Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Medicine (Baltimore) ; 103(32): e38924, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39121331

RESUMEN

This research endeavor seeks to explore the microenvironment of melanoma tumors and construct a prognostic model by focusing on genes specific to CD8+ T cells. The single-cell sequencing data of melanoma underwent processing with the Seurat package, subsequent to which cell communication network analysis was conducted using the iTALK package and transcription factor analysis was performed using the SCENIC package. Univariate COX and LASSO regression analyses were utilized to pinpoint genes linked to the prognosis of melanoma patients, culminating in the creation of a prognostic model through multivariate COX analysis. The model was validated using the GSE65904 and GSE35640 datasets. Multi-omics analysis was conducted utilizing the maftools, limma, edgeR, ChAMP, and clusterProfiler packages. The examination of single-cell sequencing data revealed the presence of 8 cell types, with the transcription factors RFXAP, CLOCK, MGA, RBBP, and ZNF836 exhibiting notably high expression levels in CD8+ T cells as determined by the SCENIC package. Utilizing these transcription factors and their associated target genes, a prognostic model was developed through COX and LASSO analyses, incorporating the genes GPR171, FAM174A, and BPI. This study validated the model with independent datasets and conducted additional analysis involving multi-omics and immune infiltration to identify a more favorable prognosis for patients in the low-risk group. The findings provide valuable insights into the tumor microenvironment of melanoma and establish a reliable prognostic model. The integration of multi-omics and immune infiltration analyses enhances our understanding of the pathogenesis of melanoma. The identification of specific genes holds promise as potential biomarkers for individuals with melanoma, serving as important indicators for predicting patient outcomes and determining their response to immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos , Biología Computacional , Melanoma , Microambiente Tumoral , Humanos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología , Melanoma/genética , Melanoma/patología , Melanoma/inmunología , Melanoma/mortalidad , Linfocitos T CD8-positivos/inmunología , Pronóstico , Biología Computacional/métodos , Masculino , Factores de Transcripción/genética , Femenino , Biomarcadores de Tumor/genética , Análisis de la Célula Individual
2.
Int J Mol Sci ; 25(13)2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-39000055

RESUMEN

Respiratory virus infections remain a significant challenge to human health and the social economy. The symptoms range from mild rhinitis and nasal congestion to severe lower respiratory tract dysfunction and even mortality. The efficacy of therapeutic drugs targeting respiratory viruses varies, depending upon infection time and the drug resistance engendered by a high frequency of viral genome mutations, necessitating the development of new strategies. The MAPK/ERK pathway that was well delineated in the 1980s represents a classical signaling cascade, essential for cell proliferation, survival, and differentiation. Since this pathway is constitutively activated in many cancers by oncogenes, several drugs inhibiting Raf/MEK/ERK have been developed and currently used in anticancer treatment. Two decades ago, it was reported that viruses such as HIV and influenza viruses could exploit the host cellular MAPK/ERK pathway for their replication. Thus, it would be feasible to repurpose this category of the pathway inhibitors for the treatment of respiratory viral infections. The advantage is that the host genes are not easy to mutate such that the drug resistance rarely occurs during short-period treatment of viruses. Therefore, in this review we will summarize the research progress on the role of the MAPK/ERK pathway in respiratory virus amplification and discuss the potential of the pathway inhibitors (MEK inhibitors) in the treatment of respiratory viral infections.


Asunto(s)
Reposicionamiento de Medicamentos , Sistema de Señalización de MAP Quinasas , Infecciones del Sistema Respiratorio , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/virología , Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Antivirales/uso terapéutico , Antivirales/farmacología , Animales , Inhibidores de Proteínas Quinasas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología
3.
Bioelectrochemistry ; 160: 108752, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38852384

RESUMEN

Replacing monopolar pulse with bipolar pulses of the same energized time can minimize unnecessary neurological side effects during irreversible electroporation (IRE). An improved neural excitation model that considers dynamic conductivity and thermal effects during brain tumor IRE ablation was proposed for the first time in this study. Nerve fiber excitation during IRE ablation by applying a monopolar pulse (100 µs) and a burst of bipolar pulses (energized time of 100 µs with both the sub-pulse length and interphase delay of 1 µs) was investigated. Our results suggest that both thermal effects and dynamic conductivity change the onset time of action potential (AP), and dynamic conductivity also changes the hyperpolarization amplitude. Considering both thermal effects and dynamic conductivity, the hyperpolarization amplitude in nerve fibers located 2 cm from the tumor center was reduced by approximately 23.8 mV and the onset time of AP was delayed by approximately 17.5 µs when a 500 V monopolar pulse was applied. Moreover, bipolar pulses decreased the excitable volume of brain tissue by approximately 68.8 % compared to monopolar pulse. Finally, bipolar pulses cause local excitation with lesser damage to surrounding healthy tissue in complete tumor ablation, demonstrating the potential benefits of bipolar pulses in brain tissue ablation.

4.
Comput Biol Med ; 177: 108678, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38833796

RESUMEN

Cells exposed to a pulsed electric field undergo electroporation(EP) and electrodeformation(ED) under electric field stress, and a coupled model of EP and ED of glioblastoma(GBM) taking into account Joule heating is proposed. The model geometry is extracted from real cell boundaries, and the effects of Joule heating-induced temperature rise on the EP and ED processes are considered. The results show that the temperature rise will increase the cell's local conductivity, leading to a decrease in the transmembrane potential(TMP). The temperature rise also causes a decrease in the dynamic Young's modulus of the cell membrane, making the cell less resistant to deformation. In addition, GBM cells are more susceptible to EP in the middle portion of the cell and ED in the three tentacle portions under pulsed electric fields, and the cells undergo significant positional shifts. The ED of the nucleus is similar to spherical cells, but the degree of ED is smaller.


Asunto(s)
Electroporación , Glioblastoma , Modelos Biológicos , Humanos , Electroporación/métodos , Calor , Línea Celular Tumoral , Neoplasias Encefálicas , Potenciales de la Membrana/fisiología , Membrana Celular
5.
Am J Cancer Res ; 14(3): 1052-1070, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38590405

RESUMEN

Melanoma is a common and fatal cutaneous malignancy with strong invasiveness and high mortality rate. Clinically, elderly melanoma patients tend to exhibit stronger invasion ability and poorer prognosis. Given the heterogeneity of tumors, we analyzed the prognosis and risk assessment of melanoma through aging-related genes rather than age stratification. FOXM1 and CCL4 were identified to be closely associated with melanoma prognosis. Single-cell transcriptome analysis showed that FOXM1 was significantly up-regulated in tumor cells, while CCL4 was markedly elevated in immune cells. A melanoma prognostic model was constructed based on the two independent prognostic factors. This model showed a high accuracy in predicting the mortality of melanoma patients over several years. The patients in low-risk group appeared to have more immune cell infiltration and better immune therapy efficacy. Cellular experiments showed that CCL4 could promote apoptosis of melanoma cells through immune cells, and apoptosis could regulate the expression of FOXM1. In addition, the results of the spatial transcriptome and immunohistochemistry suggested that CCL4 was highly expressed in macrophages and the expression of FOXM1 in melanoma cell was negatively correlated with immune cell infiltration, especially macrophages. Here, we established a novel prognostic model for melanoma, which showed promising predictive performance and may serve as a biomarker for the efficacy of immune checkpoint inhibition therapy in melanoma patients. In addition, we explored the function of two genes in the model in melanoma.

6.
J Biol Chem ; 300(4): 107208, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38521502

RESUMEN

Transforming growth factor-ß (TGF-ß) and Hippo signaling are two critical pathways engaged in cancer progression by regulating both oncogenes and tumor suppressors, yet how the two pathways coordinately exert their functions in the development of hepatocellular carcinoma (HCC) remains elusive. In this study, we firstly conducted an integrated analysis of public liver cancer databases and our experimental TGF-ß target genes, identifying CYR61 as a pivotal candidate gene relating to HCC development. The expression of CYR61 is downregulated in clinical HCC tissues and cell lines than that in the normal counterparts. Evidence revealed that CYR61 is a direct target gene of TGF-ß in liver cancer cells. In addition, TGF-ß-stimulated Smad2/3 and the Hippo pathway downstream effectors YAP and TEAD4 can form a protein complex on the promoter of CYR61, thereby activating the promoter activity and stimulating CYR61 gene transcription in a collaborative manner. Functionally, depletion of CYR61 enhanced TGF-ß- or YAP-mediated growth and migration of liver cancer cells. Consistently, ectopic expression of CYR61 was capable of impeding TGF-ß- or YAP-induced malignant transformation of HCC cells in vitro and attenuating HCC xenograft growth in nude mice. Finally, transcriptomic analysis indicates that CYR61 can elicit an antitumor program in liver cancer cells. Together, these results add new evidence for the crosstalk between TGF-ß and Hippo signaling and unveil an important tumor suppressor function of CYR61 in liver cancer.


Asunto(s)
Carcinoma Hepatocelular , Proteína 61 Rica en Cisteína , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas , Factor de Crecimiento Transformador beta , Proteínas Señalizadoras YAP , Animales , Humanos , Ratones , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/genética , Línea Celular Tumoral , Movimiento Celular , Proteína 61 Rica en Cisteína/metabolismo , Proteína 61 Rica en Cisteína/genética , Minería de Datos , Regulación Neoplásica de la Expresión Génica/genética , Vía de Señalización Hippo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/genética , Ratones Desnudos , Regiones Promotoras Genéticas , Transducción de Señal/genética , Proteína Smad2/metabolismo , Proteína Smad2/genética , Proteína smad3/metabolismo , Proteína smad3/genética , Factores de Transcripción de Dominio TEA/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/genética , Regulación hacia Arriba , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética
7.
Aging (Albany NY) ; 16(1): 911-927, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38217549

RESUMEN

OBJECTIVE: This study was conducted to screen out immune-related genes in connection with the prognosis of melanoma, construct a prognosis model and explore the relevant mechanisms. METHODS AND MATERIALS: 1973 genes associated with immune system were derived from the Immport database, and RNA-seq data of melanoma and information of patients were searched from the Xena database. Cox univariate analysis, Lasso analysis and Cox multivariate analysis were used to screen out six genes to construct the model. Then the risk scores were estimated for patients based on our constructed prognosis model. Estimate was used to affirm that the model was about immune infiltration, and CIBERSORT was used to screen out immune cells associated with prognosis. TIDE was applied to predict the efficacy of immunotherapy. Finally, GSE65904 and GSE19234 were used to confirm the effectiveness of the model. RESULTS: ADCYAP1R1, GPI, NTS might cause poor prognosis while IFITM1, KIR2DL4, LIF were more likely conductive to prognosis of melanoma patients and a model of prognosis was constructed on the basis of these six genes. The effectiveness of the model has been proven by the ROC curve, and the miRNAs targeting the screened genes were found out, suggesting that the immune system might impact on the prognosis of melanoma by T cell CD8+, T cell CD4+ memory and NK cells. CONCLUSIONS: In this study, the screened six genes were associated with the prognosis of melanoma, which was conductive to clinical prognostic prediction and individualized treatment strategy.


Asunto(s)
Melanoma , MicroARNs , Humanos , Melanoma/genética , Bases de Datos Factuales , Inmunoterapia , Células Asesinas Naturales , Pronóstico , Microambiente Tumoral/genética
8.
Mol Biol Rep ; 51(1): 203, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38270651

RESUMEN

BACKGROUND: Recovery from a foot ulcer is compromised in a diabetic status, due to the impaired tissue microenvironment that consists of altered inflammation, angiogenesis and fibrosis. Phenotypic alterations in both macrophages and fibroblasts have been detected in the diabetic wound. Recently, a fibroblast subpopulation that expresses high matrix metalloproteinase 1 (MMP1), MMP3, MMP11 and Chitinase-3-Like Protein 1 (CHI3L1) was associated with a successful diabetic wound healing. However, it is not known whether these healing-associated fibroblasts are regulated by macrophages. METHODS AND RESULTS: We used bioinformatic tools to analyze selected public databases on normal and diabetic skin from patients, and identified genes significantly altered in diabetes. In a mouse model for diabetic wound healing, we detected not only a loss of the spatiotemporal changes in interleukin 1ß (IL1ß), IL6, IL10 and vascular endothelial growth factor A (VEGF-A) in wound macrophages, but also a compromised expression of MMP1, MMP3, MMP11, CHI3L1 and VEGF-A in healing-associated wound fibroblasts in a diabetic status. Co-culture with diabetic macrophages significantly reduced the expression of MMP1, MMP3, MMP11, CHI3L1 and VEGF-A in fibroblasts from non-diabetic wound. Co-culture with non-diabetic macrophages or diabetic macrophages supplied with IL6 significantly increased the expression of MMP1, MMP3, MMP11, CHI3L1 and VEGF-A in fibroblasts from diabetic wound. Moreover, macrophage-specific expression of IL6 significantly improved wound healing and angiogenesis in diabetic mice. CONCLUSIONS: Macrophages may induce the activation of wound-healing-associated fibroblasts, while the defective macrophages in diabetes may be corrected with IL6 treatment as a promising therapy for diabetic foot disease.


Asunto(s)
Diabetes Mellitus Experimental , Factor A de Crecimiento Endotelial Vascular , Humanos , Animales , Ratones , Metaloproteinasa 3 de la Matriz , Metaloproteinasa 1 de la Matriz , Metaloproteinasa 11 de la Matriz , Interleucina-6 , Cicatrización de Heridas
9.
Int J Mol Sci ; 24(23)2023 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-38068934

RESUMEN

Cancer stem cells (CSCs) play a pivotal role in drug resistance and metastasis. Among the key players, Forkhead box O3a (FOXO3a) acts as a tumor suppressor. This study aimed to unravel the role of FOXO3a in mediating the inhibitory effect of metformin on cancer stemness derived from paclitaxel (PTX)-resistant non-small-cell lung cancer (NSCLC) cells. We showed that CSC-like features were acquired by the chronic induction of resistance to PTX, concurrently with inactivation of FOXO3a. In line with this, knockdown of FOXO3a in PTX-sensitive cells led to changes toward stemness, while overexpression of FOXO3a in PTX-resistant cells mitigated stemness in vitro and remarkably curbed the tumorigenesis of NSCLC/PTX cells in vivo. Furthermore, metformin suppressed the self-renewal ability of PTX-resistant cells, reduced the expression of stemness-related markers (c-MYC, Oct4, Nanog and Notch), and upregulated FOXO3a, events concomitant with the activation of AMP-activated protein kinase (AMPK). All these changes were recapitulated by silencing FOXO3a in PTX-sensitive cells. Intriguingly, the introduction of the AMPK dominant negative mutant offset the inhibitory effect of metformin on the stemness of PTX-resistant cells. In addition, FOXO3a levels were elevated by the treatment of PTX-resistant cells with MK2206 (an Akt inhibitor) and U0126 (a MEK inhibitor). Collectively, our findings indicate that metformin exerts its effect on FOXO3a through the activation of AMPK and the inhibition of protein kinase B (Akt) and MAPK/extracellular signal-regulated kinase (MEK), culminating in the suppression of stemness in paclitaxel-resistant NSCLC cells.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Metformina , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Metformina/farmacología , Metformina/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Línea Celular Tumoral , Quinasas de Proteína Quinasa Activadas por Mitógenos , Proliferación Celular , Resistencia a Antineoplásicos
11.
J Mol Cell Biol ; 15(2)2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-36806855

RESUMEN

Tea domain transcription factor 4 (TEAD4) plays a pivotal role in tissue development and homeostasis by interacting with Yes-associated protein (YAP) in response to Hippo signaling inactivation. TEAD4 and YAP can also cooperate with transforming growth factor-ß (TGF-ß)-activated Smad proteins to regulate gene transcription. Yet, it remains unclear whether TEAD4 plays a YAP-independent role in TGF-ß signaling. Here, we unveil a novel tumor suppressive function of TEAD4 in liver cancer via mitigating TGF-ß signaling. Ectopic TEAD4 inhibited TGF-ß-induced signal transduction, Smad transcriptional activity, and target gene transcription, consequently suppressing hepatocellular carcinoma cell proliferation and migration in vitro and xenograft tumor growth in mice. Consistently, depletion of endogenous TEAD4 by siRNAs enhanced TGF-ß signaling in cancer cells. Mechanistically, TEAD4 associates with receptor-regulated Smads (Smad2/3) and Smad4 in the nucleus, thereby impairing the binding of Smad2/3 to the histone acetyltransferase p300. Intriguingly, these negative effects of TEAD4 on TGF-ß/Smad signaling are independent of YAP, as impairing the TEAD4-YAP interaction through point mutagenesis or depletion of YAP and/or its paralog TAZ has little effect. Together, these results unravel a novel function of TEAD4 in fine tuning TGF-ß signaling and liver cancer progression in a YAP-independent manner.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Factores de Transcripción de Dominio TEA , Animales , Humanos , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Señalizadoras YAP
12.
Biomedicines ; 10(12)2022 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-36552019

RESUMEN

Prostate cancer is the most common cancer and one of the leading causes of cancer mortality in males. Androgen-deprivation therapy (ADT) is an effective strategy to inhibit tumour growth at early stages. However, 10~50% of cases are estimated to progress to metastatic castration-resistant prostate cancer (mCRPC) which currently lacks effective treatments. Clinically, salvage treatment measures, such as endocrine therapy and chemotherapy, are mostly used for advanced prostate cancer, but their clinical outcomes are not ideal. When the existing clinical therapeutic methods can no longer inhibit the development of advanced prostate cancer, human adenovirus (HAdV)-based gene therapy and viral therapy present promising effects. Pre-clinical studies have shown its powerful oncolytic effect, and clinical studies are ongoing to further verify its effect and safety in prostate cancer treatment. Targeting the prostate by HAdV alone or in combination with radiotherapy and chemotherapy sheds light on patients with castration-resistant and advanced prostate cancer. This review summarizes the advantages of oncolytic virus-mediated cancer therapy, strategies of HAdV modification, and existing preclinical and clinical investigations of HAdV-mediated gene therapy to further evaluate the potential of oncolytic adenovirus in prostate cancer treatment.

13.
Zhongguo Gu Shang ; 35(11): 1065-9, 2022 Nov 25.
Artículo en Chino | MEDLINE | ID: mdl-36415193

RESUMEN

OBJECTIVE: To analyze the value of procalcitonin (PCT) in the diagnosis of perioperative infection associated with implants in patients with primary hip arthroplasty. METHODS: A retrospective study was conducted on 150 patients who underwent primary hip arthroplasty from June 2018 to June 2020, including 86 males and 64 females, aged from 47 to 77 years old with an average of (57.04±7.43) years. All patients with primary hip arthroplasty were divided into infection group and non infection group according to whether there was infection after operation. Blood samples were collected from the elbow vein before operation (D0) and on the 4, 6, 8 days after operation(D4, D6 and D8) respectively to detect the serum PCT level and white blood cell count (WBC) level. RESULTS: Among 150 patients with primary hip arthroplasty, 34 patients with postoperative infection were in the infection group, and 116 patients without postoperative infection were in the noninfection group. In the infection group, there were 19 cases of superficial surgical site infection(55.88%, 19/34), 9 cases of urinary tract infection (26.47%, 9/34), and 6 cases of pneumonia(17.65%, 6/34). After bacterial culture in the infection group, there were 9 cases of Staphylococcus aureus, 3 cases of Escherichia coli, 3 cases of Staphylococcus epidermidis, 3 cases of Streptococcus constellation, 3 cases of Candida albicans, 6 cases of Klebsiella pneumoniae, 2 cases of Escherichia coli and Streptococcus agalactis, 3 cases of coagulase invisible staphylococcus and Burkholderia cepacia, 2 cases of Escherichia coli, Enterococcus faecalis and Pseudomonas aeruginosa. There was no significant difference in PCT levels between two groups in D0(P=0.081), D4(P=0.069) and D6(P=0.093), but there was significant difference in D8(P=0.007). There was no significant difference in WBC between two groups at any time point(P>0.05). The results of receiver operating characteristic curve(ROC) showed that the AUC of PCT diagnosis was 0.978[95%CI(0.933, 1.022)] and that of WBC was 0.562[95%CI(0.398, 0.726)], PCT was an important predictor of infection after primary hip arthroplasty(AUC>0.9). When the critical value was 0.526 ng/ml, the sensitivity and specificity of PCT diagnosis are 36% and 100%, respectively, WBC was not a significant predictor of infection after primary hip arthroplasty (0.5

Asunto(s)
Artroplastia de Reemplazo de Cadera , Polipéptido alfa Relacionado con Calcitonina , Masculino , Femenino , Humanos , Persona de Mediana Edad , Anciano , Calcitonina , Artroplastia de Reemplazo de Cadera/efectos adversos , Precursores de Proteínas , Estudios Retrospectivos , Proteína C-Reactiva/análisis , Péptido Relacionado con Gen de Calcitonina , Escherichia coli
14.
Int J Mol Sci ; 23(15)2022 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-35955931

RESUMEN

Phosphatidylethanolamine binding protein 4 (PEBP4) is an understudied multifunctional small protein. Previous studies have shown that the expression of PEBP4 is increased in many cancer specimens, which correlates to cancer progression. The present study explored the mechanism by which PEBP4 regulates the growth and progression of hepatocellular carcinoma cells. Thus, we showed that knockdown of PEBP4 in MHCC97H cells, where its expression was relatively high, diminished activities of serine/threonine protein kinase B (PKB, also known as Akt), mammalian target of rapamycin complex 1(mTORC1), and mTORC2, events that were not restored by insulin-like growth factor 1 (IGF-1). Conversely, overexpression of PEBP4 in MHCC97L cells with the low endogenous level yielded opposite effects. Furthermore, physical association of PEBP4 with Akt, mTORC1, and mTORC2 was observed. Interestingly, introduction of AktS473D mutant, bypassing phosphorylation by mTORC2, rescued mTORC1 activity, but without effects on mTORC2 signaling. In contrast, the effect of PEBP4 overexpression on the activity of mTORC1 but not that of mTORC2 was suppressed by MK2206, a specific inhibitor of Akt. In conjunction, PEBP4 knockdown-engendered reduction of cell proliferation, migration and invasion was partially rescued by Akt S473D while increases in these parameters induced by overexpression of PEBP4 were completely abolished by MK2206, although the expression of epithelial mesenchymal transition (EMT) markers appeared to be fully regulated by the active mutant of Akt. Finally, knockdown of PEBP4 diminished the growth of tumor and metastasis, whereas they were enhanced by overexpression of PEBP4. Altogether, our study suggests that increased expression of PEBP4 exacerbates malignant behaviors of hepatocellular cancer cells through cooperative participation of mTORC1 and mTORC2.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Carcinoma Hepatocelular/genética , Humanos , Neoplasias Hepáticas/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 2 de la Rapamicina/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
15.
Acta Biochim Biophys Sin (Shanghai) ; 54(8): 1140-1147, 2022 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-35880569

RESUMEN

Abnormal metabolism is a major hallmark of cancer and has been validated as a therapeutic target. Adenine monophosphate-activated protein kinase (AMPK), an αßγ heterotrimer, performs essential functions in cancer progression due to its central role in maintaining the homeostasis of cellular energy. While the contributions of AMPKα and AMPKγ subunits to cancer development have been established, specific roles of AMPKß1 and AMPKß2 isoforms in cancer development are poorly understood. Here, we show the functions of AMPKß1 and AMPKß2 in colon cancer. Specifically, deletion of AMPKß1 or AMPKß2 leads to increased cell proliferation, colony formation, migration, and tumorigenesis in HCT116 and HT29 colon cancer cells. Interestingly, the AMPKß1 and AMPKß2 isoforms have slightly different effects on regulating cancer metabolism, as colon cancer cells with AMPKß1 knockout showed decreased rates of glycolysis-related oxygen consumption, while AMPKß2 deletion led to enhanced rates of oxygen consumption due to oxidative phosphorylation. These results demonstrate that functional AMPKß1 and AMPKß2 inhibit growth and tumorigenesis in colon cancer cells, suggesting their potential as effective targets for colon cancer therapy.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Neoplasias del Colon , Proteínas Quinasas Activadas por AMP/genética , Carcinogénesis/genética , Transformación Celular Neoplásica , Neoplasias del Colon/genética , Humanos , Isoformas de Proteínas
16.
Int J Mol Sci ; 23(9)2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35563547

RESUMEN

The Ras-Raf-MEK-ERK signaling pathway, the first well-established MAPK pathway, plays essential roles in cell proliferation, survival, differentiation and development. It is activated in over 40% of human cancers owing to mutations of Ras, membrane receptor tyrosine kinases and other oncogenes. The Raf family consists of three isoforms, A-Raf, B-Raf and C-Raf. Since the first discovery of a truncated mutant of C-Raf as a transforming oncogene carried by a murine retrovirus, forty years of extensive studies have provided a wealth of information on the mechanisms underlying the activation, regulation and biological functions of the Raf family. However, the mechanisms by which activation of A-Raf and C-Raf is accomplished are still not completely understood. In contrast, B-Raf can be easily activated by binding of Ras-GTP, followed by cis-autophosphorylation of the activation loop, which accounts for the fact that this isoform is frequently mutated in many cancers, especially melanoma. The identification of oncogenic B-Raf mutations has led to accelerated drug development that targets Raf signaling in cancer. However, the effort has not proved as effective as anticipated, inasmuch as the mechanism of Raf activation involves multiple steps, factors and phosphorylation of different sites, as well as complex interactions between Raf isoforms. In this review, we will focus on the physiological complexity of the regulation of Raf kinases and their connection to the ERK phosphorylation cascade and then discuss the role of Raf in tumorigenesis and the clinical application of Raf inhibitors in the treatment of cancer.


Asunto(s)
Neoplasias , Transducción de Señal , Animales , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Fosforilación , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas B-raf/genética
17.
BMC Infect Dis ; 21(1): 200, 2021 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-33618678

RESUMEN

BACKGROUND: Coronavirus disease 2019 (COVID-19) is an infectious disease characterized by cough, fever, and fatigue and 20% of cases will develop into severe conditions resulting from acute lung injury with the manifestation of the acute respiratory distress syndrome (ARDS) that accounts for more than 50% of mortality. Currently, it has been reported that some comorbidities are linked with an increased rate of severity and mortality among COVID-19 patients. To assess the role of comorbidity in COVID-19 progression, we performed a systematic review with a meta-analysis on the relationship of COVID-19 severity with 8 different underlying diseases. METHODS: PubMed, Web of Science, and CNKI were searched for articles investigating the prevalence of comorbidities in severe and non-severe COVID-19 patients. A total of 41 studies comprising 12,526 patients were included. RESULTS: Prevalence of some commodities was lower than that in general population such as hypertension (19% vs 23.2%), diabetes (9% vs 10.9%), chronic kidney disease (CKD) (2% vs 9.5%), chronic liver diseases (CLD) (3% vs 24.8%) and chronic obstructive pulmonary disease (COPD) (3% vs 8.6%), while some others including cancer (1% vs 0.6%), cardiovascular disease (6% vs 1.8%) and cerebrovascular disease (2% vs 0.9%) exhibited greater percentage in COVID-19. Cerebrovascular disease (OR = 3.70, 95%CI 2.51-5.45) was found to be the strongest risk factor in disease exacerbation, followed by CKD (OR = 3.60, 95%CI 2.18-5.94), COPD (OR = 3.14, 95% CI 2.35-4.19), cardiovascular disease (OR = 2.76, 95% CI 2.18-3.49), malignancy (OR = 2.63, 95% CI 1.75-3.95), diabetes (OR = 2.49, 95% CI 2.10-2.96) and hypertension (OR = 2.13, 95% CI 1.81-2.51). We found no correlation between CLD and increased disease severity (OR = 1.32, 95% CI 0.96-1.82). CONCLUSION: The impact of all eight underlying diseases on COVID-19 deterioration seemed to be higher in patients outside Hubei. Based on different comorbidities, COVID-19 patients tend to be at risk of developing poor outcomes to a varying degree. Thus, tailored infection prevention and monitoring and treatment strategies targeting these high-risk subgroups might improve prognosis during the COVID-19 pandemic.


Asunto(s)
COVID-19/complicaciones , SARS-CoV-2 , COVID-19/epidemiología , Enfermedades Cardiovasculares/epidemiología , Trastornos Cerebrovasculares/epidemiología , China/epidemiología , Comorbilidad , Diabetes Mellitus/epidemiología , Humanos , Neoplasias/epidemiología , Enfermedad Pulmonar Obstructiva Crónica/epidemiología , Insuficiencia Renal Crónica/epidemiología , Factores de Riesgo
18.
J Cell Mol Med ; 25(6): 2806-2815, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33538080

RESUMEN

We have previously shown that adenine monophosphate-activated protein kinase (AMPK) regulates transforming growth factor ß (TGF-ß)-triggered Smad3 phosphorylation. Here we report that AMPK inhibits TGF-ß1 production. First, metformin reduced mRNA levels of TGF-ß1 in gastric cancer cells, in parallel to the decrease of its protein abundance. The effects were more prominent in the cells containing LKB1, an upstream kinase of AMPK. Second, knockdown of Smad3 by siRNA abrogated the expression of TGF-ß1. Third, metformin suppressed firefly luciferase activity whose transcription was driven by TGF-ß1 promoter. In accordance, deletion of the putative binding site of Smad3 in the TGF-ß1 promoter region severely impaired the promoter activity and response to metformin. Fourth, in support of our in vitro study, clinical treatment of type 2 diabetes with metformin significantly reduced the plasma level of TGF-ß1. Finally, immunohistochemical studies revealed that TGF-ß1 was highly expressed in human gastric cancer tissues as compared with adjacent normal tissues. In contrast, p-AMPK exhibited opposite changes. Furthermore, the survival rate of gastric cancer patients was positively correlated with p-AMPK and negative with TGF-ß1. Therefore, our present studies depict a mechanism underlying AMPK suppression of TGF-ß1 autoinduction, which is mediated through inhibition of Smad3 phosphorylation and activation. Collectively, our study sheds a light on the potential usage of AMPK activators in the treatment of TGF-ß1-mediated gastric cancer progression.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína smad3/metabolismo , Neoplasias Gástricas/etiología , Neoplasias Gástricas/metabolismo , Factor de Crecimiento Transformador beta1/genética , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Metformina/farmacología , Regiones Promotoras Genéticas , Neoplasias Gástricas/patología , Factor de Crecimiento Transformador beta1/sangre , Factor de Crecimiento Transformador beta1/metabolismo
19.
J Genet Genomics ; 47(9): 497-512, 2020 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-33339765

RESUMEN

Hepatocellular carcinoma (HCC) is the major form of primary liver cancer and one of the most prevalent and life-threatening malignancies globally. One of the hallmarks in HCC is the sustained cell survival and proliferative signals, which are determined by the balance between oncogenes and tumor suppressors. Transforming growth factor beta (TGF-ß) is an effective growth inhibitor of epithelial cells including hepatocytes, through induction of cell cycle arrest, apoptosis, cellular senescence, or autophagy. The antitumorigenic effects of TGF-ß are bypassed during liver tumorigenesis via multiple mechanisms. Furthermore, along with malignant progression, TGF-ß switches to promote cancer cell survival and proliferation. This dichotomous nature of TGF-ß is one of the barriers to therapeutic targeting in liver cancer. Thereafter, understanding the underlying molecular mechanisms is a prerequisite for discovering novel antitumor drugs that may specifically disable the growth-promoting branch of TGF-ß signaling or restore its tumor-suppressive arm. This review summarizes how TGF-ß inhibits or promotes liver cancer cell survival and proliferation, highlighting the functional switch mechanisms during the process.


Asunto(s)
Carcinoma Hepatocelular/genética , Proliferación Celular/genética , Neoplasias Hepáticas/genética , Factor de Crecimiento Transformador beta1/genética , Apoptosis/genética , Carcinogénesis/genética , Carcinoma Hepatocelular/patología , Supervivencia Celular/genética , Senescencia Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Hepáticas/patología
20.
Proc Natl Acad Sci U S A ; 117(14): 8013-8021, 2020 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-32193335

RESUMEN

AMP-activated protein kinase (AMPK) functions as an energy sensor and is pivotal in maintaining cellular metabolic homeostasis. Numerous studies have shown that down-regulation of AMPK kinase activity or protein stability not only lead to abnormality of metabolism but also contribute to tumor development. However, whether transcription regulation of AMPK plays a critical role in cancer metastasis remains unknown. In this study, we demonstrate that AMPKα1 expression is down-regulated in advanced human breast cancer and is associated with poor clinical outcomes. Transcription of AMPKα1 is inhibited on activation of PI3K and HER2 through ΔNp63α. Ablation of AMPKα1 expression or inhibition of AMPK kinase activity leads to disruption of E-cadherin-mediated cell-cell adhesion in vitro and increased tumor metastasis in vivo. Furthermore, restoration of AMPKα1 expression significantly rescues PI3K/HER2-induced disruption of cell-cell adhesion, cell invasion, and cancer metastasis. Together, these results demonstrate that the transcription control is another layer of AMPK regulation and suggest a critical role for AMPK in regulating cell-cell adhesion and cancer metastasis.


Asunto(s)
Proteínas Quinasas Activadas por AMP/genética , Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Mama/patología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/genética , Línea Celular Tumoral , Cromonas/farmacología , Supervivencia sin Enfermedad , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Lapatinib/farmacología , Ratones , Morfolinas/farmacología , Estadificación de Neoplasias , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Pronóstico , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Análisis de Matrices Tisulares , Transcripción Genética/efectos de los fármacos , Activación Transcripcional , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA