Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-38687468

RESUMO

High levels of oxidant stress in the form of reactive oxidant species (ROS) are prevalent in the circulation and tissues in various types of cardiovascular disease including heart failure, hypertension, peripheral arterial disease, and stroke. Here we review the role of nuclear factor erythroid 2-related factor 2 (Nrf2), an important and widespread antioxidant and anti-inflammatory transcription factor that may contribute to the pathogenesis and maintenance of cardiovascular diseases. We review studies showing that downregulation of Nrf2 exacerbates heart failure, hypertension and autonomic function. Finally, we discuss the potential for using Nrf2 modulation as a therapeutic strategy for cardiovascular diseases and autonomic dysfunction.

2.
Int J Mol Sci ; 24(20)2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37894887

RESUMO

Extracellular vesicles (EVs) are small, membrane-bound vesicles used by cells to deliver biological cargo such as proteins, mRNA, and other biomolecules from one cell to another, thus inducing a specific response in the target cell and are a powerful method of cell to cell and organ to organ communication, especially during the pathogenesis of human disease. Thus, EVs may be utilized as prognostic and diagnostic biomarkers, but they also hold therapeutic potential just as mesenchymal stem cells have been used in therapeutics. However, unmodified EVs exhibit poor targeting efficacy, leading to the necessity of engineered EVS. To highlight the advantages and therapeutic promises of engineered EVs, in this review, we summarized the research progress on engineered EVs in the past ten years, especially in the past five years, and highlighted their potential applications in therapeutic development for human diseases. Compared to the existing stem cell-derived EV-based therapeutic strategies, engineered EVs show greater promise in clinical applications: First, engineered EVs mediate good targeting efficacy by exhibiting a targeting peptide that allows them to specifically target a specific organ or even cell type, thus avoiding accumulation in undesired locations and increasing the potency of the treatment. Second, engineered EVs can be artificially pre-loaded with any necessary biomolecular cargo or even therapeutic drugs to treat a variety of human diseases such as cancers, neurological diseases, and cardiovascular ailments. Further research is necessary to improve logistical challenges in large-scale engineered EV manufacturing, but current developments in engineered EVs prove promising to greatly improve therapeutic treatment for traditionally difficult to treat diseases.


Assuntos
Vesículas Extracelulares , Neoplasias , Humanos , Vesículas Extracelulares/metabolismo , Comunicação Celular , Neoplasias/metabolismo , Peptídeos/metabolismo , Proteínas/metabolismo
3.
Cells ; 12(17)2023 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-37681877

RESUMO

Extracellular vesicles (EVs) are emerging mediators of intracellular and inter-organ communications in cardiovascular diseases (CVDs), especially in the pathogenesis of heart failure through the transference of EV-containing bioactive substances. microRNAs (miRNAs) are contained in EV cargo and are involved in the progression of heart failure. Over the past several years, a growing body of evidence has suggested that the biogenesis of miRNAs and EVs is tightly regulated, and the sorting of miRNAs into EVs is highly selective and tightly controlled. Extracellular miRNAs, particularly circulating EV-miRNAs, have shown promising potential as prognostic and diagnostic biomarkers for heart failure and as therapeutic targets. In this review, we summarize the latest progress concerning the role of EV-miRNAs in HF and their application in a therapeutic strategy development for heart failure.


Assuntos
Doenças Cardiovasculares , Vesículas Extracelulares , Insuficiência Cardíaca , MicroRNAs , Humanos , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/terapia , Movimento Celular , Vesículas Extracelulares/genética , MicroRNAs/genética
4.
Circ Res ; 131(8): 687-700, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36098045

RESUMO

BACKGROUND: Chronic heart failure (CHF) is associated with redox imbalance. Downregulation of Nrf2 (nuclear factor [erythroid-derived 2]-like 2) plays important roles in disrupting myocardial redox homeostasis and mediating sympathetic nerve activity in the setting of CHF. However, it is unclear if circulating extracellular vesicles (EVs) elicit sympathetic excitation in CHF by disrupting central redox homeostasis. We tested the hypothesis that cardiac-derived EVs circulate to the presympathetic rostral ventrolateral medulla and contribute to oxidative stress and sympathetic excitation via EV-enriched microRNA-mediated Nrf2 downregulation. METHODS: Data were collected on rats with CHF post-myocardial infarction (MI) and on human subjects with ischemic CHF. EVs were isolated from tissue and plasma, and we determined the miRNAs cargo that related to targeting Nrf2 translation. We tracked the distribution of cardiac-derived EVs using in vitro labeled circulating EVs and cardiac-specific membrane GFP+ transgenic mice. Finally, we tested the impact of exogenously loading of antagomirs to specific Nrf2-related miRNAs on CHF-EV-induced pathophysiological phenotypes in normal rats (eg, sympathetic and cardiac function). RESULTS: Nrf2 downregulation in CHF rats was associated with an upregulation of Nrf2-targeting miRNAs, which were abundant in cardiac-derived and circulating EVs from rats and humans. EVs isolated from the brain of CHF rats were also enriched with Nrf2-targeting miRNAs and cardiac-specific miRNAs. Cardiac-derived EVs were taken up by neurons in the rostral ventrolateral medulla. The administration of cardiac-derived and circulating EVs from CHF rats into the rostral ventrolateral medulla of normal rats evoked an increase in renal sympathetic nerve activity and plasma norepinephrine compared with Sham-operated rats, which were attenuated by exogenously preloading CHF-EVs with antagomirs to Nrf2-targeting miRNAs. CONCLUSIONS: Cardiac microRNA-enriched EVs from animals with CHF can mediate crosstalk between the heart and the brain in the regulation of sympathetic outflow by targeting the Nrf2/antioxidant signaling pathway. This new endocrine signaling pathway regulating sympathetic outflow in CHF may be exploited for novel therapeutics.


Assuntos
Vesículas Extracelulares , Insuficiência Cardíaca , MicroRNAs , Animais , Antagomirs/metabolismo , Antioxidantes/metabolismo , Vesículas Extracelulares/metabolismo , Insuficiência Cardíaca/metabolismo , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Norepinefrina/metabolismo , Ratos , Sistema Nervoso Simpático
5.
Natl Sci Rev ; 8(1): nwaa125, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34691552

RESUMO

Human cerebral cortex displays various dynamics patterns under different states, however the mechanism how such diverse patterns can be supported by the underlying brain network is still not well understood. Human brain has a unique network structure with different regions of interesting to perform cognitive tasks. Using coupled neural mass oscillators on human cortical network and paying attention to both global and local regions, we observe a new feature of chimera states with multiple spatial scales and a positive correlation between the synchronization preference of local region and the degree of symmetry of the connectivity of the region in the network. Further, we use the concept of effective symmetry in the network to build structural and dynamical hierarchical trees and find close matching between them. These results help to explain the multiple brain rhythms observed in experiments and suggest a generic principle for complex brain network as a structure substrate to support diverse functional patterns.

6.
Exerc Sport Sci Rev ; 49(3): 213-222, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33927165

RESUMO

This review explores the hypothesis that the repetitive contraction-relaxation that occurs during chronic exercise activates skeletal myocyte nuclear factor erythroid-derived 2-like 2 (Nrf2) to upregulate antioxidant enzymes. These proteins are secreted into the circulation within extracellular vesicles and taken up by remote cells, thus providing remote organs with cytoprotection against subsequent oxidative stress.


Assuntos
Antioxidantes , Fator 2 Relacionado a NF-E2 , Antioxidantes/metabolismo , Comunicação , Músculo Esquelético/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo
7.
Free Radic Biol Med ; 167: 218-231, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33741451

RESUMO

The balance between pro- and antioxidant molecules has been established as an important driving force in the pathogenesis of cardiovascular disease. Chronic heart failure is associated with oxidative stress in the myocardium and globally. Redox balance in the heart and brain is controlled, in part, by antioxidant proteins regulated by the transcription factor Nuclear factor erythroid 2-related factor 2 (Nrf2), which is reduced in the heart failure state. Nrf2 can, in turn, be regulated by a variety of mechanisms including circulating microRNAs (miRNAs) encapsulated in extracellular vesicles (EVs) derived from multiple cell types in the heart. Here, we review the role of the Nrf2 and antioxidant enzyme signaling pathway in mediating redox balance in the myocardium and the brain in the heart failure state. This review focuses on Nrf2 and antioxidant protein regulation in the heart and brain by miRNA-enriched EVs in the setting of heart failure. We discuss EV-mediated intra- and inter-organ communications especially, communication between the heart and brain via an EV pathway that mediates cardiac function and sympatho-excitation in heart failure. Importantly, we speculate how engineered EVs with specific miRNAs or antagomirs may be used in a therapeutic manner in heart failure.


Assuntos
Vesículas Extracelulares , Insuficiência Cardíaca , MicroRNAs , Vesículas Extracelulares/metabolismo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Miocárdio/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Transdução de Sinais
8.
Front Cell Dev Biol ; 8: 573, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850781

RESUMO

MicroRNA-124 (miR-124), a brain-enriched microRNA, is known to regulate microglial quiescence. Psychostimulants such as cocaine have been shown to activate microglia by downregulating miR-124, leading, in turn, to neuroinflammation. We thus rationalized that restoring the levels of miR-124 could function as a potential therapeutic approach for cocaine-mediated neuroinflammation. Delivering miRNA based drugs in the brain that are effective and less invasive, however, remains a major challenge in the field. Herein we engineered extracellular vesicles (EVs) and loaded them with miR-124 for delivery in the brain. Approach involved co-transfection of mouse dendritic cells with Dicer siRNA and RVG-Lamp2b plasmid to deplete endogenous miRNAs and for targeting the CNS, respectively. Mouse primary microglia (mPm) were treated with purified engineered EVs loaded with either Cy5-miR-124 or Cy5-scrambled miRNA oligos in the presence or absence of cocaine followed by assessing EV uptake and microglial activation. In vivo studies involved pretreating mice intranasally with engineered EVs followed by cocaine injection (20 mg/kg, i.p.). mPm exposed to EV-miR-124 exhibited reduced expression of miR-124 targets - TLR4 and STAT3 as well as ERK-1/2 and Iba1. In cocaine administered mice, EV-Cy5-miR-124 delivered intranasally were detected in the CNS and significantly reduced the expression of inflammatory markers TLR4, MYD88, STAT3 and NF-kB p65 while also downregulating the microglial activation marker, Iba1. Collectively, these findings suggest that engineered EVs can deliver miR-124 into the CNS, thereby alleviating cocaine-mediated microglial activation. Manipulating EV miRNAs can thus be envisioned as an efficient means for delivery of RNA-based therapeutics to target organs.

9.
J Mol Cell Cardiol ; 143: 120-131, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32370947

RESUMO

Under stress, the heart undergoes extensive remodeling resulting in cardiac fibrosis and hypertrophy, ultimately contributing to chronic heart failure (CHF). Alterations in microRNA levels are associated with dysfunctional gene expression profiles involved in the pathogenesis of heart failure. We previously showed that myocardial infarction-induced microRNA-enriched extracellular vesicles (EVs) contribute to the reduction in antioxidant enzymes by targeting Nrf2 signaling in CHF. MicroRNA-27a (miRNA-27a) is the predominant microRNA contained in cardiac fibroblast-derived EVs contributing to oxidative stress along with hypertrophic gene expression in cardiomyocytes. In the present study, we observed that miRNA-27a passenger strand (miRNA-27a*) was markedly upregulated in the non-infarcted area of the left ventricle of rats with CHF and encapsulated into EVs and secreted into the circulation. Bioinformatic analysis revealed that PDZ and LIM domain 5 (PDLIM5) is one of the major targets of miRNA-27a*, playing a major role in cardiac structure and function, and potentially contributing to the progression of cardiac hypertrophy. Our in vivo data demonstrate that PDLIM5 is down-regulated in the progression of heart failure, accompanied with the upregulation of hypertrophic genes and consistent with alterations in miRNA-27a*. Moreover, exogenous administration of miRNA27a* mimics inhibit PDLIM5 translation in cardiomyocytes whereas a miRNA27a* inhibitor enhanced PDLIM5 expression. Importantly, we confirmed that infarcted hearts have higher abundance of miRNA-27a* in EVs compared to normal hearts and further demonstrated that cultured cardiac fibroblasts secrete miRNA27a*-enriched EVs into the extracellular space in response to Angiotensin II stimulation, which inhibited PDLIM5 translation, leading to cardiomyocyte hypertrophic gene expression. In vivo studies suggest that the administration of a miRNA-27a* inhibitor in CHF rats partially blocks endogenous miR-27a* expression, prevents hypertrophic gene expression and improves myocardial contractility. These findings suggest that cardiac fibroblast-secretion of miRNA27a*-enriched EVs may act as a paracrine signaling mediator of cardiac hypertrophy that has potential as a novel therapeutic target.


Assuntos
Cardiomegalia/etiologia , Cardiomegalia/metabolismo , Vesículas Extracelulares/metabolismo , MicroRNAs/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Angiotensina II/metabolismo , Animais , Transporte Biológico , Biomarcadores , Cardiomegalia/fisiopatologia , Células Cultivadas , Técnicas de Cocultura , Suscetibilidade a Doenças , Fibroblastos/metabolismo , Expressão Gênica , Regulação da Expressão Gênica , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Organogênese , Ratos
10.
J Neuroimmune Pharmacol ; 15(3): 390-399, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31325121

RESUMO

Long non-coding RNAs (lncRNAs), including long intergenic non-coding RNAs (lincRNAs), play an important regulatory role in controlling various biological processes. Both in vitro and in vivo studies have demonstrated that lincRNA-Cox2 plays a global regulatory role in regulating the expression of immune genes. Extracellular vesicles (EVs) are cell-derived nanosized membrane vesicles that have gained increasing attention in recent years due to their ability to efficiently deliver therapeutics to specific target organs or cell types. In this study, we found that lincRNA-Cox2 controls the expression of a set of cell cycle genes in lipopolysaccharide (LPS)-stimulated microglial cells. Our in vitro study suggested that knocking down lincRNA-Cox2 reversed LPS-induced microglial proliferation. In addition, our in vivo study demonstrated that intranasally delivered lincRNA-Cox2-siRNA loaded EVs could reach the brain resulting in a significant decrease in the expression of lincRNA-Cox2 in the microglia. Importantly, lincRNA-Cox2-siRNA loaded EVs also decreased LPS-induced microglial proliferation in mice. These findings indicate that intranasal delivery of EV-loaded small RNA could be developed as therapeutics for treatment of a multitude of CNS disorders.


Assuntos
Ciclo-Oxigenase 2/genética , Vesículas Extracelulares , Lipopolissacarídeos/farmacologia , Microglia/efeitos dos fármacos , RNA Longo não Codificante/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Administração Intranasal , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Genes cdc/genética , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas
11.
Sci Rep ; 9(1): 14389, 2019 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-31591418

RESUMO

Based on the data of cerebral cortex, we present a two-layered brain network model of coupled neurons where the two layers represent the left and right hemispheres of cerebral cortex, respectively, and the links between the two layers represent the inter-couplings through the corpus callosum. By this model we show that abundant patterns of synchronization can be observed, especially the chimera state, depending on the parameters of system such as the coupling strengths and coupling phase. Further, we extend the model to a more general two-layered network to better understand the mechanism of the observed patterns, where each hemisphere of cerebral cortex is replaced by a highly clustered subnetwork. We find that the number of inter-couplings is another key parameter for the emergence of chimera states. Thus, the chimera states come from a matching between the structure parameters such as the number of inter-couplings and clustering coefficient etc and the dynamics parameters such as the intra-, inter-coupling strengths and coupling phase etc. A brief theoretical analysis is provided to explain the borderline of synchronization. These findings may provide helpful clues to understand the mechanism of brain functions.


Assuntos
Encéfalo/fisiologia , Modelos Neurológicos , Rede Nervosa/fisiologia , Encéfalo/citologia , Córtex Cerebral/citologia , Córtex Cerebral/fisiologia , Humanos , Rede Nervosa/citologia
12.
J Pharmacol Exp Ther ; 371(3): 642-651, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31601682

RESUMO

Oxidative stress plays an important role in the pathogenesis of chronic heart failure (CHF) in many tissues. Increasing evidence suggests that systemic activation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signaling can protect against postinfarct cardiac remodeling by reducing oxidative stress. However, it remains to be elucidated if Nrf2 activation exerts therapeutic effects in the CHF state. Here, we investigated the beneficial hemodynamic effects of bardoxolone methyl (2-Cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid methyl ester, CDDO-Me), a pharmacological activator of Nrf2, in a rodent model of CHF. Based on echocardiographic analysis, rats at 12 weeks post-myocardial infarction (MI) were randomly split into four groups. CDDO-Me (5 mg/kg, i.p.) was administered daily for another 2 weeks in sham and CHF rats and compared with vehicle treatment. Echocardiographic and hemodynamic analysis suggest that short-term CDDO-Me administration increased stroke volume and cardiac output in CHF rats and decreased left ventricle end-diastolic pressure. Molecular studies revealed that CDDO-Me-induced cardiac functional improvement was attributed to an increase of both Nrf2 transcription and translation, and a decrease of oxidative stress in the noninfarcted areas of the heart. Furthermore, CDDO-Me reduced NF-κB binding and increased Nrf2 binding to the CREB-binding protein, which may contribute to the selective increase of Nrf2 downstream targets, including NADPH Oxidase Quinone 1, Heme Oxygenase 1, Catalase, and Glutamate-Cysteine Ligase Catalytic Subunit, and the attenuation of myocardial inflammation in CHF rats. Our findings suggest that Nrf2 activation may provide beneficial cardiac effects in MI-mediated CHF. SIGNIFICANCE STATEMENT: Chronic heart failure (CHF) is the leading cause of death among the aged worldwide. The imbalance between pro- and antioxidant pathways is a determinant in the pathogenesis of CHF. Systemic activation of Nrf2 and antioxidant protein signaling by bardoxolone methyl may have beneficial effects on cardiac function and result in improvements by enhancing antioxidant enzyme expression and attenuating myocardial inflammation.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Fator 2 Relacionado a NF-E2/fisiologia , Ácido Oleanólico/análogos & derivados , Animais , Proteína de Ligação a CREB/metabolismo , Doença Crônica , Insuficiência Cardíaca/fisiopatologia , Hemodinâmica/efeitos dos fármacos , Masculino , NF-kappa B/metabolismo , Ácido Oleanólico/farmacologia , Ácido Oleanólico/uso terapêutico , Ratos , Ratos Sprague-Dawley , Função Ventricular Esquerda/efeitos dos fármacos
13.
Neurosci Lett ; 699: 16-23, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-30664902

RESUMO

Transplantation of dopaminergic precursors (DPs) is a promising therapeutic strategy of Parkinson's disease (PD). However, limited cell source for dopaminergic precursors has become a major obstacle for transplantation therapy. Our group demonstrated previously that mouse fibroblasts can be reprogrammed into induced dopaminergic precursors (iDPs) with high differentiation efficiency. In the current study, we hypothesized that a similar strategy can be applied to generate human iDPs for future cell therapy of PD. We overexpressed transcription factors Brn2, Sox2, and Foxa2 in human fibroblasts and observed formation of neurospheres. Subsequent characterization of the precursor colonies confirmed the generation of human induced dopaminergic precursors (hiDPs). These hiDPs were capable of self-renewal, proliferation, and differentiation. The hiDPs demonstrated high immunoreactivity for neural progenitor markers and high levels of gene expression for ventral mesencephalon-related neural progenitor markers such as Lmx1a, NIKX6.1, Corin, Otx2 and Mash1. Furthermore, the hiDPs could be differentiated into dopaminergic neurons with ˜80% efficiency, which significantly increased major functionally relevant proteins such as TH, DAT, AADC, Lmx1B, and VMAT2 compared to hiDPs. Additionally, hiDPs are more dopaminergic progenitor-restricted compare to those hiDP-like cells reprogrammed only by Brn2 and Sox2. Together, these results suggest that hiDPs with high differentiation efficiency can be generated by direct lineage reprogramming of fibroblasts with transcription factors Brn2, Sox2, and Foxa2. These hiDPs may serve as a safe and effective cell source for transplantation treatment of PD.


Assuntos
Diferenciação Celular , Linhagem da Célula/fisiologia , Neurônios Dopaminérgicos/citologia , Fibroblastos/citologia , Biomarcadores/metabolismo , Proliferação de Células/fisiologia , Autorrenovação Celular/fisiologia , Transplante de Células , Células Cultivadas , Vetores Genéticos , Fator 3-beta Nuclear de Hepatócito/biossíntese , Proteínas de Homeodomínio/biossíntese , Humanos , Fatores do Domínio POU/biossíntese , Retroviridae , Fatores de Transcrição SOXB1/biossíntese , Transfecção
14.
Neurobiol Dis ; 124: 322-334, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30528256

RESUMO

Neural stem/progenitor cells (NPCs) are known to have potent therapeutic effects in neurological disorders through the secretion of extracellular vesicles (EVs). Despite the therapeutic potentials, the numbers of NPCs are limited in the brain, curbing the further use of EVs in the disease treatment. To overcome the limitation of NPC numbers, we used a three transcription factor (Brn2, Sox2, and Foxg1) somatic reprogramming approach to generate induced NPCs (iNPCs) from mouse fibroblasts and astrocytes. The resulting iNPCs released significantly higher numbers of EVs compared with wild-type NPCs (WT-NPCs). Furthermore, iNPCs-derived EVs (iNPC-EVs) promoted NPC function by increasing the proliferative potentials of WT-NPCs. Characterizations of EV contents through proteomics analysis revealed that iNPC-EVs contained higher levels of growth factor-associated proteins that were predicted to activate the down-stream extracellular signal-regulated kinase (ERK) pathways. As expected, the proliferative effects of iNPC-derived EVs on WT-NPCs can be blocked by an ERK pathway inhibitor. Our data suggest potent therapeutic effects of iNPC-derived EVs through the promotion of NPC proliferation, release of growth factors, and activation of ERK pathways. These studies will help develop highly efficient cell-free therapeutic strategies for the treatment of neurological diseases.


Assuntos
Proliferação de Células/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Vesículas Extracelulares/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Técnicas de Reprogramação Celular/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Transdução de Sinais/fisiologia
15.
Mol Ther Nucleic Acids ; 13: 450-463, 2018 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-30388619

RESUMO

Impairment of microglial functions, such as phagocytosis and/or dysregulation of immune responses, has been implicated as an underlying factor involved in the pathogenesis of various neurodegenerative disorders. Our previous studies have demonstrated that long intergenic noncoding RNA (lincRNA)-Cox2 expression is influenced by nuclear factor κB (NF-κB) signaling and serves as a coactivator of transcriptional factors to regulate the expression of a vast array of immune-related genes in microglia. Extracellular vesicles (EVs) have been recognized as primary facilitators of cell-to-cell communication and cellular regulation. Herein, we show that EVs derived from astrocytes exposed to morphine can be taken up by microglial endosomes, leading, in turn, to activation of Toll-like receptor 7 (TLR7) with a subsequent upregulation of lincRNA-Cox2 expression, ultimately resulting in impaired microglial phagocytosis. This was further validated in vivo, wherein inhibition of microglial phagocytic activity was also observed in brain slices isolated from morphine-administrated mice compared with control mice. Additionally, we also showed that intranasal delivery of EVs containing lincRNA-Cox2 siRNA (small interfering RNA) was able to restore microglial phagocytic activity in mice administered morphine. These findings have ramifications for the development of EV-loaded RNA-based therapeutics for the treatment of various disorders involving functional impairment of microglia.

16.
Transl Neurodegener ; 7: 29, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30410751

RESUMO

BACKGROUND: Cell replacement therapy has been envisioned as a promising treatment for neurodegenerative diseases. Due to the ethical concerns of ESCs-derived neural progenitor cells (NPCs) and tumorigenic potential of iPSCs, reprogramming of somatic cells directly into multipotent NPCs has emerged as a preferred approach for cell transplantation. METHODS: Mouse astrocytes were reprogrammed into NPCs by the overexpression of transcription factors (TFs) Foxg1, Sox2, and Brn2. The generation of subtypes of neurons was directed by the force expression of cell-type specific TFs Lhx8 or Foxa2/Lmx1a. RESULTS: Astrocyte-derived induced NPCs (AiNPCs) share high similarities, including the expression of NPC-specific genes, DNA methylation patterns, the ability to proliferate and differentiate, with the wild type NPCs. The AiNPCs are committed to the forebrain identity and predominantly differentiated into glutamatergic and GABAergic neuronal subtypes. Interestingly, additional overexpression of TFs Lhx8 and Foxa2/Lmx1a in AiNPCs promoted cholinergic and dopaminergic neuronal differentiation, respectively. CONCLUSIONS: Our studies suggest that astrocytes can be converted into AiNPCs and lineage-committed AiNPCs can acquire differentiation potential of other lineages through forced expression of specific TFs. Understanding the impact of the TF sets on the reprogramming and differentiation into specific lineages of neurons will provide valuable strategies for astrocyte-based cell therapy in neurodegenerative diseases.

17.
Phys Rev E ; 97(2-1): 022220, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29548119

RESUMO

Explosive synchronization in networked second-order Kuramoto oscillators has been well studied recently and it is revealed that the synchronization process is featured by cluster explosive synchronization. However, little attention has been paid to the influence of noise or perturbation. We here study this problem and discuss the influences of noise and perturbation. For the former, we interestingly find that noise has significant influence on the cluster explosive synchronization of those nodes with smaller degrees, i.e., their synchronization will change from the first-order to second-order transition and the critical points for both the forward and backward synchronization depend on the strength of noise. Especially, when the strength of noise is in an optimal range, a synchronization of the nodes with smaller degrees will be induced in the region of coupling strength where they do not display synchronization in the absence of noise. For the latter, we find that the effect of perturbation is similar to that of noise when its duration W is small. However, the perturbation will induce a change from cluster explosive synchronization to explosive synchronization when W is large. Furthermore, a brief theory is provided to explain the influence of perturbations on the critical points.

18.
Am J Physiol Heart Circ Physiol ; 314(5): H928-H939, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29373037

RESUMO

The imbalance between the synthesis of reactive oxygen species and their elimination by antioxidant defense systems results in macromolecular damage and disruption of cellular redox signaling, affecting cardiac structure and function, thus contributing to contractile dysfunction, myocardial hypertrophy, and fibrosis in chronic heart failure [chronic heart failure (CHF)]. The Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway is an important antioxidant defense mechanism and is closely associated with oxidative stress-mediated cardiac remodeling in CHF. In the present study, we investigated the regulation of myocardial Nrf2 in the postmyocardial infarction (post-MI) state. Six weeks post-MI, Nrf2 protein was downregulated in the heart, resulting in a decrease of Nrf2-targeted antioxidant enzymes, whereas paradoxically the transcription of Nrf2 was increased, suggesting that translational inhibition of Nrf2 may contribute to the dysregulation in CHF. We therefore hypothesized that microRNAs may be involved in the translational repression of Nrf2 mRNA in the setting of CHF. Using quantitative real-time PCR analysis, we found that three microRNAs, including microRNA-27a, microRNA-28-3p, and microRNA-34a, were highly expressed in the left ventricle of infarcted hearts compared with other organs. Furthermore, in vitro analysis revealed that cultured cardiac myocytes and fibroblasts expressed these three microRNAs in response to TNF-α stimulation. These microRNAs were preferentially incorporated into exosomes and secreted into the extracellular space in which microRNA-enriched exosomes mediated intercellular communication and Nrf2 dysregulation. Taken together, these results suggest that increased local microRNAs induced by MI may contribute to oxidative stress by the inhibition of Nrf2 translation in CHF. NEW & NOTEWORTHY The results of this work provide a novel mechanism mediated by microRNA-enriched exosomes, contributing to the nuclear factor erythroid 2-related factor 2 dysregulation and subsequent oxidative stress. Importantly, these new findings will provide a promising strategy to improve the therapeutic efficacy through targeting nuclear factor erythroid 2-related factor 2-related microRNAs in the chronic heart failure state, which show potentially clinical applications.


Assuntos
Exossomos/metabolismo , Insuficiência Cardíaca/metabolismo , MicroRNAs/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Animais , Células Cultivadas , Doença Crônica , Modelos Animais de Doenças , Regulação para Baixo , Exossomos/genética , Insuficiência Cardíaca/genética , Masculino , MicroRNAs/genética , Infarto do Miocárdio/genética , Fator 2 Relacionado a NF-E2/genética , Estresse Oxidativo , Ratos Sprague-Dawley , Transdução de Sinais , Transcrição Gênica
19.
Transl Neurodegener ; 6: 10, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28439409

RESUMO

BACKGROUND: Glutaminase 1 is a phosphate-activated metabolic enzyme that catalyzes the first step of glutaminolysis, which converts glutamine into glutamate. Glutamate is the major neurotransmitter of excitatory synapses, executing important physiological functions in the central nervous system. There are two isoforms of glutaminase 1, KGA and GAC, both of which are generated through alternative splicing from the same gene. KGA and GAC both transcribe 1-14 exons in the N-terminal, but each has its unique C-terminal in the coding sequence. We have previously identified that KGA and GAC are differentially regulated during inflammatory stimulation and HIV infection. Furthermore, glutaminase 1 has been linked to brain diseases such as amyotrophic lateral sclerosis, Alzheimer's disease, and hepatic encephalopathy. Core enzyme structure of KGA and GAC has been published recently. However, how other coding sequences affect their functional enzyme activity remains unclear. METHODS: We cloned and performed serial deletions of human full-length KGA and GAC from the N-terminal and the C-terminal at an interval of approximately 100 amino acids (AAs). Prokaryotic expressions of the mutant glutaminase 1 protein and a glutaminase enzyme activity assay were used to determine if KGA and GAC have similar efficiency and efficacy to convert glutamine into glutamate. RESULTS: When 110 AAs or 218 AAs were deleted from the N-terminal or when the unique portions of KGA and GAC that are beyond the 550 AA were deleted from the C-terminal, KGA and GAC retained enzyme activity comparable to the full length proteins. In contrast, deletion of 310 AAs or more from N-terminal or deletion of 450 AAs or more from C-terminal resulted in complete loss of enzyme activity for KGA/GAC. Consistently, when both N- and C-terminal of the KGA and GAC were removed, creating a truncated protein that expressed the central 219 AA - 550 AA, the protein retained enzyme activity. Furthermore, expression of the core 219 AA - 550 AA coding sequence in cells increased extracellular glutamate concentrations to levels comparable to those of full-length KGA and GAC expressions, suggesting that the core enzyme activity of the protein lies within the central 219 AA - 550 AA. Full-length KGA and GAC retained enzyme activities when kept at 4 °C. In contrast, 219 AA - 550 AA truncated protein lost glutaminase activities more readily compared with full-length KGA and GAC, suggesting that the N-terminal and C-terminal coding regions are required for the stability KGA and GAC. CONCLUSIONS: Glutaminase isoforms KGA and GAC have similar efficacy to catalyze the conversion of glutamine to glutamate. The core enzyme activity of glutaminase 1 protein is within the central 219 AA - 550 AA. The N-terminal and C-terminal coding regions of KGA and GAC help maintain the long-term activities of the enzymes.

20.
Sci Rep ; 7(1): 561, 2017 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-28373712

RESUMO

We here study explosive synchronization transitions and network activity propagation in networks of coupled neurons to provide a new understanding of the relationship between network topology and explosive dynamical transitions as in epileptic seizures and their propagations in the brain. We model local network motifs and configurations of coupled neurons and analyze the activity propagations between a group of active neurons to their inactive neuron neighbors in a variety of network configurations. We find that neuronal activity propagation is limited to local regions when network is highly clustered with modular structures as in the normal brain networks. When the network cluster structure is slightly changed, the activity propagates to the entire network, which is reminiscent of epileptic seizure propagation in the brain. Finally, we analyze intracranial electroencephalography (IEEG) recordings of a seizure episode from a epilepsy patient and uncover that explosive synchronization-like transition occurs around the clinically defined onset of seizure. These findings may provide a possible mechanism for the recurrence of epileptic seizures, which are known to be the results of aberrant neuronal network structure and/or function in the brain.


Assuntos
Encéfalo/fisiologia , Rede Nervosa/fisiologia , Neurônios/fisiologia , Transmissão Sináptica , Encéfalo/fisiopatologia , Eletroencefalografia , Epilepsia/fisiopatologia , Humanos , Modelos Neurológicos , Plasticidade Neuronal , Convulsões/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...