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











Base de dados
Intervalo de ano de publicação
1.
J Control Release ; 364: 601-617, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37926244

RESUMO

Stem cell transplantation holds great promise for restoring function after spinal cord injury (SCI), but its therapeutic efficacy heavily depends on the innate capabilities of the cells and the microenvironment at the lesion site. Herein, a potent cell therapeutic (NCs@SCs) is engineered by artificially reprogramming bone marrow mesenchymal stem cells (BMSCs) with oxidation-responsive transcytosable gene-delivery nanocomplexes (NCs), which endows cells with robust oxidative stress resistance and improved cytokine secretion. NCs@SCs can accumulate in the injured spinal cord after intravenous administration via chemotaxis and boost successive transcytosis to deliver NCs to neurons, augmenting ciliary neurotrophic factor (CNTF) production in both BMSCs and neurons in response to elevated ROS levels. Furthermore, NCs@SCs can actively sense and eliminate ROS and re-educate recruited M1-like macrophages into the anti-inflammatory M2 phenotype via a paracrine pathway, ultimately reshaping the inflammatory microenvironment. Synergistically, NCs@SCs exhibit durable survival and provide neuroprotection against secondary damage, enabling significant locomotor function recovery in SCI rats. Transcriptome analysis reveals that regulation of the ROS/MAPK signaling pathway is involved in SCI therapy by NCs@SCs. This study presents a nanomaterial-mediated cell-reprogramming approach for developing live cell therapeutics, showing significant potential in the treatment of SCI and other neuro-injury disorders.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Traumatismos da Medula Espinal , Regeneração da Medula Espinal , Ratos , Animais , Espécies Reativas de Oxigênio/metabolismo , Traumatismos da Medula Espinal/terapia , Neurônios/metabolismo , Medula Espinal/metabolismo , Células-Tronco Mesenquimais/metabolismo , Recuperação de Função Fisiológica/fisiologia
2.
Ultrason Sonochem ; 81: 105866, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34896805

RESUMO

A new ultrasound-assisted enzymatic extraction (UAEE) method of starch from kiwifruit was established and optimized using response surface methodology (RSM). Under optimal conditions (the pectinase-to-cellulase-to-papain ratio = 1:2:1 g/kg, solid/liquid ratio = 1:6.68, extraction pH = 5.23, ultrasound power = 300 W, and extraction temperature = 52 °C), the kiwi starch (KS) yield was about 4.25%, and the starch content of KS was 873.23 mg/g. Compared to other extraction methods, UAEE can obtain KS with high yield and purity with a shorter extraction time and less solvent and enzyme. The extracted KS has a low gelatinization enthalpy (8.02 J/g) and a high peak viscosity (7933 cP), with obvious particle properties and low adhesion. In addition, KS is rich in polyphenols, has strong antioxidant activity, and has higher contents of amylose starch (30.74%) and resistant starch (60.18%). This study established a novel and highly efficient method for KS extraction and suggest several possible applications for KS in the food industry.


Assuntos
Frutas , Amido , Ultrassom , Frutas/química , Amido/química , Amido/isolamento & purificação , Amido/fisiologia
3.
Foods ; 10(9)2021 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-34574153

RESUMO

Fruit thinning is a cultivation technique that is widely applied in horticulture in order to obtain high-quality horticultural crops. This practice results in the discarding of a large number of thinned unripe fruits in orchards each year, which produces a great waste of agricultural resources and causes soil pollution that may be an important reservoir for pest and plant diseases. Current studies showed that bioactive compounds such as polyphenols, organic acids, monosaccharides and starches are present in unripe fruits. Therefore, we reviewed the bioactive components obtained from thinned unripe fruits, their revalorization for the food industry, their beneficial effects for human health and the methods for obtaining these components. We also performed a calculation of the costs and benefits of obtaining these bioactive compounds, and we proposed future research directions. This review provides a reference for the effective utilization and industrial development of thinned unripe fruits obtained from horticultural crops. Furthermore, revalorizing the waste from this cultural practice may increase the economic benefits and relieve the environmental stress.

4.
Immunity ; 54(8): 1728-1744.e7, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34343498

RESUMO

Inflammatory bowel disease (IBD) mainly includes Crohn's disease (CD) and ulcerative colitis (UC). Immune disorders play an essential role in the pathogenesis of these two IBDs, but the differences in the immune microenvironment of the colon and their underlying mechanisms remain poorly investigated. Here we examined the immunological features and metabolic microenvironment of untreated individuals with IBD by multiomics analyses. Modulation of CD-specific metabolites, particularly reduced selenium, can obviously shape type 1 T helper (Th1) cell differentiation, which is specifically enriched in CD. Selenium supplementation suppressed the symptoms and onset of CD and Th1 cell differentiation via selenoprotein W (SELW)-mediated cellular reactive oxygen species scavenging. SELW promoted purine salvage pathways and inhibited one-carbon metabolism by recruiting an E3 ubiquitin ligase, tripartite motif-containing protein 21, which controlled the stability of serine hydroxymethyltransferase 2. Our work highlights selenium as an essential regulator of T cell responses and potential therapeutic targets in CD.


Assuntos
Antioxidantes/farmacologia , Doença de Crohn/tratamento farmacológico , Doença de Crohn/imunologia , Selênio/farmacologia , Selenoproteína W/metabolismo , Células Th1/citologia , Diferenciação Celular/imunologia , Polaridade Celular , Colo/imunologia , Colo/patologia , Glicina Hidroximetiltransferase/metabolismo , Humanos , Espécies Reativas de Oxigênio/metabolismo , Ribonucleoproteínas/metabolismo , Células Th1/imunologia , Ubiquitina-Proteína Ligases/metabolismo
5.
J Exp Med ; 218(3)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33616624

RESUMO

Frequent outbreaks of viruses have caused a serious threat to public health. Previous evidence has revealed that DNA methylation is correlated with viral infections, but its role in innate immunity remains poorly investigated. Additionally, DNA methylation inhibitors promote IFN-I by upregulating endogenous retrovirus; however, studies of intrinsically demethylated tumors do not support this conclusion. This study found that Uhrf1 deficiency in myeloid cells significantly upregulated Ifnb expression, increasing resistance to viral infection. We performed whole-genome bisulfite sequencing and found that a single-nucleotide methylation site in the Ifnb promoter region disrupted IRF3 recruitment. We used site-specific mutant knock-in mice and a region-specific demethylation tool to confirm that this methylated site plays a critical role in regulating Ifnb expression and antiviral responses. These findings provide essential insight into DNA methylation in the regulation of the innate antiviral immune response.


Assuntos
Antivirais/metabolismo , Metilação de DNA/genética , Imunidade Inata/genética , Interferon Tipo I/metabolismo , Nucleotídeos/genética , Animais , Sequência de Bases , Proteínas Estimuladoras de Ligação a CCAAT/deficiência , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Cromatina/metabolismo , Ilhas de CpG/genética , Citocinas/metabolismo , Células HEK293 , Homeostase , Humanos , Sistema Imunitário/metabolismo , Vacinas contra Influenza/imunologia , Camundongos , Células Mieloides/metabolismo , Orthomyxoviridae/fisiologia , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Regiões Promotoras Genéticas/genética , Transdução de Sinais , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo , Transcrição Gênica , Transcriptoma/genética , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/metabolismo
6.
Aging (Albany NY) ; 12(14): 14791-14807, 2020 07 18.
Artigo em Inglês | MEDLINE | ID: mdl-32687062

RESUMO

Protection of telomere 1 (POT1), the telomeric single-stranded DNA (ssDNA)-binding protein in the shelterin complex, has been implicated in the DNA damage response, tumorigenesis and aging. Telomere dysfunction induced by telomere deprotection could accelerate cellular senescence in primary human cells. While previous work demonstrated the biological mechanism of POT1 in aging and cancer, how POT1 is posttranscriptionally regulated remains largely unknown. To better understand the POT1 regulatory axis, we performed bioinformatic prediction, and selected candidates were further confirmed by dual-luciferase reporter assay. Collectively, our results revealed that miR-185 can significantly reduce POT1 mRNA and protein levels by directly targeting the POT1 3'-untranslated region (3'-UTR). Overexpression of miR-185 increased telomere dysfunction-induced foci (TIF) signals in both cancer cells and primary human fibroblasts. Elevated miR-185 led to telomere elongation in the telomerase-positive cell line HTC75, which was phenotypically consistent with POT1 knocking down. Moreover, miR-185 accelerated the replicative senescence process in primary human fibroblasts in a POT1-dependent manner. Interestingly, increased serum miR-185 could represent a potential aging-related biomarker. Taken together, our findings reveal miR-185 as a novel aging-related miRNA that targets POT1 and provide insight into the telomere and senescence regulatory network at both the intracellular and extracellular levels.


Assuntos
Senescência Celular/genética , MicroRNAs/genética , Proteínas de Ligação a Telômeros/genética , Telômero/genética , Regiões 3' não Traduzidas/genética , Envelhecimento/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Biomarcadores Tumorais , Linhagem Celular Tumoral , Biologia Computacional , Técnicas de Silenciamento de Genes , Humanos , Complexo Shelterina , Transdução de Sinais/genética
7.
Oxid Med Cell Longev ; 2020: 9741369, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31998447

RESUMO

Spinal cord injury (SCI) is a devastating disease that may lead to lifelong disability. Thus, seeking for valid drugs that are beneficial to promoting axonal regrowth and elongation after SCI has gained wide attention. Metformin, a glucose-lowering agent, has been demonstrated to play roles in various central nervous system (CNS) disorders. However, the potential protective effect of metformin on nerve regeneration after SCI is still unclear. In this study, we found that the administration of metformin improved functional recovery after SCI through reducing neuronal cell apoptosis and repairing neurites by stabilizing microtubules via PI3K/Akt signaling pathway. Inhibiting the PI3K/Akt pathway with LY294002 partly reversed the therapeutic effects of metformin on SCI in vitro and vivo. Furthermore, metformin treatment weakened the excessive activation of oxidative stress and improved the mitochondrial function by activating the nuclear factor erythroid-related factor 2 (Nrf2) transcription and binding to the antioxidant response element (ARE). Moreover, treatment with Nrf2 inhibitor ML385 partially abolished its antioxidant effect. We also found that the Nrf2 transcription was partially reduced by LY294002 in vitro. Taken together, these results revealed that the role of metformin in nerve regeneration after SCI was probably related to stabilization of microtubules and inhibition of the excessive activation of Akt-mediated Nrf2/ARE pathway-regulated oxidative stress and mitochondrial dysfunction. Overall, our present study suggests that metformin administration may provide a potential therapy for SCI.


Assuntos
Axônios/fisiologia , Metformina/farmacologia , Microtúbulos , Estresse Oxidativo/efeitos dos fármacos , Regeneração/efeitos dos fármacos , Traumatismos da Medula Espinal , Animais , Cromonas/farmacologia , Microtúbulos/metabolismo , Microtúbulos/patologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Morfolinas/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Elementos de Resposta , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
8.
Cell ; 179(4): 864-879.e19, 2019 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-31675497

RESUMO

Physical or mental stress leads to neuroplasticity in the brain and increases the risk of depression and anxiety. Stress exposure causes the dysfunction of peripheral T lymphocytes. However, the pathological role and underlying regulatory mechanism of peripheral T lymphocytes in mood disorders have not been well established. Here, we show that the lack of CD4+ T cells protects mice from stress-induced anxiety-like behavior. Physical stress-induced leukotriene B4 triggers severe mitochondrial fission in CD4+ T cells, which further leads to a variety of behavioral abnormalities including anxiety, depression, and social disorders. Metabolomic profiles and single-cell transcriptome reveal that CD4+ T cell-derived xanthine acts on oligodendrocytes in the left amygdala via adenosine receptor A1. Mitochondrial fission promotes the de novo synthesis of purine via interferon regulatory factor 1 accumulation in CD4+ T cells. Our study implicates a critical link between a purine metabolic disorder in CD4+ T cells and stress-driven anxiety-like behavior.


Assuntos
Ansiedade/metabolismo , Comportamento Animal/fisiologia , Encefalopatias Metabólicas/metabolismo , Estresse Psicológico/metabolismo , Tonsila do Cerebelo/metabolismo , Tonsila do Cerebelo/patologia , Animais , Ansiedade/genética , Ansiedade/imunologia , Ansiedade/fisiopatologia , Encefalopatias Metabólicas/genética , Encefalopatias Metabólicas/fisiopatologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/patologia , Modelos Animais de Doenças , Humanos , Camundongos , Dinâmica Mitocondrial/genética , Oligodendroglia/metabolismo , Oligodendroglia/patologia , Análise de Célula Única , Estresse Psicológico/genética , Estresse Psicológico/fisiopatologia , Transcriptoma/genética , Xantina/metabolismo
9.
J Neurotrauma ; 36(24): 3394-3409, 2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31232175

RESUMO

Spinal cord injury (SCI) is a devastating neurological disorder that usually leads to a loss of motor and sensory function in patients. The expression of hypoxia inducible factor-1α (HIF-1α) is increased, and exerts a protective role after traumatic SCI. However, the endogenous activity of HIF-1α is insufficient for promoting functional recovery. The present study tested the potential effect of the sustained activation of HIF-1α by the prolylhydroxylase (PHD) inhibitor dimethyloxalylglycine (DMOG) on anti-apoptotic process and the regulation of axonal regeneration after SCI. Here, we found that treatment with DMOG significantly increased the expression of HIF-1α and that the stabilization of HIF-1α induced by DMOG not only decreased the expression of apoptotic proteins to promote neural survival, but also enhanced axonal regeneration by regulating microtubule stabilization in vivo and in vitro. In addition, we found that DMOG promoted neural survival and axonal regeneration by activating autophagy, which is induced by the HIF-1α/BNIP3 signaling pathway, and that the inhibition of HIF-1α or autophagy abrogated the protective effect of DMOG, as expected. Taken together, our results demonstrate that treatment with DMOG improves functional recovery after SCI and that DMOG may serve as a potential candidate for treating SCI.


Assuntos
Aminoácidos Dicarboxílicos/uso terapêutico , Axônios/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Regeneração Nervosa/fisiologia , Recuperação de Função Fisiológica/fisiologia , Traumatismos da Medula Espinal/metabolismo , Aminoácidos Dicarboxílicos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Axônios/efeitos dos fármacos , Feminino , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Regeneração Nervosa/efeitos dos fármacos , Células PC12 , Estabilidade Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia
10.
J Neurotrauma ; 36(12): 1949-1964, 2019 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-30543130

RESUMO

Axon retraction greatly limits functional recovery after spinal cord injury (SCI) and neuron polarization, which affects processes including axon formation and development, is a promising target for promoting axon regeneration. Increasing microtubule stability has been demonstrated to improve intrinsic axon regeneration processes and is critically related to endoplasmic reticulum (ER)-mitochondria interactions. We used real-time polymerase chain reaction, Western blotting, and immunofluorescence to screen a variety of natural compounds, and found that Loureirin B (LrB) effectively promoted neuron polarization and axon regeneration in vitro and in vivo. LrB significantly inhibited ER stress and thereby promoted mitochondrial functions by regulating mitochondrial fusion. Further, LrB reactivated the Akt/GSK-3ß pathway, which plays critical roles in cell survival and microtubule stabilization. Taken together, our results suggest that the effects of LrB on neuron regeneration involve the inhibition of ER stress-induced mitochondrial dysfunction and activation of the Akt/GSK-3ß pathway, which further promotes microtubule stabilization. LrB may therefore be a promising candidate for facilitating recovery following SCI.


Assuntos
Axônios/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Resinas Vegetais/farmacologia , Traumatismos da Medula Espinal/metabolismo , Animais , Axônios/efeitos dos fármacos , Células Cultivadas , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Mitocôndrias/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Regeneração Nervosa/fisiologia , Gravidez , Ratos , Ratos Sprague-Dawley , Resinas Vegetais/uso terapêutico , Traumatismos da Medula Espinal/tratamento farmacológico
11.
Theranostics ; 8(16): 4429-4446, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30214630

RESUMO

Proper selection and effective delivery of combination drugs targeting multiple pathophysiological pathways key to spinal cord injury (SCI) hold promise to address the thus far scarce clinical therapeutics for improving recovery after SCI. In this study, we aim to develop a clinically feasible way for targeted delivery of multiple drugs with different physiochemical properties to the SCI site, detail the underlying mechanism of neural recovery, and detect any synergistic effect related to combination therapy. Methods: Liposomes (LIP) modified with a scar-targeted tetrapeptide (cysteine-alanine-glutamine-lysine, CAQK) were first constructed to simultaneously encapsulate docetaxel (DTX) and brain-derived neurotrophic factor (BDNF) and then were further added into a thermosensitive heparin-modified poloxamer hydrogel (HP) with affinity-bound acidic fibroblast growth factor (aFGF-HP) for local administration into the SCI site (CAQK-LIP-GFs/DTX@HP) in a rat model. In vivo fluorescence imaging was used to examine the specificity of CAQK-LIP-GFs/DTX binding to the injured site. Multiple comprehensive evaluations including biotin dextran amine anterograde tracing and magnetic resonance imaging were used to detect any synergistic effects and the underlying mechanisms of CAQK-LIP-GFs/DTX@HP both in vivo (rat SCI model) and in vitro (primary neuron). Results: The multiple drugs were effectively delivered to the injured site. The combined application of GFs and DTX supported neuro-regeneration by improving neuronal survival and plasticity, rendering a more permissive extracellular matrix environment with improved regeneration potential. In addition, our combination therapy promoted axonal regeneration via moderation of microtubule function and mitochondrial transport along the regenerating axon. Conclusion: This novel multifunctional therapeutic strategy with a scar-homing delivery system may offer promising translational prospects for the clinical treatment of SCI.


Assuntos
Cicatriz , Portadores de Fármacos/administração & dosagem , Hidrogel de Polietilenoglicol-Dimetacrilato/administração & dosagem , Lipossomos/administração & dosagem , Terapia de Alvo Molecular/métodos , Regeneração , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Fator Neurotrófico Derivado do Encéfalo/administração & dosagem , Modelos Animais de Doenças , Docetaxel/administração & dosagem , Fator 1 de Crescimento de Fibroblastos/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Ratos , Sensibilidade e Especificidade , Resultado do Tratamento
12.
Cell Death Dis ; 9(10): 980, 2018 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-30250268

RESUMO

Intervertebral disc degeneration (IDD) is a complicated pathological condition blamed for low back pain. Mitochondrion is of vital importance for cellular homeostasis, and mitochondrial dysfunction is considered to be one of the major causes of cellular damage. Mitophagy is a cellular process to eliminate impaired mitochondria and showed protective effects in various diseases; however, its role in IDD is still not clear. Here, we explore the role of Parkin-mediated mitophagy in IDD. In this study, we found that Parkin was upregulated in degenerative nucleus pulposus (NP) tissues in vivo as well as in TNF-α stimulated NP cells in vitro. Knockdown of Parkin by siRNA showed that Parkin is crucial for apoptosis and mitochondrion homeostasis in NP cells. Further study showed that upregulation of Parkin by salidroside may eliminate impaired mitochondria and promote the survival of NP cells through activation of mitophagy in vitro. In in vivo study, we found that salidroside could inhibit the apoptosis of NP cells and ameliorate the progression of IDD. These results suggested that Parkin is involved in the pathogenesis of IDD and may be a potential therapeutic target for IDD.


Assuntos
Degeneração do Disco Intervertebral/patologia , Mitofagia/fisiologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Técnicas de Silenciamento de Genes , Glucosídeos/farmacologia , Glucosídeos/uso terapêutico , Humanos , Degeneração do Disco Intervertebral/tratamento farmacológico , Masculino , Mitocôndrias/metabolismo , Mitofagia/efeitos dos fármacos , Núcleo Pulposo/metabolismo , Núcleo Pulposo/patologia , Fenóis/farmacologia , Fenóis/uso terapêutico , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Transfecção , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases/genética , Regulação para Cima
13.
J Cell Mol Med ; 22(5): 2727-2738, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29512938

RESUMO

Fibroblast growth factor 1 (FGF1) is thought to exert protective and regenerative effects on neurons following spinal cord injury (SCI), although the mechanism of these effects is not well understood. The use of FGF1 as a therapeutic agent is limited by its lack of physicochemical stability and its limited capacity to cross the blood-spinal cord barrier. Here, we demonstrated that overexpression of FGF1 in spinal cord following SCI significantly reduced tissue loss, protected neurons in the ventricornu, ameliorated pathological morphology of the lesion, dramatically improved tissue recovery via neuroprotection, and promoted axonal regeneration and remyelination both in vivo and in vivo. In addition, the autophagy and the expression levels of PRDX1 (an antioxidant protein) were induced by AAV-FGF1 in PC12 cells after H2 O2 treatment. Furthermore, the autophagy levels were not changed in PRDX1-suppressing cells that were treated by AAV-FGF1. Taken together, these results suggest that FGF1 improves functional recovery mainly through inducing PRDX1 expression to increase autophagy and anti-ROS activity after SCI.


Assuntos
Autofagia , Fator 1 de Crescimento de Fibroblastos/uso terapêutico , Peroxirredoxinas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Recuperação de Função Fisiológica , Traumatismos da Medula Espinal/tratamento farmacológico , Traumatismos da Medula Espinal/fisiopatologia , Animais , Autofagia/efeitos dos fármacos , Axônios/efeitos dos fármacos , Axônios/metabolismo , Polaridade Celular/efeitos dos fármacos , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Dependovirus/genética , Feminino , Fator 1 de Crescimento de Fibroblastos/farmacologia , Vetores Genéticos/metabolismo , Atividade Motora/efeitos dos fármacos , Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Células PC12 , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Remielinização/efeitos dos fármacos , Traumatismos da Medula Espinal/patologia
14.
Food Funct ; 9(3): 1454-1464, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29473075

RESUMO

Osteoarthritis (OA) is an age-related degenerative disease and is the fourth major cause of disability, but there are no effective therapies because of its complex pathology and the side effects of the drugs. Previous research demonstrated that inflammation and ECM degradation play major roles in OA development. Monascin is an azaphilonoid pigment extracted from Monascus-fermented rice with a potential anti-inflammatory effect reported in various preclinical studies. In the present study, we investigated the protectiveness of monascin on interleukin (IL)-1ß-induced mouse chondrocytes and surgical destabilization of the medial meniscus mouse (DMM) OA models. In vitro, monascin treatment inhibited the IL-1ß-induced expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), nitric oxide (NO), prostaglandin E2 (PGE2), tumor necrosis factor alpha (TNF-α), and interleukin-6 (IL-6). In addition, the IL-1ß-stimulated matrix metalloproteinase-13 (MMP-13) and thrombospondin motifs 5 (ADAMTS-5) upregulation and type two collagen and aggrecan degradation were reversed by monascin. Mechanistically, we revealed that monascin suppressed nuclear factor kappa B (NF-κB) signalling by activating the nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in IL-1ß-induced chondrocytes. And monascin-induced protectiveness in OA development was also shown by using a DMM model. Altogether, our results suggested that monascin could be a novel therapeutic approach for OA.


Assuntos
Condrócitos/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/administração & dosagem , Interleucina-1beta/metabolismo , Osteoartrite/tratamento farmacológico , Animais , Condrócitos/metabolismo , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Interleucina-1beta/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Osteoartrite/genética , Osteoartrite/metabolismo , Transdução de Sinais/efeitos dos fármacos
15.
Mol Med Rep ; 15(6): 4055-4060, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28487971

RESUMO

Esophageal cancer is one of the most common type of malignancies. Telomerase activity, which is absent or weakly detected in the majority of human somatic cells, is elevated in esophageal cancer. Although azidothymidine (AZT), a reverse transcriptase inhibitor, has been utilized as a treatment for tumors, its role in treating esophageal cancer has not been confirmed. The aim of the present study was to determine the effect of AZT on telomerase activity and the proliferation of the human esophageal cancer cell line TE­11. A telomeric repeat amplification assay was utilized to detect telomerase activity following treatment of TE­11 cells with AZT. The effect of AZT on TE­11 cell cycle distribution was determined by flow cytometry. Cellular DNA damage was evaluated by a comet assay and an MTT assay demonstrated that AZT significantly inhibited the viability of TE­11 cells, in a time­and dose­dependent manner. In addition, TE­11 cells treated with various concentrations of AZT exhibited a significant reduction in telomerase activity and percentage of cells in the G1/G0 phase, and an increase in the percentage of cells in the S phase. High doses of AZT caused DNA damage, and enhanced the expression levels of γ­H2A histone family member X and phosphorylated checkpoint kinase 2 in TE­11 cells. These results demonstrated that AZT effectively inhibits proliferation of the TE­11 human esophageal cancer cell line in vitro. The growth inhibitory effects were associated with a reduction in telomerase activity, S and G2/M phase cell cycle arrest, and enhanced DNA damage, suggesting that AZT may be utilized in the clinic for the treatment of esophageal cancer.


Assuntos
Dano ao DNA/efeitos dos fármacos , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Telomerase/metabolismo , Zidovudina/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/efeitos dos fármacos
16.
Oncol Rep ; 34(5): 2259-66, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26323494

RESUMO

The secreted frizzled-related protein 2 (SFRP2) plays a pivotal role in the Wnt pathway, however, it functions as an agonist or an antagonist is still controversial. We profiled SFRP2 expression in several lung cancer cell lines, and found that A549 and 95-D exhibited the lowest and the highest level of SFRP2, respectively. Then we employed the SFRP2-overexpressing plasmid and siRNA to transfect A549 and 95-D cells, respectively. Through MTT assays, we found that SFRP2 knockdown inhibited cell proliferation, and halted the 95-D cells in G1 phase of the cell cycle by downregulation of CCND1 and CDK4, indicating that SFRP2 has the ability of promoting lung cancer cell proliferation. We further checked the cell properties of migration and invasion, using wound scratch assay and Transwell assays. The data showed decreased migrated and invasive 95-D cells after SFRP2 knockdown, and the observations were the opposite in the overexpressing model, implying that SFRP2 promoted lung cancer cell invasion. Moreover, the canonical Wnt pathway was also studied through detection of ß-catenin by western blotting. In the SFRP2 overexpressing model, A549 cells presented stronger expression of ß-catenin compared with controls, while it was the opposite in 95-D cells. These results suggested that SFRP2 serves as a Wnt agonist in lung cancer cells. Together, the findings of this study implied that SFRP2 is not only an agonist of Wnt pathway, but also a cancer promoting protein for lung cancer, indicating SFRP2 as a promising therapeutic target for lung cancer treatment.


Assuntos
Neoplasias Pulmonares/metabolismo , Proteínas de Membrana/fisiologia , Via de Sinalização Wnt , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Expressão Gênica , Humanos , Neoplasias Pulmonares/patologia , Invasividade Neoplásica , Proteínas Wnt/metabolismo
17.
J Cell Mol Med ; 19(9): 2172-80, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26081366

RESUMO

MicroRNAs (miRNAs) play a pivotal role in carcinogenesis. Dysregulation of miRNAs, both oncogenic miRNAs and tumour-suppressive miRNAs, is closely associated with cancer development and progression. The levels of miRNAs could be changed epigenetically by DNA methylation in the 5' untranslated region (UTR) of pre-mature miRNAs. To investigate whether DNA methylation alters the expression of miR-129 in lung cancer, we did DNA methylation assays and found that 5' UTR region of miR-129-2 gene was absolutely methylated in both A549 and SPCA-1 lung cancer cells, but totally un-methylated in 95-D cells. The expression of miR-129 was restored by 5-Aza-2'-deoxycytidine (DAC), a de-methylation agent, in both A549 and SPCA-1 cells, resulting in attenuated cell migration and invasion ability, and decreased protein level of NF-κB, which indicates the involvement of NF-κB pathway. To further illustrate the roles of miR-129 in lung tumourigenesis, we overexpressed miR-129 in lung cancer cells by transfection of miR-129 mimics, and found arrested cell proliferation at G2/M phase of cell cycle and inhibited cell invasion. These findings strongly suggest that miR-129 is a tumour suppressive miRNA, playing important roles in the development and progression of human lung cancer.


Assuntos
Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , MicroRNAs/genética , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Sequência de Bases , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Metilação de DNA/genética , Regulação para Baixo/genética , Perfilação da Expressão Gênica , Células HEK293 , Humanos , MicroRNAs/metabolismo , Dados de Sequência Molecular , Invasividade Neoplásica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína com Valosina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA