Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 489, 2023 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-36717587

RESUMO

Vascular repair is considered a key restorative measure to improve long-term outcomes after ischemic stroke. N6-methyladenosine (m6A), the most prevalent internal modification in eukaryotic mRNAs, functionally mediates vascular repair. However, whether circular RNA SCMH1 (circSCMH1) promotes vascular repair by m6A methylation after stroke remains to be elucidated. Here, we identify the role of circSCMH1 in promoting vascular repair in peri-infarct cortex of male mice and male monkeys after photothrombotic (PT) stroke, and attenuating the ischemia-induced m6A methylation in peri-infarct cortex of male mice after PT stroke. Mechanically, circSCMH1 increased the translocation of ubiquitination-modified fat mass and obesity-associated protein (FTO) into nucleus of endothelial cells (ECs), leading to m6A demethylation of phospholipid phosphatase 3 (Plpp3) mRNA and subsequently the increase of Plpp3 expression in ECs. Our data demonstrate that circSCMH1 enhances vascular repair via FTO-regulated m6A methylation after stroke, providing insights into the mechanism of circSCMH1 in promoting stroke recovery.


Assuntos
Dioxigenase FTO Dependente de alfa-Cetoglutarato , Fosfatidato Fosfatase , RNA Circular , Acidente Vascular Cerebral , Animais , Masculino , Camundongos , Adenosina/metabolismo , Dioxigenase FTO Dependente de alfa-Cetoglutarato/genética , Dioxigenase FTO Dependente de alfa-Cetoglutarato/metabolismo , Células Endoteliais/metabolismo , Infarto , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Acidente Vascular Cerebral/genética , Acidente Vascular Cerebral/terapia , Fosfatidato Fosfatase/genética , Fosfatidato Fosfatase/metabolismo
2.
Noncoding RNA Res ; 6(3): 130-138, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34377876

RESUMO

Given the important role of the blood-brain barrier (BBB) in the central nervous system (CNS), increasing studies have been carried out to determine how the structural and functional integrity of the BBB impacts the pathogenesis of CNS diseases such as stroke, traumatic brain injuries (TBIs), and gliomas. Emerging studies have revealed that noncoding RNAs (ncRNAs) help to maintain the integrity and permeability of the BBB, thereby mediating CNS homeostasis. This review summarizes recent studies that focus on the effects of ncRNAs on the BBB in CNS diseases, including regulating the biological processes of inflammation, necrosis, and apoptosis of cells, affecting the translational dysfunction of proteins and regulating tight junctions (TJs). A comprehensive and detailed understanding of the interaction between ncRNAs and the BBB will lay a solid foundation for the development of early diagnostic methods and effective treatments for CNS diseases.

3.
Autophagy ; 17(10): 2905-2922, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33317392

RESUMO

Stroke is a major public health problem leading to high rates of death and disability worldwide, but no effective pharmacological therapy is currently available except for the use of PLAT (plasminogen activator, tissue). Here we show that PARP14 (poly (ADP-ribose) polymerase family, member 14) level was significantly increased in the peri-infarct zone of photothrombotic stroke (PT) mice. Genetic knockdown and pharmacological inhibition of PARP14 aggravated functional impairment and increased infarct volume in PT mice, while overexpression of PARP14 displayed the opposite effects. Furthermore, PARP14 was abundant in microglia, and downregulation of PARP14 increased post-stroke microglial activation, whereas overexpression of PARP14 alleviated microglial activation, possibly through microglial macroautophagy/autophagy modulation. Mechanistically, overexpression of PARP14 suppressed Lpar5 (lysophosphatidic acid receptor 5) gene transcription to inhibit microglial activation post stroke. Taken together, PARP14 is a stroke-induced signal that restricts microglial activation and promotes functional recovery, and can serve as a novel target to develop new therapeutic agents for stroke. Moreover, these findings may be conducive to proper use of various PARP inhibitors.Abbreviations: 3-MA: 3-methyladenine; AIF1/Iba-1: allograft inflammatory factor 1; CNS: central nervous system; CQ: chloroquine; DAPI: 4',6-diamidino-2-phenylindole; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethyl sulfoxide; ELISA: enzyme-linked immunosorbent assay; FBS: fetal bovine serum; GFAP: glial fibrillary acidic protein; IL1B/IL-1ß: interleukin 1 beta; IL6/IL-6: interleukin 6; LPAR5: lysophosphatidic acid receptor 5; MAP1LC3B: microtubule-associated protein 1 light chain 3 beta; NOS2/iNOS: nitric oxide synthase 2, inducible; OGD: oxygen glucose deprivation; PAR: polymer of poly (ADP ribose); PARP: poly (ADP-ribose) polymerase family; PBS: phosphate-buffered saline; PLAT/tPA: plasminogen activator, tissue; PT: photothrombotic stroke; qPCR: quantitative polymerase chain reaction; Rap: rapamycin; RBFOX3/NeuN: RNA binding protein, fox-1 homolog (C. elegans) 3; SQSTM1: sequestosome 1; TNF/TNF-α: tumor necrosis factor.


Assuntos
Proteínas de Caenorhabditis elegans , Acidente Vascular Cerebral , Animais , Autofagia/fisiologia , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Regulação para Baixo , Camundongos , Microglia/metabolismo , Poli(ADP-Ribose) Polimerases/genética , Proteínas de Ligação a RNA/metabolismo , Acidente Vascular Cerebral/genética
4.
Curr Med Sci ; 40(3): 401-406, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32681245

RESUMO

N6-methyladenosine (m6A) is identified as the most widespread and abundant internal chemical modification of RNA in eukaryotes. A series of proteins including methyltransferases (also known as "writers"), demethylases (also known as "erasers"), and m6A-binding proteins (also known as "readers") were indicated to participate in the m6A methylation. m6A has emerged as a regulator of various cellular, developmental, and disease processes. Notably, there is highest abundance of m6A methylation in brain than in other organs, which indicates that m6A plays an essential role in brain functions. Here, we describe the general features, mechanisms, and functions of m6A in the brain, and discuss the emerging roles of m6A in brain physiology and diseases.


Assuntos
Adenosina/análogos & derivados , Encéfalo/metabolismo , Encéfalo/fisiologia , Adenosina/metabolismo , Animais , Proteínas de Transporte/metabolismo , Eucariotos/metabolismo , Eucariotos/fisiologia , Humanos , Metilação , Metiltransferases/metabolismo , RNA/metabolismo
5.
Biol Psychiatry ; 88(5): 392-404, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32387133

RESUMO

BACKGROUND: N6-methyladenosine (m6A) is the most abundant epigenetic modification in eukaryotic messenger RNAs and is essential for multiple RNA processing events in physiological and pathological processes. However, precisely how m6A methylation is involved in major depressive disorder (MDD) is not fully understood. METHODS: Circular RNA STAG1 (circSTAG1) was screened from the hippocampus of chronic unpredictable stress-treated mice using high-throughput RNA sequencing. Microinjection of circSTAG1 lentivirus into the mouse hippocampus was used to observe the role of circSTAG1 in depression. Sucrose preference, forced swim, and tail suspension tests were performed to evaluate the depressive-like behaviors of mice. Astrocyte dysfunction was examined by GFAP immunostaining and 3D reconstruction. Methylated RNA immunoprecipitation sequence analysis was used to identify downstream targets of circSTAG1/ALKBH5 (alkB homolog 5) axis. Cell Counting Kit-8 assay was performed to evaluate astrocyte viability in vitro. RESULTS: circSTAG1 was significantly decreased in the chronic unpredictable stress-treated mouse hippocampus and in peripheral blood of patients with MDD. Overexpression of circSTAG1 notably attenuated astrocyte dysfunction and depressive-like behaviors induced by chronic unpredictable stress. Further examination indicated that overexpressed circSTAG1 captured ALKBH5 and decreased the translocation of ALKBH5 into the nucleus, leading to increased m6A methylation of fatty acid amide hydrolase (FAAH) messenger RNA and degradation of FAAH in astrocytes with subsequent attenuation of depressive-like behaviors and astrocyte loss induced by corticosterone in vitro. CONCLUSIONS: Our findings dissect the functional link between circSTAG1 and m6A methylation in the context of MDD, providing evidence that circSTAG1 may be a novel therapeutic target for MDD.


Assuntos
Astrócitos , Transtorno Depressivo Maior , Adenosina/análogos & derivados , Amidoidrolases , Animais , Humanos , Camundongos , Proteínas Nucleares , RNA Circular , RNA Mensageiro
6.
Autophagy ; 15(11): 1860-1881, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30966861

RESUMO

Aging-related, nonresolving inflammation in both the central nervous system (CNS) and periphery predisposes individuals to the development of neurodegenerative disorders (NDDs). Inflammasomes are thought to be especially relevant to immune homeostasis, and their dysregulation contributes to inflammation and NDDs. However, few agents have been clinically shown to reduce NDD incidence by targeting inflammasomes. Our study indicated that NLRP3 (NLR family, pyrin domain containing 3) inflammasome is involved in Parkinson disease (PD) progression in patients and various murine models. In addition, the small molecule kaempferol (Ka) protected mice against LPS- and SNCA-induced neurodegeneration by inhibiting NLRP3 inflammasome activation as evidenced by the fact that Ka reduced cleaved CASP1 expression and disrupted NLRP3-PYCARD-CASP1 complex assembly with concomitant decreased IL1B secretion. Mechanically, Ka promoted macroautophagy/autophagy in microglia, leading to reduced NLRP3 protein expression, which in turn deactivated the NLRP3 inflammasome. Intriguingly, ubiquitination was involved in Ka-induced autophagic NLRP3 degradation. These findings were further confirmed in vivo as knockdown of Atg5 expression or autophagy inhibitor treatment significantly inhibited the Ka-mediated NLRP3 inflammasome inhibition and neurodegeneration amelioration. Thus, we demonstrated that Ka promotes neuroinflammatory inhibition via the cooperation of ubiquitination and autophagy, suggesting that Ka is a promising therapeutic strategy for the treatment of NDDs. Abbreviations: 3-MA: 3-methyladenine; AAV: adeno-associated virus; ACTB: actin, beta; AIF1/IBA1: allograft inflammatory factor 1; ATG5: autophagy related 5; ATG7: autophagy related 7; BafA1: bafilomycin A1; BECN1: beclin 1, autophagy related; CASP1: caspase 1; CNS: central nervous system; CQ: chloroquine; DA neurons: dopaminergic neurons; DAMPS: damage-associated molecular patterns; DAPI: 4',6-diamidino-2-phenylindole; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GFAP: glial fibrillary acidic protein; IP: immunoprecipitation; i.p.: intraperitoneally; Ka: kaempferol; KD: knockdown; KO: knockout; LPS: lipopolysaccharide; IL1B: interleukin 1 beta; IL6: interleukin 6; Ly: lysate; MAP1LC3B/LC3B: microtubule-associated protein 1 light chain 3 beta; MPTP: 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NC: negative control; NDD: neurodegenerative diseases; NLRP3: NLR family, pyrin domain containing 3; OE: overexpression; PD: Parkinson disease; poly-Ub: poly-ubiquitin; PTM: post-translational modification; PYCARD/ASC: PYD and CARD domain containing; Rapa: rapamycin; RFP: red fluorescent protein; SN: supernatant; SNCA: synuclein alpha; SNpc: substantia nigra pars compacta; SQSTM1: sequestosome 1; TH: tyrosine hydroxylase; TNF/TNF-alpha: tumor necrosis factor; Ub: ubiquitin; WT: wild type.


Assuntos
Autofagia/efeitos dos fármacos , Inflamassomos/efeitos dos fármacos , Inflamação/tratamento farmacológico , Quempferóis/uso terapêutico , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doença de Parkinson/metabolismo , Ubiquitinação/efeitos dos fármacos , Animais , Autofagia/genética , Proteína 5 Relacionada à Autofagia/genética , Proteína 5 Relacionada à Autofagia/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Células HEK293 , Humanos , Inflamassomos/metabolismo , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/toxicidade , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/patologia
7.
Toxicol Lett ; 301: 53-63, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30394308

RESUMO

Nod-like Receptor Protein 3 (NLRP3) inflammasome activation is known to lead to microglia-mediated neuroinflammation. Methamphetamine is known to induce microglial activation. However, whether NLRP3 inflammasome activation contributes to the microglial activation induced by methamphetamine remains elusive. P53-up-regulated modulator of apoptosis (PUMA) is a known apoptosis inducer; however, their role in microglial activation remains poorly understood. Methamphetamine treatment induced NLRP3 inflammasome activation as well microglial activation in animal model. Intriguingly, downregulation of PUMA significantly inhibited the activation of microglia. Methamphetamine treatment increased the expression of PUMA at protein level but not mRNA level. Further study indicated that PUMA expression was regulated at post-transcriptional level by miR-143, which was decreased in methamphetamine-treated cells via the negative transcription factor nuclear factor-kappa B1 (NF-κB1). Using gain- and loss-of-function approaches, we identified a unique role of miR-143/PUMA in mediating microglial activation via regulation of NLRP3 inflammasome activation. These findings provide new insight regarding the specific contributions of the miR-143/PUMA pathway to NLRP3 inflammasome activation in the context of drug abuse.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Inflamassomos/metabolismo , Metanfetamina/toxicidade , MicroRNAs/metabolismo , Microglia/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica , Vetores Genéticos , Lentivirus/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteínas Supressoras de Tumor/genética
8.
Metab Brain Dis ; 34(1): 61-69, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30259295

RESUMO

In this study, we demonstrated that PUMA was involved in the microglial migration induced by methamphetamine. PUMA expression was examined by western blotting and immunofluorescence staining. BV2 and HAPI cells were pretreated with a sigma-1R antagonist and extracellular signal-regulated kinase (ERK), mitogen-activated protein kinase (MAPK), c-Jun N-terminal protein kinase (JNK), and phosphatidylinositol-3 kinase (PI3K)/Akt inhibitors, and PUMA expression was detected by western blotting. The cell migration in BV2 and HAPI cells transfected with a lentivirus encoding red fluorescent protein (LV-RFP) was also examined using a wound-healing assay and nested matrix model and cell migration assay respectively. The molecular mechanisms of PUMA in microglial migration were validated using a siRNA approach. The exposure of BV2 and HAPI cells to methamphetamine increased the expression of PUMA, reactive oxygen species (ROS), the MAPK and PI3K/Akt pathways and the downstream transcription factor signal transducer and activator of transcription 3 (STAT3) pathways. PUMA knockdown in microglia transfected with PUMA siRNA attenuated the increased cell migration induced by methamphetamine, thereby implicating PUMA in the migration of BV2 and HAPI cells. This study demonstrated that methamphetamine-induced microglial migration involved PUMA up-regulation. Targeting PUMA could provide insights into the development of a potential therapeutic approach for the alleviation of microglia migration induced by methamphetamine.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Movimento Celular/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Metanfetamina/farmacologia , Microglia/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Linhagem Celular , Humanos , Camundongos , Microglia/citologia , Microglia/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptores sigma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor Sigma-1
9.
J Neuroinflammation ; 15(1): 221, 2018 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-30075790

RESUMO

BACKGROUND: Mice with pilocarpine-induced temporal lobe epilepsy (TLE) are characterized by intense hippocampal neuroinflammation, a prominent pathological hallmark of TLE that is known to contribute to neuronal hyperexcitability. Recent studies indicate that Adam10, a member of a disintegrin and metalloproteinase domain-containing protein (Adam) family, has been involved in the neuroinflammation response. However, it remains unclear whether and how Adam10 modulates neuroinflammation responses in the context of an epileptic brain or whether Adam10 affects epileptogenesis via the neuroinflammation pathway. METHODS: Adult male C57BL/6J mice were subjected to intraperitoneal injection of pilocarpine to induce TLE. Adeno-associated viral (AAV) vectors carrying Adam10 (AAV-Adam10) or lentiviral vectors carrying short hairpin RNA, which is specific to the mouse Adam10 mRNA (shRNA-Adam10), were bilaterally injected into the hippocampus to induce overexpression or knockdown of Adam10, respectively. The specific anti-inflammatory agent minocycline was administered following status epilepticus (SE) to block hippocampal neuroinflammation. Continuous video EEG recording was performed to analyze epileptic behavior. Western blot, immunofluorescence staining, and ELISA were performed to determine Adam10 expression as well as hippocampal neuroinflammation. RESULTS: In this study, we demonstrate that overexpression of Adam10 in the hippocampus suppresses neuroinflammation and reduces seizure activity in TLE mice, whereas knockdown of Adam10 exacerbates hippocampal neuroinflammation and increases seizure activity. Furthermore, increased seizure activity in Adam10 knockdown TLE mice is dependent on hippocampal neuroinflammation. CONCLUSION: These results suggest that Adam10 suppresses epilepsy through repression of hippocampal neuroinflammation. Our findings provide new insights into the Adam10 regulation of development of epilepsy via the neuroinflammation pathway and identify a potential therapeutic target for epilepsy.


Assuntos
Proteína ADAM10/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Encefalite/etiologia , Proteína Glial Fibrilar Ácida/fisiologia , Hipocampo/metabolismo , Proteínas de Membrana/metabolismo , Estado Epiléptico , Proteína ADAM10/genética , Secretases da Proteína Precursora do Amiloide/genética , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Ciclo-Oxigenase 2/metabolismo , Modelos Animais de Doenças , Encefalite/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/metabolismo , Hipocampo/efeitos dos fármacos , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Agonistas Muscarínicos/toxicidade , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Pilocarpina/toxicidade , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Estado Epiléptico/induzido quimicamente , Estado Epiléptico/complicações , Estado Epiléptico/patologia , Estado Epiléptico/terapia
10.
J Neuroimmune Pharmacol ; 13(3): 330-344, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29497921

RESUMO

Chronic neuroinflammation still remains a common underlying feature of HIV-infected patients on combined anti-retroviral therapy (cART). Previous studies have reported that despite near complete suppression of virus replication by cART, cytotoxic viral proteins such as HIV trans-activating regulatory protein (Tat) continue to persist in tissues such as the brain and the lymph nodes, thereby contributing, in part, to chronic glial activation observed in HIV-associated neurological disorders (HAND). Understanding how the glial cells cross talk to mediate neuropathology is thus of paramount importance. MicroRNAs (miR) also known as regulators of gene expression, have emerged as key paracrine signaling mediators that regulate disease pathogenesis and cellular crosstalk, through their transfer via the extracellular vesicles (EV). In the current study we have identified a novel function of miR-9, that of mediating microglial migration. We demonstrate that miR-9 released from Tat-stimulated astrocytes can be taken up by microglia resulting in their migratory phenotype. Exposure of human astrocytoma (A172) cells to HIV Tat resulted in induction and release of miR-9 in the EVs, which, was taken up by microglia, leading in turn, increased migration of the latter cells, a process that could be blocked by both an exosome inhibitor GW4869 or a specific target protector of miR-9. Furthermore, it was also demonstrated that EV miR-9 mediated inhibition of the expression of target PTEN, via its binding to the 3'UTR seed sequence of the PTEN mRNA, was critical for microglial migration. To validate the role of miR-9 in this process, microglial cells were treated with EVs loaded with miR-9, which resulted in significant downregulation of PTEN expression with a concomitant increase in microglial migration. These findings were corroborated by transfecting microglia with a specific target protector of PTEN, that blocked miR-9-mediated downregulation of PTEN as well as microglial migration. In vivo studies wherein the miR-9 precursor-transduced microglia were transplanted into the striatum of mice, followed by assessing their migration in response to a stimulus administered distally, further validated the role of miR-9 in mediating microglial migration. Collectively, our findings provide evidence that glial crosstalk via miRs released from EVs play a vital role in mediating disease pathogenesis and could provide new avenues for development of novel therapeutic strategies aimed at dampening neuropathogenesis.


Assuntos
Astrócitos/metabolismo , Exossomos/metabolismo , MicroRNAs/metabolismo , MicroRNAs/fisiologia , Microglia , Produtos do Gene tat do Vírus da Imunodeficiência Humana/farmacologia , Regiões 3' não Traduzidas , Compostos de Anilina/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Compostos de Benzilideno/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Infecções por HIV/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neostriado/citologia , PTEN Fosfo-Hidrolase/efeitos dos fármacos , Transfecção
11.
Pharmacol Ther ; 187: 31-44, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29406246

RESUMO

The emerging recognition of the functional roles of circular RNAs (circRNAs) has given rise to a new perspective regarding our understanding of cellular physiology and disease pathogenesis. Unlike linear RNAs, circRNAs are covalently closed continuous loops that act as gene regulators in mammals, and their sequence composition determines the mode of circRNA biogenesis. The availability and integrated use of advanced genome analysis platforms have allowed the identification of a large number of these molecules. Their high abundance, stability and evolutionary conservation among species endow circRNAs with numerous potential functions, such as acting as microRNA (miRNA) sponges or binding to RNA-associated proteins to form RNA-protein complexes that regulate gene transcription. Moreover, circRNAs have been shown to be expressed in a tissue-specific manner and in pathological conditions, which has stimulated significant interest in their role in human disease and cancer. In this concise review, we outline the characteristics, functions and mechanisms of action of circRNAs as well as their involvement in different diseases. Although their exact roles and mechanisms of gene regulation remain to be clarified, circRNAs have potential applications as disease biomarkers and novel therapeutic targets.


Assuntos
RNA/fisiologia , Animais , Doença , Humanos , RNA Circular
12.
Stem Cells Int ; 2017: 2707082, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28904531

RESUMO

Stem cell therapy is a promising potential therapeutic strategy to treat cerebral ischemia in preclinical and clinical trials. Currently proposed treatments for stroke employing stem cells include the replacement of lost neurons and integration into the existing host circuitry, the release of growth factors to support and promote endogenous repair processes, and the secretion of extracellular vesicles containing proteins, noncoding RNA, or DNA to regulate gene expression in recipient cells and achieve immunomodulation. Progress has been made to elucidate the precise mechanisms underlying stem cell therapy and the homing, migration, distribution, and differentiation of transplanted stem cells in vivo using various imaging modalities. Noninvasive and safe tracer agents with high sensitivity and image resolution must be combined with long-term monitoring using imaging technology to determine the optimal therapy for stroke in terms of administration route, dosage, and timing. This review discusses potential therapeutic mechanisms of stem cell transplantation for the treatment of stroke and the limitations of current therapies. Methods to label transplanted cells and existing imaging systems for stem cell labeling and in vivo tracking will also be discussed.

13.
Exp Cell Res ; 356(1): 28-39, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28408317

RESUMO

Mounting evidence indicates that methamphetamine causes blood-brain barrier damage, with emphasis on endothelial cells. The role of pericytes in methamphetamine-induced BBB damage remains unknown. Our study demonstrated that methamphetamine increased the migration of pericytes from the endothelial basement membrane. However, the detailed mechanisms underlying this process remain poorly understood. Thus, we examined the molecular mechanisms involved in methamphetamine-induced pericyte migration. The results showed that exposure of C3H/10T1/2 cells and HBVPs to methamphetamine increased PUMA expression via activation of the sigma-1 receptor, MAPK and Akt/PI3K pathways. Moreover, methamphetamine treatment resulted in the increased migration of C3H/10T1/2 cells and HBVPs. Knockdown of PUMA in pericytes transduced with PUMA siRNA attenuated the methamphetamine-induced increase in cell migration through attenuation of integrin and tyrosine kinase mechanisms, implicating a role of PUMA in the migration of C3H/10T1/2 cells and HBVPs. This study has demonstrated that methamphetamine-mediated pericytes migration involves PUMA up-regulation. Thus, targeted studies of PUMA could provide insights to facilitate the development of a potential therapeutic approach for alleviation of methamphetamine-induced pericyte migration.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Barreira Hematoencefálica/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Estimulantes do Sistema Nervoso Central/farmacologia , Metanfetamina/farmacologia , Pericitos/fisiologia , Receptores sigma/metabolismo , Proteínas Supressoras de Tumor/biossíntese , Animais , Proteínas Reguladoras de Apoptose/genética , Membrana Basal/citologia , Membrana Basal/efeitos dos fármacos , Adesão Celular , Linhagem Celular , Células Endoteliais/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/genética , Proteínas Supressoras de Tumor/genética , Receptor Sigma-1
14.
Cell Death Dis ; 8(3): e2657, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28277537

RESUMO

Following inhalation into the lungs, silica particles are engulfed by alveolar macrophages, which triggers endogenous or exogenous apoptosis signaling pathways. As an inducer of apoptosis, the role of BBC3/PUMA (BCL2-binding component 3) in macrophages during silicosis remains unknown. Here, we exposed U937 cell-derived macrophages (UDMs) to SiO2 in vitro to explore the function of BBC3 in SiO2-induced disease. We found that SiO2 induced increased BBC3 expression, as well as macrophage activation and apoptosis. Knockdown of Bbc3 with specific siRNA significantly mitigated the SiO2-induced effects. In addition, our results clearly showed increased levels of autophagy in macrophages exposed to SiO2. However, inhibition of BBC3 decreased the occurrence of autophagy. Furthermore, we observed that the blockade of autophagy with 3-MA, an autophagy inhibitor, inhibited SiO2-induced macrophage activation and apoptosis. In contrast, rapamycin, an autophagy inducer, further enhanced the effects induced by SiO2. The conditioned medium from macrophages exposed to SiO2 promoted the proliferation and migration of fibroblasts, and the inhibition of BBC3/autophagy reduced the effects of the conditioned medium on fibroblasts. In the mouse model of silicosis, Bbc3 knockout mice clearly exhibited decreased levels of autophagy and fibrosis progression. These results suggest that downregulation of BBC3 expression may become a novel therapeutic strategy for the treatment of silicosis.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Autofagia/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Fibrose Pulmonar/genética , Silicose/genética , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Autofagia/genética , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/patologia , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/patologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Fibrose Pulmonar/tratamento farmacológico , Fibrose Pulmonar/patologia , Dióxido de Silício/toxicidade , Silicose/complicações , Silicose/tratamento farmacológico , Silicose/patologia , Sirolimo/administração & dosagem , Células U937
15.
Cell Physiol Biochem ; 40(1-2): 309-325, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27866190

RESUMO

BACKGROUND: Silicosis is characterized by the accumulation of fibroblasts and the excessive deposition of extracellular matrix. Fibroblast generation via endothelial-mesenchymal transition (EndMT) is one process responsible for this accumulation of fibroblasts. However, the mechanisms underlying EndMT remain unknown. METHODS: Human umbilical vein endothelial cells (HUVECs) were exposed to SiO2 (50 µg/cm2). Specific endothelial and mesenchymal markers were evaluated using immunofluorescence and western blot analysis. Functional changes were evaluated by analyzing cell migration and proliferation. LC3-adenovirus transfections were performed, and changes in autophagy were measured using a marker of autophagy. RESULTS: SiO2 induced decreases in the endothelial cell-specific markers in HUVECs while dramatically increasing mesenchymal cell product levels and mesenchymal functions. Although MCPIP1 expression increased in parallel with the increase in specific mesenchymal cell products, the MCPIP1 expression level was not consistent with the observed decrease in specific endothelial marker expression. Autophagy mediated the effects of MCPIP1, as rapamycin and 3-MA enhanced and attenuated the effect of SiO2 on HUVECs, respectively. MAPKs and the PI3K/Akt pathway were involved in the regulation of MCPIP1 by SiO2, and Pyk2 and MLC-2 mediated cell migration. CONCLUSION: Our findings reveal a new potential function of MCPIP1, suggesting a possible mechanism of fibrosis in pulmonary silicosis.


Assuntos
Células Endoteliais da Veia Umbilical Humana/metabolismo , Mesoderma/metabolismo , Ribonucleases/metabolismo , Dióxido de Silício/farmacologia , Fatores de Transcrição/metabolismo , Autofagia/efeitos dos fármacos , Miosinas Cardíacas/metabolismo , Movimento Celular/efeitos dos fármacos , Quinase 2 de Adesão Focal/metabolismo , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Humanos , Mesoderma/efeitos dos fármacos , Cadeias Leves de Miosina/metabolismo , Fenótipo , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
16.
Part Fibre Toxicol ; 13(1): 55, 2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27782836

RESUMO

BACKGROUND: Silicosis is characterized by accumulation of fibroblasts and excessive deposition of extracellular matrix. Monocyte chemotactic protein-1-induced protein 1 (MCPIP1) plays a critical role in fibrosis induced by SiO2. However, the details of the downstream events of MCPIP1 activity in pulmonary fibrosis remain unclear. To elucidate the role of MCPIP1-induced autophagy in SiO2-induced fibrosis, both the upstream molecular mechanisms and the functional effects of SiO2 on cell apoptosis, proliferation and migration were investigated. RESULTS: Experiments using primary cultures of alveolar macrophages from healthy donors and silicosis patients as well as differentiated U937 macrophages demonstrated the following results: 1) SiO2 induced macrophage autophagy in association with enhanced expression of MCPIP1; 2) autophagy promoted apoptosis and activation of macrophages exposed to SiO2, and these events induced the development of silicosis; 3) MCPIP1 facilitated macrophage apoptosis and activation via p53 signaling-mediated autophagy; and 4) SiO2-activated macrophages promoted the proliferation and migration of fibroblasts via the MCPIP1/p53-mediated autophagy pathway. CONCLUSIONS: Our results elucidated a link between SiO2-induced fibrosis and MCPIP1/p53 signaling-mediated autophagy. These findings provide novel insight into the potential targeting of MCPIP1 or autophagy in the development of potential therapeutic strategies for silicosis.


Assuntos
Autofagia/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Fibrose Pulmonar/induzido quimicamente , Ribonucleases/fisiologia , Dióxido de Silício/toxicidade , Fatores de Transcrição/fisiologia , Apoptose/efeitos dos fármacos , Humanos , Macrófagos/imunologia , RNA Interferente Pequeno/genética , Ribonucleases/genética , Transdução de Sinais , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/metabolismo , Células U937
17.
Sci Rep ; 6: 35642, 2016 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-27767041

RESUMO

MicroRNA-143 (miR-143) plays a critical role in various cellular processes; however, the role of miR-143 in the maintenance of blood-brain barrier (BBB) integrity remains poorly defined. Silencing miR-143 in a genetic animal model or via an anti-miR-143 lentivirus prevented the BBB damage induced by methamphetamine. miR-143, which targets p53 unregulated modulator of apoptosis (PUMA), increased the permeability of human brain endothelial cells and concomitantly decreased the expression of tight junction proteins (TJPs). Silencing miR-143 increased the expression of TJPs and protected the BBB integrity against the effects of methamphetamine treatment. PUMA overexpression increased the TJP expression through a mechanism that involved the NF-κB and p53 transcription factor pathways. Mechanistically, methamphetamine mediated up-regulation of miR-143 via sigma-1 receptor with sequential activation of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol-3' kinase (PI3K)/Akt and STAT3 pathways. These results indicated that silencing miR-143 could provide a novel therapeutic strategy for BBB damage-related vascular dysfunction.


Assuntos
Transtornos Relacionados ao Uso de Anfetaminas/fisiopatologia , Barreira Hematoencefálica/fisiologia , Metanfetamina/efeitos adversos , MicroRNAs/antagonistas & inibidores , Transtornos Relacionados ao Uso de Anfetaminas/genética , Transtornos Relacionados ao Uso de Anfetaminas/terapia , Animais , Proteínas Reguladoras de Apoptose/antagonistas & inibidores , Proteínas Reguladoras de Apoptose/deficiência , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Barreira Hematoencefálica/efeitos dos fármacos , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Inativação Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/fisiologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas de Junções Íntimas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
18.
Autophagy ; 12(9): 1538-59, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27464000

RESUMO

BBC3 (BCL2 binding component 3) is a known apoptosis inducer; however, its role in microglial survival remains poorly understood. In addition to the classical transcription factor TRP53, Mir143 is involved in BBC3 expression at the post-transcriptional level. Here, we identify unique roles of Mir143-BBC3 in mediating microglial survival via the regulation of the interplay between apoptosis and autophagy. Autophagy inhibition accelerated methamphetamine-induced apoptosis, whereas autophagy induction attenuated the decrease in microglial survival. Moreover, anti-Mir143-dependent BBC3 upregulation reversed the methamphetamine-induced decrease in microglial survival via the regulation of apoptosis and autophagy. The in vivo relevance of these findings was confirmed in mouse models, which demonstrated that the microinjection of anti-Mir143 into the hippocampus ameliorated the methamphetamine-induced decrease in microglia as well as that observed in heterozygous Mir143(+/-) mice. These findings provide new insight regarding the specific contributions of Mir143-BBC3 to microglial survival in the context of drug abuse.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose , Autofagia , Metanfetamina/toxicidade , MicroRNAs/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Regiões 3' não Traduzidas , Animais , Sobrevivência Celular , Modelos Animais de Doenças , Heterozigoto , Hipocampo/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/genética , Microglia/metabolismo , Microglia/patologia , Proteínas Proto-Oncogênicas/metabolismo , Transcrição Gênica , Regulação para Cima
19.
CNS Neurol Disord Drug Targets ; 15(7): 857-65, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27189473

RESUMO

It has been well documented that methamphetamine induces microglial activation and death, however, the molecular mechanisms underlying this process remain poorly understood. In the present study, we demonstrated the involvement of sigma-1 receptor (σ-1R) in methamphetamine-mediated microglial apoptosis. Exposure of BV-2 cells to methamphetamine induces cell apoptosis through its cognate receptor σ-1R, followed by activation of the mitogen-activated protein kinases, phosphatidylinositol-3' kinase/Akt as well as the downstream transcription factor p53 pathways. Blockage of σ -1R significantly inhibited the increased pro-apoptotic proteins such as Bax, Caspase-3 and Caspase-9 induced by methamphetamine. In conclusion, these findings underscore the critical role of σ-1R in microglial apoptosis induced by methamphetamine. Understanding the link between σ -1R and apoptosis will lead to development of therapeutic strategies targeting methamphetamine-mediated microglial death/dysfunction.


Assuntos
Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Metanfetamina/toxicidade , Microglia/efeitos dos fármacos , Microglia/metabolismo , Receptores sigma/metabolismo , Animais , Western Blotting , Caspases/metabolismo , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores sigma/antagonistas & inibidores , Receptores sigma/genética , Proteína Supressora de Tumor p53/metabolismo , Receptor Sigma-1
20.
PLoS One ; 11(4): e0153756, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27093556

RESUMO

AIMS: To precisely characterize the penumbra by MRI based on a modified photothrombotic stroke mouse model. METHODS: The proximal middle cerebral artery was occluded by a convenient laser system in conjunction with an intravenous injection of Rose Bengal in mice. And the suture MCAO model was performed in seven mice as a comparison of the reproducibility. One hour after occlusion, the penumbra was defined in six random photothrombotic stroke mice by mismatch between perfusion-weighted imaging and the apparent diffusion coefficient map on a home-made workstation. After imaging, three random mice of them were chosen to perform the reperfusion surgery. And the other three mice were sacrificed to stain for several potential penumbra markers, such as c-fos and heart shock protein 90. In the remaining mice, the evolution of the lesions was detected on the apparent diffusion coefficient map, diffusion-weighted imaging and T2-weighted imaging at 1, 3, 6, 12 and 24 hours. After evaluating the neurological deficit scores, the brains were sectioned and stained by triphenyltetrazolium chloride and Nissl. RESULTS: The mice subjected to photothrombosis showed significant behavioral deficits. One hour after occlusion, the low perfusion areas on the perfusion-weighted imaging interlaced with the hypointense areas on the apparent diffusion coefficient map, demonstrating that the penumbra was located both surrounding and inside the lesions. This phenomenon was subsequently confirmed by the c-fos and heart shock protein 90 staining. The final T2-weighted images of the mice subjected to the reperfusion surgery were also consistent with the penumbra images at one hour. At early stages, the lesions were clearly identified on the apparent diffusion coefficient map; the volumes of the lesions on the diffusion-weighted imaging and T2-weighted imaging did not reach a maximum until 12 hours. The coefficient of variation (CV) of the final lesions in the photothrombotic stroke mice was 21.7% (0.08 of 0.37) on T2-weighted imaging and 27.8% (0.10 of 0.35) on triphenyltetrazolium chloride, representing a high reproducibility (n = 7). While the CV of the lesions in the MCAO stroke mice was only 70% (0.24 of 0.34, n = 4). CONCLUSIONS: This study has provided a precise imaging definition of the penumbra based on a reproducible photothrombotic stroke mouse model.


Assuntos
Isquemia Encefálica/patologia , Encéfalo/patologia , Acidente Vascular Cerebral/patologia , Animais , Imagem de Difusão por Ressonância Magnética/métodos , Modelos Animais de Doenças , Processamento de Imagem Assistida por Computador/métodos , Infarto da Artéria Cerebral Média/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Artéria Cerebral Média/patologia , Reperfusão/métodos , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA