Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Biotechnol ; 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38575817

RESUMEN

Pancreatic cancer stands as one of the most lethal malignancies, characterized by delayed diagnosis, high mortality rates, limited treatment efficacy, and poor prognosis. Disulfidptosis, a recently unveiled modality of cell demise induced by disulfide stress, has emerged as a critical player intricately associated with the onset and progression of various cancer types. It has emerged as a promising candidate biomarker for cancer diagnosis, prognosis assessment, and treatment strategies. In this study, we have effectively established a prognostic risk model for pancreatic cancer by incorporating multiple differentially expressed long non-coding RNAs (DElncRNAs) closely linked to disulfide-driven cell death. Our investigation delved into the nuanced relationship between the DElncRNA-based predictive model for disulfide-driven cell death and the therapeutic responses to anticancer agents. Our findings illuminate that the high-risk subgroup exhibits heightened susceptibility to the small molecule compound AZD1208, positioning it as a prospective therapeutic agent for pancreatic cancer. Finally, we have elucidated the underlying mechanistic potential of AZD1208 in ameliorating pancreatic cancer through its targeted inhibition of the peroxisome proliferator-activated receptor-γ (PPARG) protein, employing an array of comprehensive analytical methods, including molecular docking and molecular dynamics (MD) simulations. This study explores disulfidptosis-related genes, paving the way for the development of targeted therapies for pancreatic cancer and emphasizing their significance in the field of oncology. Furthermore, through computational biology approaches, the drug AZD1208 was identified as a potential treatment targeting the PPARG protein for pancreatic cancer. This discovery opens new avenues for exploring targets and screening drugs for pancreatic cancer.

2.
Mol Biotechnol ; 2024 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-38520499

RESUMEN

Acute pancreatitis (AP) and chronic pancreatitis (CP) are considered to be two separate pancreatic diseases in most studies, but some clinical retrospective analyses in recent years have found some degree of correlation between the two in actual treatment, however, the exact association is not clear. In this study, bioinformatics analysis was utilized to examine microarray sequencing data in mice, with the aim of elucidating the critical signaling pathways and genes involved in the progression from AP to CP. Differential gene expression analyses on murine transcriptomes were conducted using the R programming language and the R/Bioconductor package. Additionally, gene network analysis was performed using the STRING database to predict correlations among genes in the context of pancreatic diseases. Functional enrichment and gene ontology pathways common to both diseases were identified using Metascape. The hub genes were screened in the cytoscape algorithm, and the mRNA levels of the hub genes were verified in mice pancreatic tissues of AP and CP. Then the drugs corresponding to the hub genes were obtained in the drug-gene relationship. A set of hub genes, including Jun, Cd44, Epcam, Spp1, Anxa2, Hsp90aa1, and Cd9, were identified through analysis, demonstrating their pivotal roles in the progression from AP to CP. Notably, these genes were found to be enriched in the Helper T-cell factor (Th17) signaling pathway. Up-regulation of these genes in both AP and CP mouse models was validated through quantitative real-time polymerase chain reaction (qRT-PCR) results. The significance of the Th17 signaling pathway in the transition from AP to CP was underscored by our findings. Specifically, the essential genes driving this progression were identified as Jun, Cd44, Epcam, Spp1, Anxa2, Hsp90aa1, and Cd9. Crucial insights into the molecular mechanisms underlying pancreatitis progression were provided by this research, offering promising avenues for the development of targeted therapeutic interventions.

3.
Future Med Chem ; 15(5): 437-451, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37013861

RESUMEN

Aim: To identify novel inhibitors of SIRT1 and to understand their mechanism of action in hepatocellular carcinoma. Materials & methods: Molecular docking and dynamic simulations were conducted to identify potential SIRT1 inhibitors. The in vitro efficacy of the inhibitors was evaluated by methyl thiazolyl tetrazolium assays, flow cytometry and western blot analysis. Additionally, the in vivo antitumor activity of the inhibitor was evaluated. Results: Tipranavir, a US FDA-approved anti-HIV-1 medication, was found to possess potential as a SIRT1 inhibitor. Tipranavir selectively inhibited HepG2 cell proliferation without causing toxicity to normal human hepatic cells. Additionally, tipranavir treatment resulted in a reduction of SIRT1 expression and induction of apoptosis in HepG2 cells. Furthermore, tipranavir was found to suppress tumorigenesis in a xenograft mouse model and decreased the expression of SIRT1 in vivo. Conclusion: Tipranavir holds desirable potential as a promising therapeutic agent against hepatoma.


Asunto(s)
Fármacos Anti-VIH , Carcinoma Hepatocelular , Neoplasias Hepáticas , Sirtuina 1 , Animales , Humanos , Ratones , Fármacos Anti-VIH/farmacología , Apoptosis , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Neoplasias Hepáticas/metabolismo , Simulación del Acoplamiento Molecular , Piridinas/farmacología , Sirtuina 1/antagonistas & inhibidores
4.
Bioorg Chem ; 133: 106382, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36716580

RESUMEN

Acute pancreatitis (AP) is a frequent abdominal inflammatory disease. Despite the high morbidity and mortality, the management of AP remains unsatisfactory. Disulfiram (DSF) is an FDA-proved drug with potential therapeutic effects on inflammatory diseases. In this study, we aim to investigate the effect of DSF on pancreatic acinar cell necrosis, and to explore the underlying mechanisms. Cell necrosis was induced by sodium taurocholate or caerulein, AP mice model was induced by nine hourly injections of caerulein. Network pharmacology, molecular docking, and molecular dynamics simulation were used to explore the potential targets of DSF in protecting against cell necrosis. The results indicated that DSF significantly inhibited acinar cell necrosis as evidenced by a decreased ratio of necrotic cells in the pancreas. Network pharmacology, molecular docking, and molecular dynamics simulation identified RIPK1 as a potent target of DSF in protecting against acinar cell necrosis. qRT-PCR analysis revealed that DSF decreased the mRNA levels of RIPK1 in freshly isolated pancreatic acinar cells and the pancreas of AP mice. Western blot showed that DSF treatment decreased the expressions of RIPK1 and MLKL proteins. Moreover, DSF inhibited NF-κB activation in acini. It also decreased the protein expression of TLR4 and the formation of neutrophils extracellular traps (NETs) induced by damage-associated molecular patterns released by necrotic acinar cells. Collectively, DSF could ameliorate the severity of mouse acute pancreatitis by inhibiting RIPK-dependent acinar cell necrosis and the following formation of NETs.


Asunto(s)
Pancreatitis , Ratones , Animales , Pancreatitis/tratamiento farmacológico , Pancreatitis/inducido químicamente , Células Acinares , Disulfiram/efectos adversos , Ceruletida/efectos adversos , Enfermedad Aguda , Simulación del Acoplamiento Molecular , Necrosis , Proteína Serina-Treonina Quinasas de Interacción con Receptores/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/uso terapéutico
5.
J Biol Chem ; 298(10): 102339, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35931121

RESUMEN

Family with sequence similarity 83 A (FAM83A) is a newly discovered proto-oncogene that has been shown to play key roles in various cancers. However, the function of FAM83A in other physiological processes is not well known. Here, we report a novel function of FAM83A in adipocyte differentiation. We used an adipocyte-targeting fusion oligopeptide (FITC-ATS-9R) to deliver a FAM83A-sgRNA/Cas9 plasmid to knockdown Fam83a (ATS/sg-FAM83A) in white adipose tissue in mice, which resulted in reduced white adipose tissue mass, smaller adipocytes, and mitochondrial damage that was aggravated by a high-fat diet. In cultured 3T3-L1 adipocytes, we found loss or knockdown of Fam83a significantly repressed lipid droplet formation and downregulated the expression of lipogenic genes and proteins. Furthermore, inhibition of Fam83a decreased mitochondrial ATP production through blockage of the electron transport chain, associated with enhanced apoptosis. Mechanistically, we demonstrate FAM83A interacts with casein kinase 1 (CK1) and promotes the permeability of the mitochondrial outer membrane. Furthermore, loss of Fam83a in adipocytes hampered the formation of the TOM40 complex and impeded CK1-driven lipogenesis. Taken together, these results establish FAM83A as a critical regulator of mitochondria maintenance during adipogenesis.


Asunto(s)
Adipocitos Blancos , Adipogénesis , Quinasa de la Caseína I , Mitocondrias , Proteínas de Neoplasias , Proto-Oncogenes , Animales , Ratones , Células 3T3-L1 , Adipocitos Blancos/citología , Adipocitos Blancos/metabolismo , Adipogénesis/genética , Quinasa de la Caseína I/metabolismo , Diferenciación Celular , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
6.
Cell Regen ; 11(1): 14, 2022 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-35362877

RESUMEN

The Chchd10 gene encodes a coiled-coil-helix-coiled-coil-helix-domain containing protein predicted to function in the mitochondrion and nucleus. Mutations of Chchd10 are associated with ALS, dementia and myopathy in humans and animal models, but how knockout of Chchd10 (Chchd10KO) affects various tissues especially skeletal muscle and adipose tissues remains unclear. Here we show that Chchd10 expression increases as myoblasts and preadipocytes differentiate. During myogenesis, CHCHD10 interacts with TAR DNA binding protein 43 (TDP-43) in regenerating myofibers in vivo and in newly differentiated myotubes ex vivo. Surprisingly, Chchd10KO mice had normal skeletal muscle development, growth and regeneration, with moderate defects in grip strength and motor performance. Chchd10KO similarly had no effects on development of brown and white adipose tissues (WAT). However, Chchd10KO mice had blunted response to acute cold and attenuated cold-induced browning of WAT, with markedly reduced UCP1 levels. Together, these results demonstrate that Chchd10 is dispensable for normal myogenesis and adipogenesis but is required for normal motility and cold-induced, mitochondrion-dependent browning of adipocytes. The data also suggest that human CHCHD10 mutations cause myopathy through a gain-of-function mechanism.

7.
J Biol Chem ; 296: 100037, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33158991

RESUMEN

With the improvement of people's living standards, the number of obese patients has also grown rapidly. It is reported that the level of oxidative stress in obese patients has significantly increased, mainly caused by the increase in reactive oxygen species (ROS) levels in adipose tissue. Studies have shown that the use of siRNA to interfere with bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) expression could promote adipocyte differentiation, and under hypoxic conditions, BAMBI could act as a regulator of HIF1α to regulate the polarity damage of epithelial cells. In view of these results, we speculated that BAMBI may regulate adipogenesis by regulating the level of ROS. In this study, we generated adipose-specific BAMBI knockout mice (BAMBI AKO) and found that compared with control mice, BAMBI AKO mice showed obesity when fed with high-fat diet, accompanied by insulin resistance, glucose intolerance, hypercholesterolemia, and increased inflammation in adipose tissue. Interestingly, adipose-specific deficiency of BAMBI could cause an increase in the expression level of Nox4, thereby promoting ROS production in cytoplasm and mitochondria and the DNA-binding activity of C/EBPß and ultimately promoting adipogenesis. Consistently, our findings indicated that BAMBI may be a reactive oxygen regulator to affect adipogenesis, thereby controlling obesity and metabolic syndrome.


Asunto(s)
Adipogénesis , Tejido Adiposo/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas de la Membrana/genética , Especies Reactivas de Oxígeno/metabolismo , Acetilcisteína/farmacología , Tejido Adiposo/citología , Animales , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Dieta Alta en Grasa , Hígado Graso/genética , Humanos , Resistencia a la Insulina/genética , Ratones , Ratones Noqueados
8.
FASEB J ; 35(1): e21154, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33140469

RESUMEN

Myogenesis includes sequential stages of progenitor cell proliferation, myogenic commitment and differentiation, myocyte fusion, and myotube maturation. Different stages of myogenesis are orchestrated and regulated by myogenic regulatory factors and various downstream cellular signaling. Here we identify phosphatase orphan 1 (Phospho1) as a new player in myogenesis. During activation, proliferation, and differentiation of quiescent satellite cells, the expression of Phospho1 gradually increases. Overexpression of Phospho1 inhibits myoblast proliferation but promotes their differentiation and fusion. Conversely, knockdown of Phospho1 accelerates myoblast proliferation but impairs myotube formation. Moreover, knockdown of Phospho1 decreases the OXPHO protein levels and mitochondria density, whereas overexpression of Phospho1 upregulates OXPHO protein levels and promotes mitochondrial oxygen consumption. Finally, we show that Phospho1 expression is controlled by myogenin, which binds to the promoter of Phospho1 to regulate its transcription. These results indicate a key role of Phospho1 in regulating myogenic differentiation and mitochondrial function.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Regulación Enzimológica de la Expresión Génica , Desarrollo de Músculos , Mioblastos Esqueléticos/enzimología , Monoéster Fosfórico Hidrolasas/biosíntesis , Animales , Ratones , Mitocondrias Musculares/genética , Mitocondrias Musculares/metabolismo , Miogenina/genética , Miogenina/metabolismo , Monoéster Fosfórico Hidrolasas/genética
9.
Mol Ther Nucleic Acids ; 22: 722-732, 2020 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-33230469

RESUMEN

Skeletal muscle is an important metabolic organ of the body, and impaired skeletal muscle differentiation can result in a wide range of metabolic diseases. It has been shown that microRNAs (miRNAs) play an important role in skeletal muscle differentiation. The aim of this study was to investigate the role of mmu-miR-324-5p in the differentiation of C2C12 myoblasts and lipid droplet deposition in myotubes for future targeted therapies. We found that mmu-miR-324-5p was highly expressed in mouse skeletal muscle. Overexpression of miR-324-5p significantly inhibited C2C12 myoblast differentiation while promoting oleate-induced lipid accumulation and ß-oxidation in C2C12 myoblasts. Conversely, inhibition of mmu-miR-324-5p promoted C2C12 myoblast differentiation and inhibited lipid deposition in myotubes. Mechanistically, mmu-miR-324-5p negatively regulated the expression of long non-coding Dum (lncDum) and peptidase M20 domain containing 1 (Pm20d1) in C2C12 myoblasts. Reduced lncDum expression was associated with a significant decrease in the expression of myogenesis-related genes. Knockdown of mmu-miR-324-5p increased the levels of lncDum and myogenesis-related gene expression. Following oleate-induced lipid deposition in C2C12 myoblasts, overexpression of mmu-miR-324-5p decreased the expression of Pm20d1 while increasing the expression of mitochondrial ß-oxidation and long-chain fatty acid synthesis-related genes. In conclusion, we provide evidence that miR-324-5p inhibits C2C12 myoblast differentiation and promotes intramuscular lipid deposition by targeting lncDum and Pm20d1, respectively.

10.
Life Sci ; 258: 118240, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32781072

RESUMEN

As a dicarboxylic acid with the structural formula HOOCCH (OH) COOH, tartronic acid is considered as an inhibitor of the transformation of carbohydrates into fat under fat-deficient diet conditions. However, the effect of tartronic acid on lipogenesis under high-fat diet conditions has yet to be established. In this work, we investigated the regulatory role of tartronic acid in lipogenesis in 3T3-L1 adipocytes and C57BL/6J mice. The results confirmed that tartronic acid promoted weight gain (without affecting food intake) and induced adipocyte hypertrophy in epididymal white adipose tissue and lipid accumulation in the livers of high-fat diet-induced obese mice. In vitro, tartronic acid promoted 3T3-L1 adipocyte differentiation by increasing the protein expression of FABP-4, PPARγ and SREBP-1. Moreover, the contents of both acetyl-CoA and malonyl-CoA were significantly upregulated by treatment with tartronic acid, while the protein expression of CPT-1ß were inhibited. In summary, we proved that tartronic acid promotes lipogenesis by serving as substrates for fatty acid synthesis and inhibiting CPT-1ß, providing a new perspective for the study of tartronic acid.


Asunto(s)
Acetilcoenzima A/biosíntesis , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Lipogénesis/efectos de los fármacos , Malonil Coenzima A/biosíntesis , Tartronatos/farmacología , Regulación hacia Arriba/efectos de los fármacos , Células 3T3-L1 , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Dieta Alta en Grasa/efectos adversos , Lipogénesis/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Regulación hacia Arriba/fisiología
11.
Biochem Biophys Res Commun ; 527(2): 574-580, 2020 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-32423805

RESUMEN

Adipogenesis, a differentiation process that transitions preadipocytes to adipocytes, is key to understanding the biology of fat accumulation and obesity. During this process, there many crucial transcription factors, such as PPARγ and the C/EBP family. Here we show a transcription factor in preadipocytes --- Sox5, that has a function in porcine adipogenesis. In our porcine subcutaneous-derived preadipocyte differentiation model, we found Sox5 expression displayed a significant upregulation after initial induction and decreased afterwards, which resembles the PPARγ expression pattern. siRNA knockdown of Sox5 in porcine preadipocytes significantly promoted cell growth and accelerated cell cycle progression. After inducing differentiation, knockdown of Sox5 notably down-regulated the expression of adipogenic marker genes: PPARγ, aP2, FAS and impaired lipid accumulation. Mechanistically, the deletion of Sox5 down-regulated the BMP R-Smads signal pathway, a crucial signal pathway for controlling preadipocyte fate commitment and adipogenesis. After using BMP4 recombinant protein to activate the BMP R-Smads signal, Sox5 function was partially rescued. In conclusion, our findings uncovered a function of Sox5 in porcine adipogenesis and reveal an interaction between Sox5 and BMP signaling.


Asunto(s)
Adipogénesis , Proteínas Morfogenéticas Óseas/metabolismo , Factores de Transcripción SOXD/genética , Transducción de Señal , Proteínas Smad/metabolismo , Porcinos/fisiología , Animales , Proteínas Morfogenéticas Óseas/genética , Células Cultivadas , Regulación hacia Abajo , Interferencia de ARN , Factores de Transcripción SOXD/metabolismo , Proteínas Smad/genética , Porcinos/genética , Regulación hacia Arriba
12.
Genes (Basel) ; 10(10)2019 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-31615047

RESUMEN

Adipose tissue is one of the main organs for the energy storage and supply of organisms. Adipose deposition and metabolism are controlled by a cascade of transcription factors and epigenetic regulatory mechanisms. Previous studies have also shown that miR-106a plays a considerable role in the development of organisms. The regulatory mechanism of miR-106a on porcine preadipocytes is still not clear. In this study, preadipocytes were isolated from the neck subcutaneous deposits of 3-5-day old Chinese native Guanzhong black pigs using 5-ethynyl-20-deoxyuridine (EdU) staining and a CCK-8 assay to detect the number of proliferous cells and real-time qPCR (RT-qPCR) and western blot analysis to detect gene expression, as well as Oil Red O and BODIPY staining dye lipid droplets and flow cytometry (FCM) to detect cell cycles. We also used the double luciferase method to detect the relative luciferase activities. Upregulated miR-106a increased the number of proliferous cells and enhanced the expression of cell proliferation-related genes in porcine adipocytes. The double luciferase reporter vector confirmed that p21 was a target gene of miR-106a in the cell proliferation phase. miR-106a upregulation increased the number of lipid droplets and the expression of lipogenic genes and directly targeted BMP and activin membrane-bound inhibitor (BAMBI) in the process of differentiation. Our results indicated that miR-106a promotes porcine preadipocyte proliferation and differentiation by targeting p21 and BAMBI.


Asunto(s)
Adipocitos/metabolismo , Diferenciación Celular/genética , MicroARNs/genética , Porcinos/genética , Adipocitos/citología , Adipogénesis/genética , Animales , Proliferación Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Transducción de Señal , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...