Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Pharmacol Res ; 188: 106676, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36693599

RESUMEN

Age-related gastrointestinal decline contributes to whole-organism frailty and mortality. Genistein is known to have beneficial effects on age-related diseases, but its precise role in homeostasis of the aging gut remains to be elucidated. Here, wild-type aging mice and Zmpste24-/- progeroid mice were used to investigate the role of genistein in lifespan and homeostasis of the aging gut in mammals. A series of longitudinal, clinically relevant measurements were performed to evaluate the effect of genistein on healthspan. It was found that dietary genistein promoted a healthier and longer life and was associated with a decrease in the levels of systemic inflammatory cytokines in aging mice. Furthermore, dietary genistein ameliorated gut dysfunctions, such as intestinal inflammation, leaky gut, and impaired epithelial regeneration. A distinct genistein-mediated alteration in gut microbiota was observed by increasing Lachnospira abundance and short-chain fatty acid (SCFA) production. Further fecal microbiota transplantation and dirty cage sharing experiments indicated that the gut microbiota from genistein-fed mice rejuvenated the aging gut and extended the lifespan of progeroid mice. It was demonstrated that genistein-associated SCFAs alleviated tumor necrosis factor alpha-induced intestinal organoid damage. Moreover, genistein-associated propionate promoted regulatory T cell-derived interleukin 10 production, which alleviated macrophage-derived inflammation. This study provided the first data, to the authors' knowledge, indicating that dietary genistein modulates homeostasis in the aging gut and extends the healthspan and lifespan of aging mammals. Moreover, the existence of a link between genistein and the gut microbiota provides a rationale for dietary interventions against age-associated frailty.


Asunto(s)
Fragilidad , Microbioma Gastrointestinal , Ratones , Animales , Longevidad , Genisteína/farmacología , Ácidos Grasos Volátiles/farmacología , Envejecimiento , Inflamación , Homeostasis , Ratones Endogámicos C57BL , Mamíferos
2.
J Proteome Res ; 21(8): 1876-1893, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35786973

RESUMEN

Gastrointestinal (GI) cancers constitute the largest portion of all human cancers, and the most prevalent GI cancers in China are colorectal cancer (CRC), gastric cancer (GC), and hepatocellular carcinoma (HCC). Exosomes are nanosized vesicles containing proteins, lipids, glycans, and nucleic acid, which play important roles in the tumor microenvironment and progression. Aberrant glycosylation is closely associated with GI cancers; however, little is known about the glycopattern of the exosomes from GI cancer cells. In this study, glycopatterns of HCC, CRC, and GC cell lines and their exosomes were detected using lectin microarrays. For all exosomes, (GlcNAcß1-4)n and Galß1-4GlcNAc (DSA) were the most abundant glycans, but αGalNAc and αGal (GSL-II and SBA) were the least. Different cancers had various characteristic glycans in either cells or exosomes. Glycans altered in cell-derived exosomes were not always consistent with the host cells in the same cancer. However, Fucα1-6GlcNAc (core fucose) and Fucα1-3(Galß1-4)GlcNAc (AAL) were altered consistently in cells and exosomes although they were decreased in HCC and CRC but increased in GC. The study drew the full-scale glycan fingerprint of cells and exosomes related to GI cancer, which may provide useful information for finding specific biomarkers and exploring the underlying mechanism of glycosylation in exosomes.


Asunto(s)
Carcinoma Hepatocelular , Exosomas , Neoplasias Gastrointestinales , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Línea Celular , Exosomas/metabolismo , Neoplasias Gastrointestinales/metabolismo , Glicoproteínas/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Polisacáridos/metabolismo , Microambiente Tumoral
3.
Mol Med ; 27(1): 95, 2021 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-34470609

RESUMEN

BACKGROUND: Long noncoding RNAs (lncRNAs), a type of pervasive genes that regulates various biological processes, are differentially expressed in different types of malignant tumors. The role of lncRNAs in the carcinogenesis of pancreatic ductal adenocarcinoma (PDAC) remains unclear. Here, we investigated the role of the lncRNA DKFZp434J0226 in PDAC. METHODS: Aberrantly expressed mRNAs and lncRNAs among six PDAC and paired non-tumorous tissues were profiled using microarray analysis. Quantitative real-time polymerase chain reaction was used to evaluate DKFZp434J0226 expression in PDAC tissues. CCK-8 assay, wound-healing assay, soft agar colony formation assay, and transwell assay were performed to assess the invasiveness and proliferation of PDAC cells. Furthermore, RNA pull-down, immunofluorescence, RNA immunoprecipitation, and western blotting assays were performed to investigate the association between DKFZp434J0226 and SF3B6. Tumor xenografts in mice were used to test for tumor formation in vivo. RESULTS: In our study, 222 mRNAs and 128 lncRNAs were aberrantly expressed (≥ twofold change). Of these, 66 mRNAs and 53 lncRNAs were upregulated, while 75 lncRNAs and 156 mRNAs were downregulated. KEGG pathway analysis and the Gene ontology category indicated that these genes were associated with the regulation of mRNA alternative splicing and metabolic balance. Clinical analyses revealed that overexpression of DKFZp434J0226 was associated with worse tumor grading, frequent perineural invasion, advanced tumor-node-metastasis stage, and decreased overall survival and time to progression. Functional assays demonstrated that DKFZp434J0226 promoted PDAC cell migration, invasion, and growth in vitro and accelerated tumor proliferation in vivo. Mechanistically, DKFZp434J0226 interacted with the splicing factor SF3B6 and promoted its phosphorylation, which further regulated the alternative splicing of pre-mRNA. CONCLUSIONS: This study indicates that DKFZp434J0226 regulates alternative splicing through phosphorylation of SF3B6 in PDAC and leads to an oncogenic phenotype in PDAC.


Asunto(s)
Empalme Alternativo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Factores de Empalme de ARN/metabolismo , ARN Largo no Codificante , Animales , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Biología Computacional/métodos , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Ratones , Neoplasias Pancreáticas/patología , Fosforilación , Pronóstico , Unión Proteica , Transporte de Proteínas , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
4.
Am J Physiol Cell Physiol ; 318(3): C605-C614, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31940247

RESUMEN

Hu-antigen R (HuR) is involved in the carcinogenesis and progression of multiple types of cancer. However, its precise role in gastric cancer (GC) and the relevant molecular mechanism remain largely unclear. In the present study, we found that HuR expression level was higher in GC tissues and cell lines than in adjacent normal tissues and normal gastric epithelial cell lines, and this elevated expression was found to have a significant association with lymph node metastasis. Moreover, silencing HuR with RNA interference inhibited cell viability and induced cell apoptosis through the apoptosis-related regulators (Bcl-2 and Bax) in GC cells. In addition, bioinformatic analysis revealed that HuR expression was inversely correlated with miR-145 expression in GC tissue samples, and HuR was identified as a direct target of miR-145 with the dual-luciferase reporter. Enforced expression of miR-145 inhibited the HuR expression at both mRNA and protein levels and induced similar biologic effects of silencing HuR in GC cells. Additionally, we also found that restoration of HuR could eliminate the effects induced by miR-145 in GC cells. Taken together, these findings demonstrate the exact role of the miR-145-HuR axis in the progression of GC and indicate a potential target for GC therapy.


Asunto(s)
Progresión de la Enfermedad , Proteína 1 Similar a ELAV/metabolismo , MicroARNs/metabolismo , Neoplasias Gástricas/metabolismo , Línea Celular , Proteína 1 Similar a ELAV/antagonistas & inhibidores , Proteína 1 Similar a ELAV/genética , Marcación de Gen/métodos , Células HEK293 , Humanos , MicroARNs/genética , Neoplasias Gástricas/patología
5.
FASEB J ; 33(4): 5411-5424, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30698996

RESUMEN

The epithelial-mesenchymal transition (EMT) is crucial for cancer progression. Evidence has shown that miR-22 and miR-214 play a key role in colon cancer progression; however, the underlying mechanism remains to be known. The effects of miR-22 and miR-214 on EMT are contradictory in different cancers, and whether miR-22 and miR-214 are involved in the colon cancer EMT process needs to be elucidated. In this study, we evaluated the exact role and the regulation mechanism of miR-22 and miR-214 in colon cancer. After transfection with miR-22 expression vector, the cell proliferation and migration capacity of HCT116 and RKO cells were significantly suppressed. Also, E-cadherin was increased and vimentin was decreased by miR-22 overexpression. Similar effects were also observed after miR-214 expression vector transfection. Dual-luciferase reporter confirmed that BCL9L is the target gene of both miR-22 and miR-214. Silencing of BCL9L inhibits cell proliferation and migration, and the expression of E-cadherin and vimentin was also altered by BCL9L knockdown, which was consistent with miR-22 or miR-214 transfection. Furthermore, miR-22 and miR-214 inhibited tumor growth in nude mice. Moreover, although the association between BCL9L's lower expression and longer survival time was statistically nonsignificant, a trend existed; further studies in a larger cohort are needed. Collectively, these data suggest that miR-22 and miR-214 inhibit cell proliferation, migration, and EMT of colon cancer, most likely by targeting BCL9L.-Sun, R., Liu, Z., Han, L., Yang, Y., Wu, F., Jiang, Q., Zhang, H., Ma, R., Miao, J., He, K., Wang, X., Zhou, D., Huang, C. miR-22 and miR-214 targeting BCL9L inhibit proliferation, metastasis, and epithelial-mesenchymal transition by down-regulating Wnt signliang in colon cancer.


Asunto(s)
Proliferación Celular/genética , Neoplasias del Colon/genética , Proteínas de Unión al ADN/genética , Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , Factores de Transcripción/genética , Vía de Señalización Wnt/genética , Animales , Apoptosis/genética , Cadherinas/genética , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Neoplasias del Colon/patología , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Vimentina/genética
6.
Anticancer Drugs ; 31(6): 583-591, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32282367

RESUMEN

Colorectal cancer (CRC) is one of most common cancers worldwide. Although miR-203a is reported as a tumor suppressor involved in cell progression in some cancers, the role of miR-203a in CRC is still controversial and the underling mechanism of miR-203a in CRC remains unclear. Here, we demonstrated that low expression of miR-203a had poorer survival in CRC patients. miR-203a was down-regulated in most human colon cancer cells. Overexpression of miR-203a could inhibit colon cancer cell proliferation and arrest cell cycle in G1 phase. Bioinformatics and dual luciferase reporter assay confirmed that RING-finger protein 6 (RNF6) was a target gene of miR-203a. Silencing RNF6 inhibited cell proliferation and arrest cell cycle in G1 phase. RNF6 overexpression reversed the effects of miR-203a overexpression in colon cancer cells. Taken together, our data indicate that miR-203a inhibits colon cancer cell proliferation by targeting RNF6, offer novel insights into the regulatory network of miR-203a-modulated cell cycle and proliferation, and suggest that miR-203a a potential therapeutic target in CRC treatment.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proliferación Celular , Neoplasias Colorrectales/patología , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Proteínas de Unión al ADN/genética , Humanos , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas
7.
Cell Mol Biol (Noisy-le-grand) ; 64(5): 40-45, 2018 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-29729692

RESUMEN

Gastric cancer (GC) is one of the most common malignant cancer around the world, however the mechanisms is still unclear. In the present study, we investigated the function of CREB3 regulatory factor (CREBRF) in human GC and explored its relevant molecular mechanism. We found that CREBRF was highly expressed in primary GC tissues and the expression level was associated with the clinicopathologic characteristics of GC. CREBRF silencing inhibited GC cell proliferation and induced G1/G0 to S phase cell cycle arrest through regulating Cyclin A, Cyclin D1 and CDK2 expressions. Furthermore, the results showed that knockdown of CREBRF suppressed the activation of AKT signaling pathway. We further discovered that activating of AKT rescued the effect of CREBRF silencing on cell growth and drove cell re-enter into the S phase of the cell cycle with SC79 (a AKT activator). Taken together, our study demonstrated that CREBRF might promote GC cell proliferation and induce G1-S phase transition through activating AKT signaling pathway. These findings suggest that CREBRF acts as a novel oncogene and may be a potential therapeutic target in therapy of GC.


Asunto(s)
Puntos de Control de la Fase G1 del Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , Neoplasias Gástricas/genética , Proteínas Supresoras de Tumor/genética , Anciano , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular , Ciclina A/genética , Ciclina A/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/metabolismo , Femenino , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Proteínas Proto-Oncogénicas c-akt/agonistas , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo
9.
World J Gastrointest Oncol ; 16(3): 577-582, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38577447

RESUMEN

We conducted a comprehensive review of existing prediction models pertaining to the efficacy of immune-checkpoint inhibitor (ICI) and the occurrence of immune-related adverse events (irAEs). The predictive potential of neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) in determining ICI effectiveness has been extensively investigated, while limited research has been conducted on predicting irAEs. Furthermore, the combined model incorporating NLR and PLR, either with each other or in conjunction with additional markers such as carcinoembryonic antigen, exhibits superior predictive capabilities compared to individual markers alone. NLR and PLR are promising markers for clinical applications. Forthcoming models ought to incorporate established efficacious models and newly identified ones, thereby constituting a multifactor composite model. Furthermore, efforts should be made to explore effective clinical application approaches that enhance the predictive accuracy and efficiency.

10.
Front Microbiol ; 15: 1298262, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38567072

RESUMEN

Introduction: Dietary nutrient content is crucial for energy metabolism and development of gut microbiota. Herein, this study aimed to explore the effects of fat-to-fiber ratios on nutrient transporter, energy metabolism and gut microbiota when ingredients composition was altered. Methods: A total of 240 as-hatched broiler chickens were randomly assigned into three groups including low fat-high dietary fiber (LF-HD), medium fat-medium dietary fiber (MF-MD) and high fat-low dietary fiber (HF-LD), with diets being iso-protein, and broilers were offered the same commercial diets from 21 to 42 d. The data were analyzed using one-way ANOVA of SPSS. Results and Discussion: Results showed that HF-LD diet significantly increased glucose content and decreased triglyceride in serum of broilers (p < 0.05). The mRNA abundance of jejunal gene involved in glucose transporter and tricarboxylic acid (TCA) cycle was significantly increased in broilers fed with HF-LD diets. Compared with LF-HD, HF-LD had a lower abundance of Anaerofilum and CHKCI001, and an increased proportion of beneficial bacteria such as Alistipes, Catenibacillus, Intestinimonas, Lactobacillus, and Peptococcus (p < 0.05). Functional prediction of these microbial changes indicated that HF-LD diet drove caecal microbiota to participate in carbohydrate metabolism and TCA cycle (p < 0.05). Dietary HF-LD-induced microbiota changes were positively correlated with growth performance of broilers (p < 0.05). Therefore, HF-LD diet increased glucose transporters and energy metabolism in intestine and shaped microbial structure and metabolic pathways, which may benefit the growth performance of broilers.

11.
Animals (Basel) ; 14(8)2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38672346

RESUMEN

Accurately determining the energy values of ingredients is crucial for meeting energy requirements and achieving maximum production performance of animals. This study was conducted to measure the available energy values of three expanded soybean meals (ESBMs) for Arbor Acres male broilers from 14 to 16 day and 28 to 30 day using the difference method. A corn-soybean basal diet was formulated, and test diets were developed with 25% ESBMs as substitutes for energy-yielding ingredients. A completely randomized design was used for determining heat production and energy balance of broilers in 12 open-circuit respiration chambers, with six replicates per group. Prior to measurement, four (14 to 16 day) or two (28 to 30 day) birds per chamber were given a 4-day adaption to diets and chambers. The period lasted for 3 days to determine the apparent metabolizable energy (AME), nitrogen balance, gas exchanges, and heat production. Broilers fed test diets with 25% ESBM exhibited higher nitrogen intake (p < 0.05), nitrogen excreta (p < 0.05), and increased energy deposition as protein irrespective of age (p < 0.05). Furthermore, results showed that AME, nitrogen corrected AME (AMEn), and net energy (NE) values of 3 ESBMs averaged 10.48, 8.93, and 6.88 MJ/kg for broilers from 14 to 16 day, while averaged 11.91, 10.42, and 6.43 MJ/kg for broilers from 28 to 30 day. Broilers from 28 to 30 day showed significantly higher AMEn values but lower NE/AME values of ESBMs compared with those from 14 to 16 day (p < 0.05). Therefore, age-dependent energy values of a single ingredient should be considered in feed formulations to optimize economic returns.

12.
J Immunother Cancer ; 12(1)2024 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-38272562

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is an exceptionally immunosuppressive malignancy characterized by limited treatment options and a dismal prognosis. Macrophages constitute the primary and heterogeneous immune cell population within the HCC microenvironment. Our objective is to identify distinct subsets of macrophages implicated in the progression of HCC and their resistance to immunotherapy. METHODS: Intratumoral macrophage-specific marker genes were identified via single-cell RNA sequencing analyses. The clinical relevance of phospholipase A2 Group VII (PLA2G7), a pivotal enzyme in phospholipid metabolism, was assessed in patients with HCC through immunohistochemistry and immunofluorescence. Flow cytometry and an in vitro co-culture system were used to elucidate the specific role of PLA2G7 in macrophages. Orthotopic and subcutaneous HCC mouse models were employed to evaluate the potential of the PLA2G7 inhibitor in complementing immune checkpoint blockade (ICB) therapy. RESULTS: Single-cell RNA sequencing analyses disclosed predominant PLA2G7 expression in intratumoral macrophages within the HCC microenvironment. The macrophage-specific PLA2G7 was significantly correlated with poorer prognosis and immunotherapy resistance in patients with HCC. PLA2G7high macrophages represent a highly immunosuppressive subset and impede CD8 T-cell activation. Pharmacological inhibition of PLA2G7 by darapladib improved the therapeutic efficacy of anti-programmed cell death protein 1 antibodies in the HCC mouse models. CONCLUSIONS: Macrophage-specific PLA2G7 serves as a novel biomarker capable of prognosticating immunotherapy responsiveness and inhibiting PLA2G7 has the potential to enhance the efficacy of ICB therapy for HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Ratones , Animales , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Macrófagos , Inmunoterapia , Pronóstico , Microambiente Tumoral , 1-Alquil-2-acetilglicerofosfocolina Esterasa/uso terapéutico
13.
J Hepatocell Carcinoma ; 11: 1357-1373, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011124

RESUMEN

Background: CD276 is an emerging immune checkpoint molecule that has been implicated in various cancers. However, its specific role in hepatocellular carcinoma (HCC) remains unclear. This study examined the impact of CD276 on patient prognosis and the tumor microenvironment (TME). Methods: The Cancer Genome Atlas (TCGA) database was utilized to evaluate CD276 expression in HCC and the association between CD276 and immune indicators was also analyzed. The signaling pathways correlated with CD276 expression were identified by gene set enrichment analysis (GSEA). Different algorithms were used to assess immune cell infiltration. The effect of CD276 knockdown on HCC cell phenotypes and its relationship with macrophage polarization was examined using the cell counting kit 8 (CCK-8) assay and co-culture system. Results: CD276 was upregulated in HCC and associated with unfavorable clinical outcomes. Hgh CD276 expression was associated with enrichment of the G2/M checkpoint, E2F targets, and mitotic spindles. CD276 expression was correlated with the infiltration of immune cells, including high level of tumor-associated macrophages and low levels of CD8+ T cells. Knockdown of CD276 decreased HCC cell proliferation and increased apoptosis. CD276 silencing in HCC cells and co-culture with THP-1-derived macrophages had a regulatory effect on macrophage polarization and macrophage-mediated cell proliferation and migration. Conclusion: CD276 expression in HCC is associated with unfavorable clinical outcomes and may contribute to the development of an immunosuppressive microenvironment. Specifically, CD276 was associated with alterations in immune cell infiltration, immune marker expression, and macrophage polarization during HCC progression, suggesting its potential as a prognostic indicator and promising target for immunotherapeutic intervention in HCC.

14.
Cell Cycle ; 23(5): 573-587, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38701194

RESUMEN

Myogenic differentiation (MyoD) 1, which is known as a pivotal transcription factor during myogenesis, has been proven dysregulated in several cancers. However, litter is known about the precise role and downstream genes of MyoD1 in gastric cancer (GC) cells. Here, we report that MyoD1 is lowly expressed in primary GC tissues and cells. In our experiments, overexpression of MyoD1 inhibited cell proliferation. Downstream genes of MyoD1 regulation were investigated using RNA-Seq. As a result, 138 up-regulated genes and 20 down-regulated genes and 27 up-regulated lncRNAs and 20 down-regulated lncRNAs were identified in MyoD1 overexpressed MKN-45 cells, which participated in epithelial cell signaling in Helicobacter pylori infection, glycosaminoglycan biosynthesis (keratan sulfate), notch signaling pathway, and others. Among these genes, BIK was directly regulated by MyoD1 in GC cells and inhibited cancer cell proliferation. The BIK knockdown rescued the effects of MyoD1 overexpression on GC cells. In conclusion, MyoD1 inhibited cell proliferation via 158 genes and 47 lncRNAs downstream directly or indirectly that participated in multiple signaling pathways in GC, and among these, MyoD1 promotes BIK transcription by binding to its promoter, then promotes BIK-Bcl2-caspase 3 axis and regulates GC cell apoptosis.


Asunto(s)
Apoptosis , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Proteína MioD , ARN Largo no Codificante , Neoplasias Gástricas , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Neoplasias Gástricas/metabolismo , Humanos , Apoptosis/genética , Proteína MioD/metabolismo , Proteína MioD/genética , Proliferación Celular/genética , Línea Celular Tumoral , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Transducción de Señal/genética , Transcripción Genética/genética
15.
J Control Release ; 368: 157-169, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38367861

RESUMEN

Gene delivery to macrophages holds great promise for cancer immunotherapy. However, traditional gene delivery methods exhibit low transfection efficiency in macrophages. The star-shaped topological structure of polymers is known to encapsulate genes inside their cores, thereby facilitating sustained release of the genetic material. Herein, combining the structural advantages of star polymers and the transfection advantages of poly (ß-amino ester)s (PAEs), we developed a novel linear oligomer grafting-onto strategy to synthesize a library of multi-terminal star structured PAEs (SPAEs), and evaluated their gene delivery efficiency in various tissue cells. The transfection with human hepatocellular carcinoma cells (HepG2, HCC-LM3 cells and MHCC-97H cells), rat normal liver cells (BRL-3 A cells), human ovarian cancer cells (A2780 cells), African green monkey kidney cells (Vero cells), human cervical cancer cells (HeLa cells), human chondrosarcoma cells (SW1353 cells), and difficult-to-transfect human epidermal keratinocytes (HaCaT cells) and normal human fibroblast cells (NHF cells) showed that SPAEs exhibited superior transfection profile. The GFP transfection efficiency of top-performing SPAEs in HeLa cells (96.1%) was 2.1-fold, and 3.2-fold higher compared to jetPEI and Lipo3000, respectively, indicating that the star-shaped topological structure can significantly enhance the transfection efficiency of PAEs. More importantly, the top-performing SPAEs could efficiently deliver Nod2 DNA to difficult-to-transfect RAW264.7 macrophages, with a high transfection efficiency of 33.9%, which could promote macrophage M1 polarization and enhanced CD8+ T cell response in co-incubation experiments. This work advances gene therapy by targeting difficult-to-transfect macrophages and remodeling the tumor immune microenvironment.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Neoplasias Ováricas , Ratas , Humanos , Animales , Chlorocebus aethiops , Femenino , Células HeLa , Línea Celular Tumoral , Células Vero , Ésteres , Transfección , Terapia Genética , Polímeros/química , Macrófagos , Microambiente Tumoral
16.
Cell Death Dis ; 15(6): 427, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890303

RESUMEN

As the second most common malignant tumor in the urinary system, renal cell carcinoma (RCC) is imperative to explore its early diagnostic markers and therapeutic targets. Numerous studies have shown that AURKB promotes tumor development by phosphorylating downstream substrates. However, the functional effects and regulatory mechanisms of AURKB on clear cell renal cell carcinoma (ccRCC) progression remain largely unknown. In the current study, we identified AURKB as a novel key gene in ccRCC progression based on bioinformatics analysis. Meanwhile, we observed that AURKB was highly expressed in ccRCC tissue and cell lines and knockdown AURKB in ccRCC cells inhibit cell proliferation and migration in vitro and in vivo. Identified CDC37 as a kinase molecular chaperone for AURKB, which phenocopy AURKB in ccRCC. AURKB/CDC37 complex mediate the stabilization of MYC protein by directly phosphorylating MYC at S67 and S373 to promote ccRCC development. At the same time, we demonstrated that the AURKB/CDC37 complex activates MYC to transcribe CCND1, enhances Rb phosphorylation, and promotes E2F1 release, which in turn activates AURKB transcription and forms a positive feedforward loop in ccRCC. Collectively, our study identified AURKB as a novel marker of ccRCC, revealed a new mechanism by which the AURKB/CDC37 complex promotes ccRCC by directly phosphorylating MYC to enhance its stability, and first proposed AURKB/E2F1-positive feedforward loop, highlighting AURKB may be a promising therapeutic target for ccRCC.


Asunto(s)
Aurora Quinasa B , Carcinoma de Células Renales , Proteínas de Ciclo Celular , Progresión de la Enfermedad , Factor de Transcripción E2F1 , Neoplasias Renales , Proteínas Proto-Oncogénicas c-myc , Humanos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Carcinoma de Células Renales/metabolismo , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F1/genética , Neoplasias Renales/patología , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Fosforilación , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Aurora Quinasa B/metabolismo , Aurora Quinasa B/genética , Proliferación Celular , Animales , Regulación Neoplásica de la Expresión Génica , Ratones Desnudos , Ratones , Movimiento Celular/genética , Chaperoninas
17.
Int J Biol Sci ; 20(8): 3008-3027, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38904013

RESUMEN

SET domain containing 7(SETD7), a member of histone methyltransferases, is abnormally expressed in multiple tumor types. However, the biological function and underlying molecular mechanism of SETD7 in clear cell renal cell carcinoma (ccRCC) remain unclear. Here, we explored the biological effects of SETD7-TAF7-CCNA2 axis on proliferation and metastasis in ccRCC. We identified both SETD7 and TAF7 were up-regulated and significantly promoted the proliferation and migration of ccRCC cells. Concurrently, there was a significant positive correlation between the expression of SETD7 and TAF7, and the two were colocalized in the nucleus. Mechanistically, SETD7 methylates TAF7 at K5 and K300 sites, resulting in the deubiquitination and stabilization of TAF7. Furthermore, re-expression of TAF7 could partially restore SETD7 knockdown inhibited ccRCC cells proliferation and migration. In addition, TAF7 transcriptionally activated to drive the expression of cyclin A2 (CCNA2). And more importantly, the methylation of TAF7 at K5 and K300 sites exhibited higher transcriptional activity of CCNA2, which promotes formation and progression of ccRCC. Our findings reveal a unique mechanism that SETD7 mediated TAF7 methylation in regulating transcriptional activation of CCNA2 in ccRCC progression and provide a basis for developing effective therapeutic strategies by targeting members of SETD7-TAF7-CCNA2 axis.


Asunto(s)
Carcinoma de Células Renales , Movimiento Celular , Proliferación Celular , N-Metiltransferasa de Histona-Lisina , Neoplasias Renales , Humanos , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Proliferación Celular/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Movimiento Celular/genética , Neoplasias Renales/metabolismo , Neoplasias Renales/genética , Neoplasias Renales/patología , Línea Celular Tumoral , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factores Asociados con la Proteína de Unión a TATA/genética , Metilación , Factor de Transcripción TFIID/metabolismo , Factor de Transcripción TFIID/genética , Regulación Neoplásica de la Expresión Génica
18.
Front Biosci (Landmark Ed) ; 28(10): 235, 2023 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-37919059

RESUMEN

BACKGROUND: Pyroptosis-related genes (PRG) are closely associated with the progression and metastasis of hepatocellular carcinoma (HCC). The predictive power of PRGs could be used to assess the clinical outcomes of HCC. METHODS: The Cancer Genome Atlas (TCGA) RNA-seq data and clinical information from patients with liver hepatocellular carcinoma (LIHC) were used to identify PRG with differentially expressed between HCC and normal samples. Univariate Cox regression, least absolute shrinkage and selection operator (LASSO) Cox method, and multivariate Cox regression analysis were used to develop a prognostic model that included three PRGs. Gene set enrichment analysis (GSEA) was performed to identify differential immune cells and their associated pathways. The expression of Gasdermin C (GSDMC) in the HCC samples was detected by western blotting, and the function of GSDMC in HCC proliferation and metastasis was detected by the Cell Counting Kit-8 (CCK-8), colony formation, cell invasion, and wound healing assays. RESULTS: Of 52 PRGs, GSDMC, Bcl-2 homologusantagonist/ killer 1 (BAK1), and NOD-like receptor thermal protein domain associated protein 6 (NLRP6) were selected to establish a prognostic model. The model successfully differentiated HCC patients with varied survival in the TCGA training and test cohorts, as well as the International Cancer Genome Consortium (ICGC) validation cohorts. The risk score was proven to be an independent prognostic factor. In addition, we also reported a marked upregulation of GSDMC in HCC tissues, which could be induced by CD274 (PD-L1). Overexpression of GSDMC contributes to HCC cells invasion, proliferation, and migration. CONCLUSIONS: The three PRGs signatures containing GSDMC independently predicted HCC prognosis. As a new driver molecule, GSDMC could play a tumor-promoting role by facilitating HCC growth and metastasis.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/genética , Piroptosis/genética , Neoplasias Hepáticas/genética , Activación Transcripcional , Biomarcadores de Tumor/genética , Proteínas Citotóxicas Formadoras de Poros
19.
Anim Nutr ; 12: 72-76, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36514375

RESUMEN

Optimizing the energy utilization of nutrients and ensuring maximum benefits are continuous goals for livestock producers. The net energy (NE) value of feed reflects its nutritional value in the precision feeding system. An experiment was conducted to determine the apparent metabolizable energy (AME) and NE values of 3 types of dephenolized cottonseed protein (DCP) for Hy Line Brown hens aged 42 to 45 weeks using the reference diet substitution method. A reference diet based on corn soybean meal was used to meet the nutritional needs of Hy Line Brown laying hens. To render the crude protein and energy values of the 3 test diets similar, 10.5%, 12%, and 16% of the gross energy yielding ingredients from the reference diet were replaced with DCP 1, DCP 2, and DCP 3, respectively. The birds were fed 4 diets during a 7-d adaptation period. After the dietary adaptation period, 2 birds per replicate from each treatment group were placed in an individual open circuit respiratory calorimetry chamber for a 3-d experimental period. Daily O2 consumption and CO2 production were recorded, and excreta samples were collected. The AME values of DCP 1, DCP 2, and DCP 3 were 3,049.05, 2,820.13, and 2,982.31 kcal/kg of dry matter (DM), respectively. The NE values of DCP 1, DCP 2, DCP 3 were 1,475.77, 1,910.31, and 1,905.37 kcal/kg of DM, respectively, and the NE:AME ratios were 48.40%, 67.74%, and 63.89%, respectively. Our data show that the AME value of DCP does not reflect the nutritional value of the feed. The NE value of DCP with a high ME value was not necessarily high.

20.
Cancer Gene Ther ; 30(5): 727-737, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36599972

RESUMEN

The molecular mechanism of network regulation in the occurrence and development of colorectal cancer (CRC) has been constantly improved. Here, we investigated the biological effects of TEAD4-MAD2L1 axis on proliferation and metastasis of human CRC cells. This study revealed that the expressions of MAD2L1 and TEAD4 in CRC tissues and CRC cell lines were significantly higher than those in adjacent epithelial tissues and normal intestinal epithelial cell line NCM460, and their expressions were significantly positively correlated; Moreover, inhibiting the expression of MAD2L1 or TEAD4 can inhibit the proliferation and migration of CRC cells and promote apoptosis. In addition, the promoter region of MAD2L1 gene has a TEAD4 binding site (motif sequence), and the transcription of MAD2L1 is positively regulated by TEAD4 protein; The inhibition of promotion/migration and promotion of apoptosis of CRC cells by silencing TEAD4 can be saved by the high expression of MAD2L1. In conclusion, our study suggests that the transcription and expression of MAD2L1 is regulated by TEAD4, which further promotes the proliferation and migration of CRC cells in vitro and in vivo, and inhibits apoptosis. MAD2L1 and TEAD4 are potential biomarkers for colorectal cancer.


Asunto(s)
Neoplasias Colorrectales , Factores de Transcripción , Humanos , Línea Celular Tumoral , Factores de Transcripción/metabolismo , Proliferación Celular/genética , Proteínas de Ciclo Celular/genética , Neoplasias Colorrectales/patología , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Dominio TEA , Proteínas Mad2/genética , Proteínas Mad2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA