Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 144
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 38(14): e23804, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39037422

RESUMO

Natural Nicotinamide Adenine Dinucleotide (NAD+) precursors have attracted much attention due to their positive effects in promoting ovarian health. However, their target tissue, synthesis efficiency, advantages, and disadvantages are still unclear. This review summarizes the distribution of NAD+ at the tissue, cellular and subcellular levels, discusses its biosynthetic pathways and the latest findings in ovary, include: (1) NAD+ plays distinct roles both intracellularly and extracellularly, adapting its distribution in response to requirements. (2) Different precursors differs in target tissues, synthetic efficiency, biological utilization, and adverse effects. Importantly: tryptophan is primarily utilized in the liver and kidneys, posing metabolic risks in excess; nicotinamide (NAM) is indispensable for maintaining NAD+ levels; nicotinic acid (NA) constructs a crucial bridge between intestinal microbiota and the host with diverse functions; nicotinamide riboside (NR) and nicotinamide mononucleotide (NMN) increase NAD+ systemically and can be influenced by delivery route, tissue specificity, and transport efficiency. (3) The biosynthetic pathways of NAD+ are intricately intertwined. They provide multiple sources and techniques for NAD+ synthesis, thereby reducing the dependence on a single molecule to maintain cellular NAD+ levels. However, an excess of a specific precursor potentially influencing other pathways. In addition, Protein expression analysis suggest that ovarian tissues may preferentially utilize NAM and NMN. These findings summarize the specific roles and potential of NAD+ precursors in enhancing ovarian health. Future research should delve into the molecular mechanisms and intervention strategies of different precursors, aiming to achieve personalized prevention or treatment of ovarian diseases, and reveal their clinical application value.


Assuntos
NAD , Niacinamida , Ovário , Humanos , NAD/metabolismo , NAD/biossíntese , Ovário/metabolismo , Feminino , Animais , Niacinamida/metabolismo , Niacinamida/biossíntese , Vias Biossintéticas , Mononucleotídeo de Nicotinamida/metabolismo
2.
Biol Proced Online ; 26(1): 1, 2024 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-38178023

RESUMO

BACKGROUND: Gastric cancer (GC) is a common malignancy and a leading cause of cancer-related death with high morbidity and mortality. Methyl-CpG binding domain protein 3 (MBD3), a key epigenetic regulator, is abnormally expressed in several cancers, participating in progression and metastasis. However, the role of MBD3 in GC remains unknown. METHODS: MBD3 expression was assessed via public databases and validated by western blotting and quantitative real-time polymerase chain reaction (qRT-PCR). The prognosis of MBD3 was analysed via bioinformatics based on the TCGA dataset. The migration, invasion and proliferation of GC cells were examined by transwell, wound healing, cell counting kit (CCK)-8, colony-formation and xenograft mouse models. Epithelial-mesenchymal transition (EMT) and phosphatidylinositide 3-kinases/ protein Kinase B (PI3K/AKT) pathway markers were evaluated by Western blotting. RNA sequencing was used to identify the target of MBD3. RESULTS: MBD3 expression was higher in GC tissues and cells than in normal tissues and cells. Additionally, high MBD3 levels were associated with poor prognosis in GC patients. Subsequently, we proved that MBD3 enhanced the migration, invasion and proliferation abilities of GC cells. Moreover, western blot results showed that MBD3 promoted EMT and activated the PI3K/AKT pathway. RNA sequencing analysis showed that MBD3 may increase actin γ1 (ACTG1) expression to promote migration and proliferation in GC cells. CONCLUSION: MBD3 promoted migration, invasion, proliferation and EMT by upregulating ACTG1 via PI3K/AKT signaling activation in GC cells and may be a potential diagnostic and prognostic target.

3.
Biochem Biophys Res Commun ; 648: 11-20, 2023 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-36724555

RESUMO

Non-small cell lung cancer (NSCLC) is a major global health threat with high incidence and mortality. Modulator of apoptosis-1 (MOAP1), also named MAP-1, belongs to the PNMA gene family and plays a key role in regulating apoptosis and tumor growth. However, its influences on NSCLC are largely unclear, and thus were explored in our present study, particularly the underlying mechanisms. Here, we initially find that MOAP1 expression is significantly decreased in NSCLC patients compared with the normal ones, and negatively correlated with the TNM and pathologic stages among patients. Additionally, MOAP1 low expression predicts a poorer prognosis than that of the NSCLC patients expressing higher MOAP1. Our in vitro studies confirm much lower MOAP1 expression in NSCLC cell lines. Of note, promoting MOAP1 expression strongly reduces the proliferation and induces apoptosis in NSCLC cells, accompanied with cell cycle arrest distributed in G0/G1 phase. Moreover, we find that MOAP1 has a negative correlation with Th2 cells' infiltration, but a positive correlation with the infiltration levels of eosinophils. Epithelial mesenchymal transition (EMT) process is also greatly restrained in NSCLC cells with MOAP1 over-expression, as proved by the reduced migration and invasion of cells. We further identify a positive correlation between MOAP1 and tripartite motif-containing 68 (TRIM68) in patients with NSCLC. Further analysis shows that TRIM68 directly interacts with MOAP1 and stabilizes MOAP1. Importantly, TRIM68 can activate MOAP1 by inducing the K63-linked polyubiquitination of MOAP1. Finally, animal studies verify that promoting MOAP1 efficiently suppresses tumor growth and lung metastasis in the nude mice. Collectively, our results reveal a novel mechanism through which MOAP1 stabilized by TRIM68 inhibits NSCLC development and targeting MOAP1 for its up-regulation may be a promising therapeutic strategy for NSCLC treatment.


Assuntos
Proteínas Reguladoras de Apoptose , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Proteínas com Motivo Tripartido , Animais , Camundongos , Apoptose , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/patologia , Camundongos Nus , Humanos , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas com Motivo Tripartido/metabolismo
4.
Arch Biochem Biophys ; 748: 109783, 2023 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-37816421

RESUMO

PURPOSE: Long non-coding RNA urothelial cancer associated 1 (UCA1) serves as an oncogene in various cancers. However, the mechanism underlying the role of UCA1 in pancreatic cancer remains unclear. This study aimed to explore the role of UCA1 in pancreatic cancer. METHODS: The expression and prognosis of UCA1 were analyzed using The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) databases. The results were validated by immunohistochemistry (IHC) and qRT-PCR. The biofunctions of UCA1 were analyzed using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA). The migration abilities and mitochondrial dynamics of PC cells were examined using the Transwell assay, mitochondrial membrane potential (MMP), and fluorescence. The mitochondrial-related protein and MAPK/ERK pathway markers were evaluated using western blotting. RESULTS: UCA1 expression was significantly higher in pancreatic cancer tissues than in normal tissues. High UCA1 expression indicated poor clinical outcomes and was associated with clinical features in patients with pancreatic cancer. Additionally, high UCA1 expression is a potential independent marker for poor prognosis. Subsequently, we demonstrated that UCA1 enhanced the migration capability, increased MMP, enhanced mitochondrial fusion, and inhibited mitochondrial autophagy in pancreatic cancer cells via the MAPK/ERK pathway. CONCLUSION: UCA1 promotes the migration by regulating the mitochondrial dynamics of pancreatic cancer cells via the MAPK/ERK pathway. Our findings suggest that UCA1 may serve as a potential biomarker in pancreatic cancer prognosis.


Assuntos
MicroRNAs , Neoplasias Pancreáticas , RNA Longo não Codificante , Neoplasias da Bexiga Urinária , Humanos , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Dinâmica Mitocondrial , Neoplasias da Bexiga Urinária/genética , Movimento Celular , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Neoplasias Pancreáticas
5.
Exp Cell Res ; 411(1): 112937, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34863709

RESUMO

Spinal cord injury (SCI) often causes severe neurological dysfunction, and facilitating neurite elongation is particularly important in its treatment. Astrocytes (AS) play an important role in the central nervous system (CNS), and their high plasticity and versatility provide a feasible entry point for relevant research. Our purpose was to explore whether extracellular vesicles (EVs) from astrocytes (AS-EVs) and lipopolysaccharide (LPS)-preactivated astrocytes (LPAS-EVs) facilitate neurite elongation, to explore the underlying mechanism, and to verify whether these EVs promote locomotor recovery in rats. We used LPS to preactivate astrocytes and cocultured them with PC12 cells to observe neurite changes, then extracted and identified AS-EVs and LPAS-EVs and the role and mechanism of these EVs in facilitating neurite elongation was examined in vivo and vitro. We demonstrated that AS-EVs and LPAS-EVs facilitated the elongation of neurites and the recovery of rats with SCI. LPAS-EVs had a stronger effect than AS-EVs, by activating the Hippo pathway, promoting monopole spindle binding protein 1 (MOB1) expression, and reducing Yes-associated protein (YAP) levels. The data also suggest a feedback regulation between MOB1 and p-YAP/YAP. In sum, AS-EVs and LPAS-EVs can play an active role in facilitating neurite elongation by activating the Hippo pathway. These findings provide a new strategy for treating SCI and other CNS-related injuries.


Assuntos
Astrócitos/citologia , Vesículas Extracelulares/transplante , Via de Sinalização Hippo , Neuritos/fisiologia , Neurônios/citologia , Traumatismos da Medula Espinal/terapia , Animais , Astrócitos/metabolismo , Vesículas Extracelulares/metabolismo , Células PC12 , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/patologia
6.
Anal Chem ; 94(6): 2803-2811, 2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35104110

RESUMO

Fluorescence imaging the interplay between lipid droplets (LDs) and protein aggregates (PAs) is extremely valuable for elucidating molecular mechanisms of aging. Here, we describe the first dual-functional fluorescent probe, LW-1, for simultaneously imaging LDs and PAs in distinct fluorescence channels to dissect interplaying roles between LDs and PAs during aging. Notably, based on an intriguing mechanism of hydrogen bonds regulating single bond rotation, LW-1 selectively detected LDs in a red channel. Meanwhile, based on another mechanism of the hydrogen bond regulating intramolecular charge transfer efficiency, probe LW-1 further detected PAs in an NIR channel. Practical applications showed that LW-1 was capable of concurrently detecting LDs and PAs in living cells. Moreover, simultaneously imaging LDs and PAs in intestine tissues of mice at different aging degrees was conducted. The results denoted that the PAs level in the intestine tissue increased dramatically with aging, accompanying the buildup of LDs. Significantly, the interplay between LDs and PAs during aging was observed. These evidences demonstrated that the PAs level was closely related with aging processes in intestine tissues, while LDs were formed correspondingly to interact with PAs, suggesting that excessive PAs can be loaded into LDs and then be removed by lipophagy.


Assuntos
Corantes Fluorescentes , Gotículas Lipídicas , Envelhecimento , Animais , Corantes Fluorescentes/química , Gotículas Lipídicas/química , Camundongos , Imagem Óptica/métodos , Agregados Proteicos
7.
Exp Cell Res ; 393(1): 112091, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32422133

RESUMO

Methyl-CpG-binding domain 3 (MBD3), as an induced stem cells reprogramming barrier, has an abnormal expression in various prevalent malignancies. However, in pancreatic cancer cell stemness, the roles of MBD3 remain unclear. In our study, the effects of MBD3 were investigated on the proliferation, stemness and the underlying mechanism in pancreatic cancer cells. Firstly, MBD3 knockdown was proved to promote proliferation and sphere formation of pancreatic cancer cells and tumorigenesis, while MBD3 upregulation inhibited the above results. Also, MBD3 downregulation notably increased stemness markers level of OCT4, NANOG and SOX2, and MBD3 upregulation resulted in the opposite effects. Mechanically, it was found that MBD3 involved in activation of Hippo pathway. There was a negative correlation between MBD3 and YAP expression in TCGA database. MBD3 knockdown improved YAP expression, and promoted YAP nuclear translocation increased TEAD luciferase activity, while MBD3 overexpression reversed the above results. Further evidence revealed that YAP could bind to MBD3, and decreased MBD3 expression. Collectively, MBD3 bound to YAP to significantly inhibit proliferation and weaken stemness maintenance in pancreatic cancer cells, as well as reduce tumorigenesis via Hippo signaling. Thus, MBD3 may serve as a potential molecular biomarker for exploring new therapeutic strategies to treat pancreatic cancer.


Assuntos
Proteínas de Ligação a DNA/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Via de Sinalização Hippo , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Neoplasias Pancreáticas
8.
J Nanobiotechnology ; 19(1): 142, 2021 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-34001148

RESUMO

BACKGROUND: Therapeutic tumor vaccine (TTV) that induces tumor-specific immunity has enormous potentials in tumor treatment, but high heterogeneity and poor immunogenicity of tumor seriously impair its clinical efficacy. Herein, a novel NIR responsive tumor vaccine in situ (HA-PDA@IQ/DOX HG) was prepared by integrating hyaluronic acid functionalized polydopamine nanoparticles (HA-PDA NPs) with immune adjuvants (Imiquimod, IQ) and doxorubicin (DOX) into thermal-sensitive hydrogel. RESULTS: HA-PDA@IQ NPs with high photothermal conversion efficiency (41.2%) and T1-relaxation efficiency were using HA as stabilizer by the one-pot oxidative polymerization. Then, HA-PDA@IQ loaded DOX via π-π stacking and mixed with thermal-sensitive hydrogel to form the HA-PDA@IQ/DOX HG. The hydrogel-confined delivery mode endowed HA-PDA@IQ/DOX NPs with multiple photothermal ablation performance once injection upon NIR irradiation due to the prolonged retention in tumor site. More importantly, this mode enabled HA-PDA@IQ/DOX NPs to promote the DC maturation, memory T cells in lymphatic node as well as cytotoxic T lymphocytes in spleen. CONCLUSION: Taken together, the HA-PDA@IQ/DOX HG could be served as a theranostic tumor vaccine for complete photothermal ablation to trigger robust antitumor immune responses.


Assuntos
Vacinas Anticâncer/uso terapêutico , Tratamento Farmacológico/métodos , Imunidade/efeitos dos fármacos , Imunidade/efeitos da radiação , Terapia Fototérmica/métodos , Doxorrubicina/administração & dosagem , Ácido Hialurônico , Hidrogéis , Indóis/química , Nanopartículas/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Polímeros/administração & dosagem , Polímeros/química
9.
BMC Oral Health ; 21(1): 555, 2021 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-34717590

RESUMO

BACKGROUND: Nattokinase (NK) is a promising alternative in the prevention and treatment of cardiovascular diseases due to its potent fibrinolytic activity. In this study, we investigated the effect of crude nattokinase extract on the healing of acetic acid-induced oral mucositis in mice. METHODS: Bacillus subtilis culture media (BSCM) was isolated into the supernatant, named nattokinase crude extract (NCE), and the pellet was named Bacillus subtilis mass (BSM). An oral mucositis model was established in mice by applying 50% glacial acetic acid to the buccal mucosa. According to the treatment conditions, the mice were divided into BSCM, NCE, BSM and phosphate buffered saline (PBS) groups. The weight of the mice, oral mucositis healing score and histopathological examination were used to evaluate the treatment. RESULTS: Fibrinolytic activities of BSCM, NCE and BSM were approximately 8069, 10,800 and 80 U/ml, respectively. The weight gain of mice in the NCE group was significantly different from the PBS group after three days' treatment (p < 0.05). The oral mucositis score of NCE group was significantly higher than other groups (p < 0.05). The differences in histopathology scores between the NCE and other groups were statistically significant (p < 0.01). CONCLUSIONS: NCE could possess remarkable potential to reduce pain and promote oral mucositis healing with minimal safety concerns. In this study, we first report that NCE from the supernatant of Bacillus subtilis can promote the healing of oral mucositis, which extends the application scope of NK.


Assuntos
Estomatite , Subtilisinas , Animais , Misturas Complexas , Fermentação , Camundongos , Estomatite/induzido quimicamente , Estomatite/tratamento farmacológico , Subtilisinas/metabolismo
10.
J Cell Mol Med ; 24(2): 1969-1979, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31851779

RESUMO

Protein arginine methyltransferase 5 (PRMT5) has been implicated in the development and progression of human cancers. However, few studies reveal its role in epithelial-mesenchymal transition (EMT) of pancreatic cancer cells. In this study, we find that PRMT5 is up-regulated in pancreatic cancer, and promotes proliferation, migration and invasion in pancreatic cancer cells, and promotes tumorigenesis. Silencing PRMT5 induces epithelial marker E-cadherin expression and down-regulates expression of mesenchymal markers including Vimentin, collagen I and ß-catenin in PaTu8988 and SW1990 cells, whereas ectopic PRMT5 re-expression partially reverses these changes, indicating that PRMT5 promotes EMT in pancreatic cancer. More importantly, we find that PRMT5 knockdown decreases the phosphorylation level of EGFR at Y1068 and Y1172 and its downstream p-AKT and p-GSK3ß, and then results in down-regulation of ß-catenin. Expectedly, ectopic PRMT5 re-expression also reverses the above changes. It is suggested that PRMT5 promotes EMT probably via EGFR/AKT/ß-catenin pathway. Taken together, our study demonstrates that PRMT5 plays oncogenic roles in the growth of pancreatic cancer cell and provides a potential candidate for pancreatic cancer treatment.


Assuntos
Transição Epitelial-Mesenquimal , Receptores ErbB/metabolismo , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/patologia , Proteína-Arginina N-Metiltransferases/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Animais , Carcinogênese/genética , Carcinogênese/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Proteína-Arginina N-Metiltransferases/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Pancreáticas
11.
J Cell Biochem ; 121(1): 632-641, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31452251

RESUMO

Large intergenic noncoding RNA regulator of reprogramming (Linc-RoR) was first identified as a regulator to increase the emergence of induced pluripotent stem cells through reprogramming differentiated cells and is abnormal expression in a variety of malignant tumors. However, the function of Linc-RoR in pancreatic cancer progression needs further clarification. The data from this study demonstrated that Linc-RoR knockdown suppressed cell proliferative capacity and colony formation, while Linc-RoR overexpression promoted these behaviors. In particular, Linc-RoR overexpression promoted the level of mesenchymal markers, inhibited the expression of epithelial markers, as well as enhanced pancreatic cancer cells migration and invasion, whereas Linc-RoR knockdown inhibited the expression of mesenchymal markers, promoted the expression of epithelial markers, as well as weakened pancreatic cancer cells migration and invasion. Further study revealed that Linc-RoR knockdown brought about a significant fall in YAP phosphorylation and a rise in total YAP, while Linc-RoR overexpression produced the opposite results. Specifically, Linc-RoR promoted YAP in the cytoplasm into the nucleus. Taken together, we conjectured that Linc-RoR promoted proliferation, migration, and invasion of pancreatic cancer cells by activating the Hippo/YAP pathway. YAP might be an underlying target of Linc-RoR and mediate epithelial-mesenchymal transition (EMT) in pancreatic cancer (PC); thus, Linc-RoR might be a very meaningful biomarker for PC.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , RNA Longo não Codificante/genética , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Via de Sinalização Hippo , Humanos , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Fatores de Transcrição/genética , Células Tumorais Cultivadas , Proteínas de Sinalização YAP
12.
Int J Cancer ; 146(10): 2901-2912, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31633800

RESUMO

Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer mortality worldwide. Emerging evidence indicates that tumour cells release substantial amounts of RNA into the bloodstream, in which RNA strongly resists RNases and is present at sufficient levels for quantitative analyses. Our study aimed to discover blood-based markers for the early detection of CRC and to ascertain their efficiency in discriminating healthy controls, patients with polyps and adenomas and cancer patients. We first analysed and screened ZFAS1, SNHG11, LINC00909 and LINC00654 in a bioinformatics database and then collected clinical plasma samples for preliminary small-scale analysis and further large-scale verification. We then explored the mechanism of dominant lncRNA SNHG11 expression in CRC by in vitro and in vivo assays. The combination of ZFAS1, SNHG11, LINC00909 and LINC00654 showed high diagnostic performance for CRC (AUC: 0.937), especially early-stage disease (AUC: 0.935). Plasma levels of the four candidate lncRNAs were significantly reduced in postoperative samples compared to preoperative samples. A panel including these four lncRNAs performed well in distinguishing patient groups with different stages of colon disease, and SNHG11 exhibited the greatest diagnostic ability to identify precancerous lesions and early-stage tumour formation. Mechanistically, high SNHG11 expression promotes proliferation and metastasis by targeting the Hippo pathway. Taken together, the data indicate that SNHG11 may be a novel therapeutic target for the treatment of CRC and a potential biomarker for the early detection of CRC.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Colorretais/sangue , Neoplasias Colorretais/diagnóstico , Detecção Precoce de Câncer/métodos , RNA Longo não Codificante/sangue , Adenocarcinoma/sangue , Adenocarcinoma/diagnóstico , Adenocarcinoma/patologia , Adenoma/sangue , Adenoma/diagnóstico , Adenoma/patologia , Animais , Neoplasias Colorretais/patologia , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Prognóstico
13.
EMBO J ; 35(6): 668-84, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26912724

RESUMO

A key step of Wnt signaling activation is the recruitment of ß-catenin to the Wnt target-gene promoter in the nucleus, but its mechanisms are largely unknown. Here, we identified FoxM1 as a novel target of Wnt signaling, which is essential for ß-catenin/TCF4 transactivation. GSK3 phosphorylates FoxM1 on serine 474 which induces FoxM1 ubiquitination mediated by FBXW7. Wnt signaling activation inhibits FoxM1 phosphorylation by GSK3-Axin complex and leads to interaction between FoxM1 and deubiquitinating enzyme USP5, thereby deubiquitination and stabilization of FoxM1. FoxM1 accumulation in the nucleus promotes recruitment of ß-catenin to Wnt target-gene promoter and activates the Wnt signaling pathway by protecting the ß-catenin/TCF4 complex from ICAT inhibition. Subsequently, the USP5-FoxM1 axis abolishes the inhibitory effect of ICAT and is required for Wnt-mediated tumor cell proliferation. Therefore, Wnt-induced deubiquitination of FoxM1 represents a novel and critical mechanism for controlling canonical Wnt signaling and cell proliferation.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Linhagem Celular , Endopeptidases/metabolismo , Proteína Forkhead Box M1 , Humanos , Ativação Transcricional , Ubiquitinação , Via de Sinalização Wnt
14.
Anal Chem ; 92(8): 6072-6080, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32216261

RESUMO

Hypochlorite (ClO-) and singlet oxygen (1O2) commonly coexist in living systems and exert important interplaying roles in many diseases. To dissect their complex inter-relationship, it is urgently required to construct a fluorescent probe that can discriminate ClO- and 1O2 in living organisms. Herein, by taking the 3-(aliphaticthio)-propan-1-one group as the unique recognition unit for both ClO- and 1O2, we proposed the first fluorescent probe, Hy-2, to simultaneously discriminate ClO- and 1O2 with high sensitivity and selectivity. Probe Hy-2 itself showed fluorescence in blue channel. After treatment with ClO- and 1O2, respectively, pronounced fluorescence enhancements were observed in the green channel and red channel correspondingly. Moreover, upon development of the probe with aggregation-induced emission (AIE) characteristics, the probe could work well in a solution with high water volume fraction. Probe Hy-2 was also able to accumulate into mitochondria and was utilized as an effective tool to image exogenous and endogenous ClO- and 1O2 in mitochondria. Significantly, as the first trial, probe Hy-2 was employed to simultaneously monitor the variation of ClO- and 1O2 level in cecal tissues of rat in the cecal ligation and puncture (CLP)-induced polymicrobial sepsis model. The results demonstrated that the expressed ClO- and 1O2 levels were tightly correlated with the severity of sepsis, inferring that the overproduction of ClO- and 1O2 is an important factor in the pathogenesis of sepsis. The probe illustrated herein may provide a guide for further exploring the functions of ClO- and 1O2 in various diseases.


Assuntos
Corantes Fluorescentes/química , Ácido Hipocloroso/análise , Imagem Óptica , Oxigênio/análise , Sepse/diagnóstico por imagem , Animais , Modelos Animais de Doenças , Corantes Fluorescentes/síntese química , Ratos , Ratos Sprague-Dawley
15.
Nanotechnology ; 31(25): 255601, 2020 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-32126545

RESUMO

To enhance the therapeutic efficiency and reduce side effects from drug delivery and chemotherapy, image-guided nanoscale systems have attracted tremendous attention in recent decades. In this study, we developed a novel method to fabricate a colchicine/gadolinium-loaded tubulin self-assembly nanocarrier (Col-Gd@Tub NC) for the image-guided chemotherapy of glioma. The Col-Gd@Tub NCs were spontaneously formed via tubulin self-assembly and were subsequently functionalized by colchicine and gadolinium elements. These resultant Col-Gd@Tub NCs with a diameter of 45 nm exhibited uniform particle size distribution and favorable stability without any leakage of gadolinium in water. Meanwhile, the introduction of gadolinium endowed Col-Gd@Tub NCs with high T 1-weighted MRI performance in vitro. After tail vein injection, Col-Gd@Tub NCs exhibited excellent MRI contrast capability and relatively long circulation time (∼12 h) and were finally cleared out from the bladder. More significantly, the binding colchicine still exerted an anti-tumor effect after the Col-Gd@Tub NCs were taken up by the tumor cells. These results show that the Col-Gd@Tub NCs may be served as a versatile nanoscale platform for the integration of biomedical imaging probes and therapeutic molecules for tumor therapy.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Colchicina/administração & dosagem , Gadolínio/química , Glioma/tratamento farmacológico , Tubulina (Proteína)/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias Encefálicas/diagnóstico por imagem , Linhagem Celular Tumoral , Colchicina/química , Colchicina/farmacologia , Difusão Dinâmica da Luz , Feminino , Glioma/diagnóstico por imagem , Células HEK293 , Humanos , Imageamento por Ressonância Magnética , Nanopartículas Metálicas , Camundongos , Microscopia Eletrônica de Transmissão , Tamanho da Partícula , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Nanotechnology ; 31(16): 165101, 2020 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-31766034

RESUMO

The antioxidant activity of ceria nanoparticles is tightly regulated by size distribution and heteroatom doping. Inspired by this rule, cerium and praseodymium codoped carbon quantum dots (Ce/Pr-CQDs) were synthesized through the one-pot hydrothermal carbonization method. Taking intrinsic advantage of CQDs, the resultant Ce/Pr-CQDs exhibited uniform and ultra-small morphology with an average size of 2.8 nm, which led to an increased proportion of Ce3+. In addition, the doping of Pr into Ce-CQDs improved the redox properties. As we expected, the Ce/Pr-CQDs possessed enhanced hydroxyl radical scavenging properties compared with the cerium-doped carbon quantum dots (Ce-CQDs). Furthermore, Ce/Pr-CQDs with favorable biocompatibility and negligible cytotoxicity are readily internalized into cytoplasm, decreasing the level of reactive oxygen species (ROS). Taken together, the resultant Ce/Pr-CQDs displayed great potential for applications relating to oxidative-stress-associated disease.


Assuntos
Antioxidantes/farmacologia , Carbono/química , Cério/química , Sequestradores de Radicais Livres/farmacologia , Radical Hidroxila/química , Praseodímio/química , Pontos Quânticos/química , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular , Camundongos , Oxirredução , Pontos Quânticos/ultraestrutura , Difração de Raios X
18.
Nanotechnology ; 30(32): 325101, 2019 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-30909174

RESUMO

Excessive reactive oxygen species (ROS) can lead to irreversible damage to the human body in vivo, therefore it is highly desirable to exploit an efficient antioxidant. Recently, cerium oxide nanoparticles have attracted extensive attention in the field of biomedicine due to their excellent antioxidant properties. In this study, cerium-doped carbon quantum dots (Ce-doped CQDs) with hydroxyl radical scavenging capacity were synthesized for first time by one-step hydrothermal carbonization method. The resultant Ce-doped CQDs with the average particle size of 2.5 nm possessed the properties of good water solubility, colloid stability, and strong fluorescence, which are similar to traditional CQDs. Meanwhile, the Ce-doped CQDs had good biocompatibility and negligible cytotoxicity. Taking advantage of inherent ultra-small size, the Ce-doped CQDs exhibited a high Ce3+/Ce4+ ratio at the surface of particles. The radical scavenging capability of the Ce-doped CQDs was proved by a simple photometric system in vitro, which provided direct evidence for its antioxidant potency. Furthermore, the Ce-doped CQDs had a high ability to protect cells from hydrogen peroxide-induced damage by scavenging hydroxyl radicals. These results suggest that Ce-doped CQDs as a new ROS scavenger may provide potential prospects for the treatment of oxidative stress-related diseases.

19.
Exp Cell Res ; 363(2): 179-187, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29305963

RESUMO

The failure of neuronal proliferation and differentiation is a major obstacle for neural repair and regeneration after traumatic central nervous system (CNS) injury. PTEN acts as an intrinsic brake on the neuronal cells, but its roles and mechanism still remain to be clarified. Herein, for the first time we confirmed that PTEN had a dual effect on the neuronal cells in vitro. Firstly, we found that PTEN knockdown significantly promoted cell proliferation and differentiation. Then, PTEN knockdown activated PI3K/Akt and Wnt/ß-catenin pathways in vitro. Further evidence revealed that GSK3ß as a key node involved in PTEN controlling cell proliferation and differentiation in PC12 cells. In addition, we identified that PTEN-GSK3ß pathway modulated neuronal proliferation via ß-catenin. Taken together, these results suggest that PTEN silencing enhances neuronal proliferation and differentiation by activating PI3K/Akt/GSK3ß pathway that it may be a promising therapeutic approach for CNS injury.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Animais , Inativação Gênica/fisiologia , Neurônios/metabolismo , Células PC12 , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
20.
Exp Cell Res ; 362(1): 43-50, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29097184

RESUMO

Protein arginine methyltransferase 1 (PRMT1) is up-regulated and promotes migration, invasion and proliferation in wide range of cancers. However, we for the first time identify that PRMT1 promotes migration and invasion and inhibits proliferation in gastric cancer cells, a phenomenon called "migration-proliferation dichotomy". First, we find that PRMT1 overexpression promotes migration and invasion and inhibits proliferation, whereas PRMT1 knockdown reverses the above abilities. Next, PRMT1 reduces the expression of epithelial marker E-cadherin and increases the expression of mesenchymal markers including N-cadherin, Vimentin, snail and ß-catenin in gastric cancer cells. Furthermore, our studies show that PRMT1 silencing promotes the phosphorylation of LATS1, and then induces YAP phosphorylation, while overexpression of PRMT1 down-regulates the phosphorylation of LATS1 and YAP, indicating that PRMT1 inhibits EMT probably via Hippo signaling. Collectively, the present study reveals important roles of PRMT1 in progression of gastric cancer. Given the dual functions of PRMT1, it is as a potential drug target of gastric cancer with extreme caution.


Assuntos
Proliferação de Células/genética , Transição Epitelial-Mesenquimal/genética , Proteína-Arginina N-Metiltransferases/fisiologia , Proteínas Repressoras/fisiologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Movimento Celular/genética , Progressão da Doença , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA