Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Intervalo de año de publicación
1.
Int Immunopharmacol ; 134: 112139, 2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38739978

RESUMEN

Capping protein regulatory factor and myosin 1 linker 1 is termed CARMIL1. CARMIL1 is involved in several physiological processes; it forms an actin filament network and plasma membrane-bound cellular projection tissues and positively regulates the cellular components and tissues. CARMIL1 exhibits important biological functions in cancer; nonetheless, these functions have not been completely explored. We aimed to investigate the novel functions of CARMIL1 in liver cancer, particularly in cell proliferation. The cell counting kit-8, 5-ethynyl-2'-deoxyuridine, Component A experiments, and subcutaneous tumor formation model suggest that CARMIL1 is central to the proliferation of liver cancer cells both in vivo and in vitro. We extracted CARMIL1 samples from The Cancer Genome Atlas Program and analyzed its enrichment. CARMIL1 regulated the pathway activity by affecting the expression of star molecular proteins of the extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin (mTOR). Moreover, it influenced the proliferation ability of liver cancer cells. Western blotting suggested that CARMIL1 downregulation could affect ERK and mTOR phosphorylation. Results of the co-immunoprecipitation demonstrated that CARMIL1 binds to tripartite motif (TRIM)27, which in turn binds to p53. Subsequently, CARMIL1 can regulate p53 stability and promote its degradation through TRIM27. Additionally, CARMIL1 inhibition enhanced the sensitivity of liver cancer cells to sorafenib. Tumor growth was significantly inhibited in the group treated with sorafenib and CARMIL1, compared with the group treated with CARMIL1 alone. Sorafenib is a first-line targeted chemotherapeutic drug for hepatocellular carcinoma treatment. It increases the long-term survival of hepatocellular carcinoma by 44%. In this study, downregulated CARMIL1 combined with sorafenib significantly reduced the tumor volume and weight of the mouse subcutaneous tumor model, indicating the potential possibility of combining CARMIL1 with sorafenib in hepatocellular carcinoma treatment. In summary, CARMIL1 promotes liver cancer cell proliferation by regulating the TRIM27/p53 axis and activating the ERK/mTOR pathway.


Asunto(s)
Proliferación Celular , Neoplasias Hepáticas , Serina-Treonina Quinasas TOR , Proteínas de Motivos Tripartitos , Proteína p53 Supresora de Tumor , Animales , Humanos , Masculino , Ratones , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Microfilamentos/metabolismo , Proteínas de Microfilamentos/genética , Transducción de Señal , Sorafenib/farmacología , Sorafenib/uso terapéutico , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Motivos Tripartitos/metabolismo , Proteínas de Motivos Tripartitos/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética
2.
Biochem Pharmacol ; 217: 115811, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37717692

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is considered one of the most aggressive solid tumours in humans. Despite its high mortality rate, effective targeted therapeutic strategies remain limited due to incomplete understanding of the underlying biological mechanisms. The NAP1L gene family has been implicated in the development and progression of various human tumours. However, the specific function and role of NAP1L5 (nucleosome assembly protein-like 5) in PDAC have not been fully elucidated. Therefore, in this study, we aimed to investigate the role of NAP1L5 in PDAC and explore the regulatory relationship between NAP1L5 and its potential downstream molecule PHLPP1 (PH domain Leucine-rich repeat Protein Phosphatase 1) in PDAC. Our study revealed that NAP1L5 is notably upregulated in PDAC. Moreover, both in vivo and in vitro experiments demonstrated that knockdown of NAP1L5 suppressed the proliferation of PDAC cells. Mechanistically, NAP1L5 was found to promote PDAC progression by activating the AKT/mTOR signalling pathway in a PHLPP1-dependent manner. Specifically, NAP1L5 binds to PHLPP1 and facilitates the ubiquitination-mediated degradation of PHLPP1, ultimately resulting in reduced PHLPP1 expression. Notably, TRIM29, recruited by NAP1L5, was found to be involved in facilitating K48-linked ubiquitination of PHLPP1. Our findings indicate that NAP1L5 overexpression promotes the proliferation of PDAC cells by inhibiting PHLPP1 expression. These novel insights suggest that NAP1L5 may serve as a potential therapeutic target for PDAC.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Neoplasias Pancreáticas/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Transducción de Señal , Ubiquitinación , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción/metabolismo , Neoplasias Pancreáticas
3.
Biol Direct ; 18(1): 18, 2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37069693

RESUMEN

TM4SF1, a member of the transmembrane 4 superfamily, is crucial for both healthy and malignant human tissues. The significant function of TM4SF1 in the incidence and progression of cancer has been widely recognized in recent years. Although some achievements have been made in the study of TM4SF1, the effect of TM4SF1 on cancer stemness in hepatocellular carcinoma (HCC) and its molecular basis are yet to be reported. We found through abundant in vitro and in vivo experiments which the expression of TM4SF1 was positively correlated with the progression and cancer stemness of HCC. We identified the downstream protein MYH9 of TM4SF1 and its final regulatory target NOTCH pathway using bioinformatics analysis and protein mass spectrometry. We cultivated a Lenvatinib-resistant strain from HCC cells to examine the relationship between cancer stemness and tumor drug resistance. The study confirmed that TM4SF1 could regulate the NOTCH pathway by upregulating MYH9, thus promoting cancer stemness and Lenvatinib resistance in HCC. This study not only provided a new idea for the pathogenesis of HCC but also confirmed that TM4SF1 might become a new intervention point to improve the clinical efficacy of Lenvatinib in treating HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Cadenas Pesadas de Miosina , Proteínas de Neoplasias , Receptores Notch , Humanos , Antígenos de Superficie/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores Notch/metabolismo
4.
Aging (Albany NY) ; 14(22): 9000-9019, 2022 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-36374212

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. Nucleosome assembly protein 1-like 5 (NAP1L5) is a protein-coding gene that encodes a protein similar to nucleosome assembly protein 1 (NAP1). It is a histone chaperone that plays an important role in gene transcription in organisms. However, the role of NAP1L5 in the pathogenesis of hepatocellular carcinoma remains to be elucidated. In this study, low expression of NAP1L5 was found in hepatocellular carcinoma, and the downregulation of NAP1L5 was related to shorter survival and disease-free survival. In addition, its expression is also related to the tumor size and recurrence of hepatocellular carcinoma. The overexpression and knockdown of NAP1L5 by plasmid and siRNA showed that NAP1L5 inhibited the proliferation, migration and invasion and induced apoptosis of hepatoma cells. In vivo experiments confirmed that NAP1L5 can inhibit the growth and metastasis of hepatocellular carcinoma cells. In the mechanistic study, we found that NAP1L5 affects the occurrence and development of hepatocellular carcinoma by regulating MYH9 to inhibit the PI3K/AKT/mTOR signaling pathway. As a functional tumor suppressor, NAP1L5 is expressed at low levels in HCC. NAP1L5 inhibits the PI3K/AKT/mTOR signaling pathway in hepatocellular carcinoma by regulating MYH9. It may be a new potential target for liver cancer treatment.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas Nucleares , Humanos , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Neoplasias Hepáticas/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Nucleares/genética
5.
Exp Cell Res ; 421(2): 113377, 2022 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-36252649

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is the most common primary liver cancer in the world. In malignant liver cancer, the increase of aerobic glycolysis indicates that the possibility of tumorigenesis is greatly enhanced. TRIM37 is a member of the TRIM family of proteins that possesses E3 ubiquitin ligase activity and has been implicated in the occurrence and prognosis of many different tumors. However, the stability of P53 plays an important role in preventing tumorigenesis. The mechanism by which TRIM37 regulates the stability of P53 through ubiquitin in the progression of hepatocellular carcinoma is still unclear. MATERIALS AND METHODS: Real-time quantitative polymerase chain reaction (qRT-PCR) and Western blotting were used to detect the expression of mRNA and protein in HCC cells. Lactic acid production, glucose uptake, and ATP levels were measured by BioVision kit. The following were used to assess the in vitro function of TRIM37 in HCC cells: cell counting kit-8 (CCK-8), colony formation assay, cell migration and invasion assay, and flow cytometry. We observed the effect of TRIM37 on the growth of transplanted tumors in nude mice. Co-Immunoprecipitation (Co-IP) revealed a binding relationship between TRIM37 and P53. RESULTS: The expression of TRIM37 in hepatocellular carcinoma (HCC) tissues was higher than that of normal tissues according to an analysis of The Cancer Genome Atlas (TCGA) database.Loss-of-function assays indicated that TRIM37 inhibited the proliferation, colony formation, migration, and invasion of liver cancer cells. The mechanism is as follows: TRIM37 interacts with the P53 protein to induce E3 ligase activity, ubiquitination, and degradation, further promoting the malignant characteristics of hepatocellular carcinoma, thus promoting the process of glycolysis. Genetic knockdown of P53 reversed the promoting function of TRIM37 on the growth and metastasis of hepatocellular carcinoma cells. CONCLUSIONS: Our study showed that the TRIM37-P53 axis plays a role in the progression of liver cancer, and thus is a potential target for the treatment of liver cancer.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Ratones , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis , Neoplasias Hepáticas/patología , Ratones Desnudos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
Cell Prolif ; 55(9): e13273, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35748106

RESUMEN

As a member of the deoxyribonuclease 1 family, DNASE1L3 plays a significant role both inside and outside the cell. However, the role of DNASE1L3 in hepatocellular carcinoma (HCC) and its molecular basis remains to be further investigated. In this study, we report that DNASE1L3 is downregulated in clinical HCC samples and evaluate the relationship between its expression and HCC clinical features. In vivo and in vitro experiments showed that DNASE1L3 negatively regulates the proliferation, invasion and metastasis of HCC cells. Mechanistic studies showed that DNASE1L3 recruits components of the cytoplasmic ß-catenin destruction complex (GSK-3ß and Axin), promotes the ubiquitination degradation of ß-catenin, and inhibits its nuclear transfer, thus, decreasing c-Myc, P21 and P27 level. Ultimately, cell cycle and EMT signals are restrained. In general, this study provides new insight into the mechanism for HCC and suggests that DNASE1L3 can become a considerable target for HCC.


Asunto(s)
Carcinoma Hepatocelular , Endodesoxirribonucleasas/metabolismo , Neoplasias Hepáticas , beta Catenina/metabolismo , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Neoplasias Hepáticas/genética , Ubiquitina/metabolismo , Vía de Señalización Wnt
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA