and sporadic cancer. Herein, the purpose of our study is to validate effect of Apc gene on IA formation and rupture by regulating the NF-κB signaling pathway mediated inflammatory response. Methods: We collected IA specimens (from incarceration of IA) and normal cerebral arteries (from surgery of traumatic brain injury) to examine expression of Apc and the NF-κB signaling pathway related factors (NF-κB p65 and IκBα). ELISA was used to determine levels of monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß (IL-1ß), and IL-6. IA model was established in rats, and Apc-siRNA was treated to verify effect of Apc on IA formation and rupture. Next, regulation of Apc on the NF-κB signaling pathway was investigated. Results: Reduced expression of Apc and IκBα, and increased expression of NF-κB p65 were found in IA tissues. MCP-1, TNF-α, IL-1ß, and IL-6 exhibited higher levels in unruptured and ruptured IA, which suggested facilitated inflammatory responses. In addition, the IA rats injected with Apc-siRNA showed further enhanced activation of NF-κB signaling pathway, and up-regulated levels of MCP-1, TNF-α, IL-1ß, IL-6, MMP-2, and MMP-9 as well as extent of p65 phosphorylation in IA. Conclusion: Above all, Apc has the potential role to attenuate IA formation and rupture by inhibiting inflammatory response through repressing the activation of the NF-κB signaling pathway.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Citocinas/genética , Aneurisma Intracraniano/genética , NF-kappa B/genética , Ruptura Espontânea/genética , Transdução de Sinais/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Adulto , Idoso , Animais , Citocinas/metabolismo , Feminino , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Inflamação/genética , Inflamação/metabolismo , Aneurisma Intracraniano/metabolismo , Aneurisma Intracraniano/patologia , Masculino , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Ratos Sprague-Dawley , Ruptura Espontânea/metabolismo , Adulto Jovem
4.
Biomed Res Int ; 2018: 8354350, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29516010

RESUMO

Although extensive studies have identified large number of microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) in ischemic stroke, the RNA regulation network response to focal ischemia remains poorly understood. In this study, we simultaneously interrogate the expression profiles of lncRNAs, miRNAs, and mRNAs changes during focal ischemia induced by transient middle cerebral artery occlusion. A set of 1924 novel lncRNAs were identified and may involve brain injury and DNA repair as revealed by coexpression network analysis. Furthermore, many short interspersed elements (SINE) mediated lncRNA:mRNA duplexes were identified, implying that lncRNAs mediate Staufen1-mediated mRNA decay (SMD) which may play a role during focal ischemia. Moreover, based on the competitive endogenous RNA (ceRNA) hypothesis, a stroke regulatory ceRNA network which reveals functional lncRNA:miRNA:mRNA interactions was revealed in ischemic stroke. In brief, this work reports a large number of novel lncRNAs responding to focal ischemia and constructs a systematic RNA regulation network which highlighted the role of ncRNAs in ischemic stroke.


Assuntos
Isquemia/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Acidente Vascular Cerebral/genética , Animais , Modelos Animais de Doenças , Regulação da Expressão Gênica/genética , Redes Reguladoras de Genes/genética , Humanos , Isquemia/fisiopatologia , Estabilidade de RNA/efeitos dos fármacos , Ratos , Elementos Nucleotídeos Curtos e Dispersos , Acidente Vascular Cerebral/fisiopatologia
5.
Metab Brain Dis ; 32(5): 1427-1435, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28391551

RESUMO

Posttraumatic hydrocephalus (PTH) is a disorder of disturbed cerebrospinal fluid (CSF) dynamics after traumatic brain injury (TBI). It can lead to brain metabolic impairment and dysfunction and has a high risk of clinical deterioration and worse outcomes. The incidence and risk factors for the development of PTH after decompressive craniectomy (DC) has been assessed in previous studies, but rare studies identify patients with higher risk for PTH among all TBI patients. This study aimed to develop and validate a risk scoring system to predict PTH after TBI. Demographics, injury severity, duration of coma, radiologic findings, and DC were evaluated to determine the independent predictors of PTH during hospitalization until 6 months following TBI through logistic regression analysis. A risk stratification system was created by assigning a number of points for each predictor and validated in an independent cohort. The model accuracy was assessed by the area under the receiver operating characteristic curve (AUC). Of 526 patients in the derivation cohort, 57 (10.84%) developed PTH during 6 months follow up. Age > 50 yrs (Odd ratio [OR] = 1.91, 95% confidence interval [CI] 1.09-3.75, 4 points), duration of coma ≥1 w (OR = 5.68, 95% CI 2.57-13.47, 9 points), Fisher grade III (OR = 2.19, 95% CI 1.24-4.36, 5 points) or IV (OR = 3.87, 95% CI 1.93-8.43, 7 points), bilateral DC (OR = 6.13, 95% CI 2.82-18.14, 9 points), and extra herniation after DC (OR = 2.36, 95% CI 1.46-4.92, 5 points) were independently associated with PTH. Rates of PTH for the low- (0-12 points), intermediate- (13-22 points) and high-risk (23-34 points) groups were 1.16%, 35.19% and 78.57% (p < 0.0001). The corresponding rates in the validation cohort, where 17/175 (9.71%) developed PTH, were 1.35%, 37.50% and 81.82% (p < 0.0001). The risk score model exhibited good-excellent discrimination in both cohorts, with AUC of 0.839 versus 0.894 (derivation versus validation) and good calibration (Hosmer-Lemshow p = 0.56 versus 0.68). This model will be useful to identify patients at high risk for PTH who may be candidates for preventive interventions, and to improve their outcomes.


Assuntos
Hidrocefalia/epidemiologia , Hidrocefalia/etiologia , Adulto , Fatores Etários , Área Sob a Curva , China/epidemiologia , Estudos de Coortes , Craniectomia Descompressiva , Feminino , Escala de Coma de Glasgow , Hérnia/complicações , Hérnia/etiologia , Humanos , Hidrocefalia/diagnóstico por imagem , Masculino , Pessoa de Meia-Idade , Modelos Estruturais , Valor Preditivo dos Testes , Prognóstico , Curva ROC , Reprodutibilidade dos Testes , Estudos Retrospectivos , Medição de Risco , Fatores de Risco , Tomografia Computadorizada por Raios X
6.
Biomed Res Int ; 2017: 2957538, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28127553

RESUMO

Acetylation or deacetylation of chromatin proteins and transcription factors is part of a complex signaling system that is involved in the control of neurological disorders. Recent studies have demonstrated that histone deacetylases (HDACs) exert protective effects in attenuating neuronal injury after ischemic insults. Class IIa HDAC4 is highly expressed in the brain, and neuronal activity depends on the nucleocytoplasmic shuttling of HDAC4. However, little is known about HDAC4 and its roles in ischemic stroke. In this study, we report that phosphorylation of HDAC4 was remarkably upregulated after stroke and blockade of HDAC4 phosphorylation with GÖ6976 repressed stroke-induced angiogenesis. Phosphorylation of HDAC4 was also increased in endothelial cells hypoxia model and suppression of HDAC4 phosphorylation inhibited the tube formation and migration of endothelial cells in vitro. Furthermore, in addition to the inhibition of angiogenesis, blockade of HDAC4 phosphorylation suppressed the expression of genes downstream of HIF-VEGF signaling in vitro and in vivo. These data indicate that phosphorylated HDAC4 may serve as an important regulator in stroke-induced angiogenesis. The protective mechanism of phosphorylated HDAC4 is associated with HIF-VEGF signaling, implicating a novel therapeutic target in stroke.


Assuntos
Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Histona Desacetilases/metabolismo , Neovascularização Fisiológica , Acidente Vascular Cerebral/metabolismo , Animais , Carbazóis/farmacologia , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/química , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Mol Neurobiol ; 54(10): 8179-8190, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27900677

RESUMO

Angiogenesis after ischemic brain injury contributes to the restoration of blood supply in the ischemic zone. Strategies to improve angiogenesis may facilitate the function recovery after stroke. Recent researches have demonstrated that dysfunction of long non-coding RNAs are associated with angiogenesis. We have previously reported that long non-coding RNAs (lncRNAs) are aberrantly expressed in ischemic stroke. However, little is known about long non-coding RNAs and theirs role in angiogenesis after stroke. In this study, we identified a rat lncRNAs, Meg3, and found that Meg3 was significantly decreased after ischemic stroke. Overexpression of Meg3 suppressed functional recovery and decreased capillary density after ischemic stroke. Downregulation of Meg3 ameliorated brain lesion and increased angiogenesis after ischemic stroke. Silencing of Meg3 resulted in a proangiogenic effect evidenced by increased endothelial cell migration, proliferation, sprouting, and tube formation. Mechanistically, we showed that Meg3 negatively regulated notch pathway both in vivo and in vitro. Inhibition of notch signaling in endothelial cells reversed the proangiogenic effect induced by Meg3 downregulation. This study revealed the function of Meg3 in ischemic stroke and elucidated its mechanism in angiogenesis after ischemic stroke.


Assuntos
Isquemia Encefálica/metabolismo , Regulação para Baixo/fisiologia , Neovascularização Patológica/metabolismo , RNA Longo não Codificante/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Animais , Isquemia Encefálica/patologia , Masculino , Neovascularização Patológica/patologia , Ratos , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
8.
Sci Rep ; 6: 20850, 2016 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-26865164

RESUMO

Ischemic injuries will lead to necrotic tissue damage, and post-ischemia angiogenesis plays critical roles in blood flow restoration and tissue recovery. Recently, several types of small RNAs have been reported to be involved in this process. In this study, we first generated a rat brain ischemic model to investigate the involvement of new types of small RNAs in ischemia. We utilized deep sequencing and bioinformatics analyses to demonstrate that the level of small RNA fragments derived from tRNAs strikingly increased in the ischemic rat brain. Among these sequences, tRNA(Val)- and tRNA(Gly)-derived small RNAs account for the most abundant segments. The up-regulation of tRNA(Val)- and tRNA(Gly)-derived fragments was verified through northern blot and quantitative PCR analyses. The levels of these two fragments also increased in a mouse hindlimb ischemia model and cellular hypoxia model. Importantly, up-regulation of the tRNA(Val)- and tRNA(Gly)-derived fragments in endothelial cells inhibited cell proliferation, migration and tube formation. Furthermore, we showed that these small RNAs are generated by angiogenin cleavage. Our results indicate that tRNA-derived fragments are involved in tissue ischemia, and we demonstrate for the first time that tRNA(Val)- and tRNA(Gly)-derived fragments inhibit angiogenesis by modulating the function of endothelial cells.


Assuntos
Isquemia Encefálica/genética , Neovascularização Fisiológica/genética , Pequeno RNA não Traduzido/genética , RNA de Transferência de Glicina/genética , RNA de Transferência de Valina/genética , Animais , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Hipóxia Celular , Movimento Celular , Proliferação de Células , Biologia Computacional , Regulação da Expressão Gênica , Membro Posterior/irrigação sanguínea , Membro Posterior/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Proteólise , Clivagem do RNA , Pequeno RNA não Traduzido/metabolismo , RNA de Transferência de Glicina/metabolismo , RNA de Transferência de Valina/metabolismo , Ratos , Ratos Sprague-Dawley , Ribonuclease Pancreático/genética , Ribonuclease Pancreático/metabolismo , Transdução de Sinais
9.
Stem Cell Res Ther ; 6: 10, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-26268554

RESUMO

INTRODUCTION: 'Patient-specific' induced pluripotent stem cells (iPSCs) are attractive because they can generate abundant cells without the risk of immune rejection for cell therapy. Studies have shown that iPSC-derived mesenchymal stem cells (iMSCs) possess powerful proliferation, differentiation, and therapeutic effects. Recently, most studies indicate that stem cells exert their therapeutic effect mainly through a paracrine mechanism other than transdifferentiation, and exosomes have emerged as an important paracrine factor for stem cells to reprogram injured cells. The objective of this study was to evaluate whether exosomes derived from iMSCs (iMSCs-Exo) possess the ability to attenuate limb ischemia and promote angiogenesis after transplantation into limbs of mice with femoral artery excision. METHODS: Human iPSCs (iPS-S-01, C1P33, and PCKDSF001C1) were used to differentiate into iMSCs in a modified one-step method. iMSCs were characterized by flow cytometry and multipotent differentiation potential analysis. Ultrafiltration combined with a purification method was used to isolate iMSCs-Exo, and transmission electron microscopy and Western blotting were used to identify iMSCs-Exo. After establishment of mouse hind-limb ischemia with excision of femoral artery and iMSCs-Exo injection, blood perfusion was monitored at days 0, 7, 14, and 21; microvessel density in ischemic muscle was also analyzed. In vitro migration, proliferation, and tube formation experiments were used to analyze the ability of pro-angiogenesis in iMSCs-Exo, and quantitative reverse-transcriptase polymerase chain reaction and enzyme-linked immunosorbent assay were used to identify expression levels of angiogenesis-related molecules in human umbilical vein endothelial cells (HUVECs) after being cultured with iMSCs-Exo. RESULTS: iPSCs were efficiently induced into iMSC- with MSC-positive and -negative surface antigens and osteogenesis, adipogenesis, and chondrogenesis differentiation potential. iMSCs-Exo with a diameter of 57 ± 11 nm and expressed CD63, CD81, and CD9. Intramuscular injection of iMSCs-Exo markedly enhanced microvessel density and blood perfusion in mouse ischemic limbs, consistent with an attenuation of ischemic injury. In addition, iMSCs-Exo could activate angiogenesis-related molecule expression and promote HUVEC migration, proliferation, and tube formation. CONCLUSION: Implanted iMSCs-Exo was able to protect limbs from ischemic injury via the promotion of angiogenesis, which indicated that iMSCs-Exo may be a novel therapeutic approach in the treatment of ischemic diseases.


Assuntos
Exossomos/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Isquemia/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Animais , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Camundongos
10.
Mol Cell Biochem ; 395(1-2): 291-8, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24972705

RESUMO

Neural stem cells (NSCs) derived from induced pluripotent stem cells (iPSCs) are becoming an appealing source of cell-based therapies of brain diseases. As such, it is important to understand the molecular mechanisms that regulate the differentiation of iPSCs toward NSCs. It is well known that Notch signaling governs the retention of stem cell features and drives stem cells fate. However, further studies are required to investigate the role of Notch signaling in the NSCs differentiation of iPSCs. In this study, we successfully generated NSCs from human iPSCs using serum-free medium supplemented with retinoic acid (RA) in vitro. We then assessed changes in the expression of Notch signaling-related molecules and some miRNAs (9, 34a, 200b), which exert their regulation by targeting Notch signaling. Moreover, we used a γ-secretase inhibitor (DAPT) to disturb Notch signaling. Data revealed that the levels of the Notch signaling-related molecules decreased, whereas those miRNAs increased, during this differentiation process. Inhibition of Notch signaling accelerated the formation of the neural rosette structures and the expression of NSC and mature neurocyte marker genes. This suggests that Notch signaling negatively regulated the neuralization of human iPSCs, and that this process may be regulated by some miRNAs.


Assuntos
Dipeptídeos/farmacologia , Células-Tronco Pluripotentes Induzidas/fisiologia , Células-Tronco Neurais/citologia , Receptores Notch/metabolismo , Transdução de Sinais , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , MicroRNAs/metabolismo , Transdução de Sinais/efeitos dos fármacos
11.
Tissue Eng Part A ; 20(13-14): 1794-806, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24387670

RESUMO

Stem cells in human urine have gained attention in recent years; however, urine-derived stem cells (USCs) are far from being well elucidated. In this study, we compared the biological characteristics of USCs with adipose-derived stem cells (ASCs) and investigated whether USCs could serve as a potential cell source for neural tissue engineering. USCs were isolated from voided urine with a modified culture medium. Through a series of experiments, we examined the growth rate, surface antigens, and differentiation potential of USCs, and compared them with ASCs. USCs showed robust proliferation ability. After serial propagation, USCs retained normal karyotypes. Cell surface antigen expression of USCs was similar to ASCs. With lineage-specific induction factors, USCs could differentiate toward the osteogenic, chondrogenic, adipogenic, and neurogenic lineages. To assess the ability of USCs to survive, differentiate, and migrate, they were seeded onto hydrogel scaffold and transplanted into rat brain. The results showed that USCs were able to survive in the lesion site, migrate to other areas, and express proteins that were associated with neural phenotypes. The results of our study demonstrate that USCs possess similar biological characteristics with ASCs and have multilineage differentiation potential. Moreover USCs can differentiate to neuron-like cells in rat brain. The present study shows that USCs are a promising cell source for tissue engineering and regenerative medicine.


Assuntos
Neurologia , Transplante de Células-Tronco , Células-Tronco/citologia , Urina/citologia , Tecido Adiposo/citologia , Adulto , Animais , Biomarcadores/metabolismo , Encéfalo/citologia , Diferenciação Celular , Linhagem da Célula , Membrana Celular/metabolismo , Movimento Celular , Proliferação de Células , Forma Celular , Sobrevivência Celular , Proteínas de Fluorescência Verde/metabolismo , Humanos , Cariotipagem , Masculino , Neurônios/citologia , Neurônios/metabolismo , Ratos , Células-Tronco/metabolismo , Adulto Jovem
12.
Neurol Sci ; 35(4): 531-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24057116

RESUMO

The objective of the study was to investigate the role of neuregulin-ErbB signaling in neuropathic pain in different types of injury. Neuregulin-1(NRG-1) was injected into animals with either formalin-induced pain model or spared nerve injury (SNI) model. Formalin tests or paw withdrawal tests were performed to study the role of NRG-1 in neuropathic pain. siRNA specific to different erbB receptors were then introduced to test which specific signaling pathway was required for NRG-1 signaling in the different pain models. NRG-1 inhibits neuropathic pain after SNI in a dose-dependent manner, while NRG-1 aggravates formalin-induced neuropathic pain. ErbB2 and erbB4 receptors were activated after neuregulin administration. Knockdown of ErbB2 relieves the aggravation of NRG-1 on formalin-induced neuropathic pain, and knockdown of ErbB4 could relieve the inhibition of NRG-1 on neuropathic pain in the SNI model. NRG-1 has two distinct functions depending on the different receptor activation in different models of neuropathic pain. These novel findings may provide new therapeutic approaches for the treatment of neuropathic pain in different injury types.


Assuntos
Neuralgia/metabolismo , Neuregulina-1/fisiologia , Receptor ErbB-2/metabolismo , Animais , Formaldeído , Hiperalgesia/complicações , Hiperalgesia/metabolismo , Masculino , Neuralgia/induzido quimicamente , Neuralgia/complicações , Neuregulina-1/farmacologia , Ratos
13.
Stem Cell Res Ther ; 4(3): 73, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23769173

RESUMO

INTRODUCTION: Stroke is a major cause of permanent neurologic damage, with few effective treatments available to restore lost function. Induced pluripotent stem cells (iPSCs) have the potential to generate all cell types in vitro and can be generated from a stroke patient. Therefore, iPSCs are attractive donor sources of genetically identical "patient-specific" cells to hold promise in therapy for stroke. In the present study, we established a four-stage culture system by using serum-free medium and retinoic acid (RA) to differentiate iPSCs into neural stem cells (NSCs) effectively and stably. Our hypothesis was that iPSC-derived NSCs would survive, migrate, and differentiate in vivo, and improve neurologic function after transplantation into the brains of rats with ischemic stroke. METHODS: Human iPSCs (iPS-S-01) and human ESCs (HuES17) were used to differentiate into NSCs by using our four-stage culture system. iPSCs and differentiated NSCs were characterized by immunocytochemistry staining and reverse transcription-polymerase chain reaction (RT-PCR) analysis. After establishment of focal cerebral ischemia with occlusion of the middle cerebral artery (MCA) and cell transplantation, animals were killed at 1 week and 2 weeks to analyze survival, migration, and differentiation of implanted cells in brain tissue. Animal behavior was evaluated via rope grabbing, beam walking, and Morris water maze tests. RESULTS: iPSCs were efficiently induced into NSCs by using a newly established four-stage induction system in vitro. iPSCs expressed pluripotency-associated genes Oct4, Sox2, and Nanog before NSC differentiation. The iPSC-derived NSCs spontaneously differentiated into neurons and astrocytes, which highly express ß-tubulin and glial fibrillary acidic protein (GFAP), respectively. On transplantation into the striatum, CM-DiI labeled iPSC-derived NSCs were found to migrate into the ischemia area at 1 week and 2 weeks, and animal-function recovery was significantly improved in comparison with control groups at 3 weeks. CONCLUSIONS: The four-stage induction system is stable and effective to culture, differentiate, and induce iPSCs to NSCs by using serum-free medium combined with retinoic acid (RA). Implanted iPSC-derived NSCs were able to survive, migrate into the ischemic brain area to differentiate into mature neural cells, and seem to have potential to restore lost neurologic function from damage due to stroke in a rat model.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Infarto da Artéria Cerebral Média/terapia , Células-Tronco Neurais/transplante , Neurônios/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Células-Tronco Embrionárias/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/patologia , Masculino , Atividade Motora , Proteína Homeobox Nanog , Nestina/genética , Nestina/metabolismo , Células-Tronco Neurais/citologia , Neurônios/patologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Tubulina (Proteína)/metabolismo
14.
Mol Cell Biochem ; 370(1-2): 45-51, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22833359

RESUMO

The compensatory angiogenesis that occurs after cerebral ischemia increases blood flow to the injured area and limits extension of the ischemic penumbra. In this way, it improves the local blood supply. Fostering compensatory angiogenesis is an effective treatment for ischemic cerebrovascular disease. However, angiogenesis in the adult organism is a complex, multi-step process, and the mechanisms underlying the regulation of angiogenesis are not well understood. Although Notch signaling reportedly regulates the vascularization process that occurs in ischemic tissues, little is known about the role of Notch signaling in the regulation of ischemia-induced angiogenesis after ischemic stroke. Recent research has indicated that miR-210, a hypoxia-induced microRNA, plays a crucial role in regulating the biological processes that occur in blood vessel endothelial cells under hypoxic conditions. This study was undertaken to investigate the role of miR-210 in regulating angiogenesis in response to brain ischemia injury and the role of the Notch pathway in the body's response. We found miR-210 to be significantly up-regulated in adult rat ischemic brain cortexes in which the expression of Notch1 signaling molecules was also increased. Hypoxic models of human umbilical vein endothelial cells (HUVE-12) were used to assess changes in miR-210 and Notch1 expression in endothelial cells. Results were consistent with in vivo findings. To determine the molecular mechanisms behind these phenomena, we transfected HUVE-12 cells with miR-210 recombinant lentiviral vectors. We found that miR-210 overexpression caused up-regulation of Notch1 signaling molecules and induced endothelial cells to migrate and form capillary-like structures on Matrigel. These data suggest that miR-210 is involved in the regulation of angiogenesis in response to ischemic injury to the brain. Up-regulation of miR-210 can activate the Notch signaling pathway, which may contribute to angiogenesis after cerebral ischemia.


Assuntos
Isquemia Encefálica/complicações , MicroRNAs/metabolismo , Neovascularização Patológica/etiologia , Neovascularização Patológica/genética , Receptor Notch1/metabolismo , Transdução de Sinais , Animais , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Hipóxia Celular/genética , Modelos Animais de Doenças , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Masculino , MicroRNAs/genética , Microvasos/patologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética
15.
Kidney Blood Press Res ; 35(3): 182-91, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22123256

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are endogenous, non-coding, small RNAs that regulate gene expression and function, but little is known about regulation of miRNAs in the kidneys under normal or pathologic conditions. Here, we sought to investigate the potential involvement of miRNAs in renal ischemia/reperfusion (I/R) injury and angiogenesis and to define some of the miRNAs possibly associated with renal angiogenesis. METHODS AND RESULTS: Male Balb/c mice were subjected to a standard renal I/R. CD31 immunostaining indicated a significant increase of microvessels in the ischemic region. VEGF and VEGFR2 expression were increased in renal I/R at both the mRNA and protein levels which were detected by qRT-PCR and Western blot, respectively. More importantly, 76 microRNAs exhibited more than 2-fold changes using Agilent microRNA microarray, which contains downregulation of 40 miRNAs and upregulation of 36 miRNAs. Upregulation of miR-210 was confirmed by qRT-PCR with prominent changes at 4 and 24 h after reperfusion. Furthermore, overexpression of miR-210 in HUVEC-12 cells enhances VEGF and VEGFR2 expression and promotes angiogenesis on Matrigel in vitro. CONCLUSION: These findings suggest miR-210 may be involved in targeting the VEGF signaling pathway to regulate angiogenesis after renal I/R injury, which provides novel insights into the angiogenesis mechanism of renal I/R injury.


Assuntos
MicroRNAs/biossíntese , Neovascularização Patológica/metabolismo , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neovascularização Patológica/fisiopatologia , Distribuição Aleatória , Traumatismo por Reperfusão/fisiopatologia , Fator A de Crescimento do Endotélio Vascular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA