Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.402
Filtrar
1.
Sci Rep ; 14(1): 20794, 2024 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-39242716

RESUMEN

Cytokine-induced apoptosis inhibitor 1 (CIAPIN1) is a protein that regulates apoptosis and programmed cell death. This research aims to evaluate its potential role in inhibiting breast cancer cell proliferation, migration, and glycolysis and uncover its underlying molecular mechanism. We collected breast cancer tissue samples from eight patients between January 2019 and June 2023 in our Hospital to analyse CIAPIN1 expression. We transfected human breast cancer cell lines (MCF7, MDA-MB-231, MDA-MB-453, and MDA-MB-468) with siRNA of CIAPIN1. Finally, we determined protein expression using RT-qPCR and Western blotting. CIAPIN1 expression was elevated in both breast cancer tissue and serum. Overexpression of CIAPIN1 detected in the breast cancer cell lines MCF7 and MDA-MB-468. In addition, CIAPIN1 overexpression increased cell proliferation and migration rate. CIAPIN1 downregulation suppressed cell proliferation while elevated cellular apoptosis, reactive oxygen species (ROS) production and oxidative stress in breast cancer cells. Moreover, CIAPIN1 inhibition remarkably suppressed pyruvate, lactate and adenosine triphosphate (ATP) production and reduced the pyruvate kinase M2 (PKM2) protein expression and phosphorylation of signal transducer and activator of transcription 3 (STAT3) in breast cancer cells. Downregulation of CIAPIN1 suppresses cell proliferation, migration and glycolysis capacity in breast cancer cells by inhibiting the STAT3/PKM2 pathway.


Asunto(s)
Neoplasias de la Mama , Movimiento Celular , Proliferación Celular , Regulación hacia Abajo , Glucólisis , Péptidos y Proteínas de Señalización Intracelular , Factor de Transcripción STAT3 , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Glucólisis/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Movimiento Celular/genética , Femenino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Transducción de Señal , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Apoptosis/genética , Células MCF-7 , Especies Reactivas de Oxígeno/metabolismo
2.
J Cell Mol Med ; 28(17): e70053, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39224032

RESUMEN

MitoAMPK was proved to inhibit the Warburg effect, but the specific mechanisms on non-small-cell lung cancer remain unclear. Here, we selected SIRT6 and MZF1 to clarify the mechanism. By western blotting, quantitative polymerase chain reaction, the CCK-8 assay, and immunohistochemistry assays, we found SIRT6 expression was lower in NSCLC tissues and cell lines than normal tissues and cells. Moreover, SIRT6 could inhibit the Warburg effect by regulating glycolysis-related genes of SLC2A2, SLC2A4 and PKM2. Finally, we demonstrated the interaction between SIRT6 and MZF1 using ChIP-qPCR. In conclusion, mitoAMPK inhibits the Warburg effect by regulating the expression of the MZF1-SIRT6 complex.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción de Tipo Kruppel , Neoplasias Pulmonares , Sirtuinas , Efecto Warburg en Oncología , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/metabolismo , Sirtuinas/metabolismo , Sirtuinas/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Línea Celular Tumoral , Glucólisis/genética , Femenino , Masculino
3.
Cell Death Dis ; 15(9): 645, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39227375

RESUMEN

lncRNA can regulate tumorigenesis development and distant metastasis of colorectal cancer (CRC). However, the detailed molecular mechanisms are still largely unknown. Using RNA-sequencing data, RT-qPCR, and FISH assay, we found that HIF1A-AS2 was upregulated in CRC tissues and associated with poor prognosis. Functional experiments were performed to determine the roles of HIF1A-AS2 in tumor progression and we found that HIF1A-AS2 can promote the proliferation, metastasis, and aerobic glycolysis of CRC cells. Mechanistically, HIF1A-AS2 can promote FOXC1 expression by sponging miR-141-3p. SP1 can transcriptionally activate HIF1A-AS2. Further, HIF1A-AS2 can be packaged into exosomes and promote the malignant phenotype of recipient tumor cells. Taken together, we discovered that SP1-induced HIF1A-AS2 can promote the metabolic reprogramming and progression of CRC via miR-141-3p/FOXC1 axis. HIF1A-AS2 is a promising diagnostic marker and treatment target in CRC.


Asunto(s)
Neoplasias Colorrectales , Progresión de la Enfermedad , Factores de Transcripción Forkhead , Regulación Neoplásica de la Expresión Génica , MicroARNs , Humanos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Animales , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Línea Celular Tumoral , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ratones , Ratones Desnudos , Proliferación Celular/genética , Factor de Transcripción Sp1/metabolismo , Factor de Transcripción Sp1/genética , Glucólisis/genética , Ratones Endogámicos BALB C , Masculino , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Movimiento Celular/genética , Reprogramación Metabólica
5.
Cell Mol Biol Lett ; 29(1): 106, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39095708

RESUMEN

BACKGROUND: The RNA N6-methyladenosine (m6A) modification has become an essential hotspot in epigenetic modulation. Serine-arginine protein kinase 1 (SRPK1) is associated with the pathogenesis of various cancers. However, the m6A modification of SRPK1 and its association with the mechanism of in lung adenocarcinoma (LUAD) remains unclear. METHODS: Western blotting and polymerase chain reaction (PCR) analyses were carried out to identify gene and protein expression. m6A epitranscriptomic microarray was utilized to the assess m6A profile. Loss and gain-of-function assays were carried out elucidate the impact of METTL3 and SRPK1 on LUAD glycolysis and tumorigenesis. RNA immunoprecipitation (RIP), m6A RNA immunoprecipitation (MeRIP), and RNA stability tests were employed to elucidate the SRPK1's METTL3-mediated m6A modification mechanism in LUAD. Metabolic quantification and co-immunoprecipitation assays were applied to investigate the molecular mechanism by which SRPK1 mediates LUAD metabolism. RESULTS: The epitranscriptomic microarray assay revealed that SRPK1 could be hypermethylated and upregulated in LUAD. The main transmethylase METTL3 was upregulated and induced the aberrant high m6A levels of SRPK1. Mechanistically, SRPK1's m6A sites were directly methylated by METTL3, which also stabilized SRPK1 in an IGF2BP2-dependent manner. Methylated SRPK1 subsequently promoted LUAD progression through enhancing glycolysis. Further metabolic quantification, co-immunoprecipitation and western blot assays revealed that SRPK1 interacts with hnRNPA1, an important modulator of PKM splicing, and thus facilitates glycolysis by upregulating PKM2 in LUAD. Nevertheless, METTL3 inhibitor STM2457 can reverse the above effects in vitro and in vivo by suppressing SRPK1 and glycolysis in LUAD. CONCLUSION: It was revealed that in LUAD, aberrantly expressed METTL3 upregulated SRPK1 levels via an m6A-IGF2BP2-dependent mechanism. METTL3-induced SRPK1 fostered LUAD cell proliferation by enhancing glycolysis, and the small-molecule inhibitor STM2457 of METTL3 could be an alternative novel therapeutic strategy for individuals with LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Adenosina , Glucólisis , Neoplasias Pulmonares , Metiltransferasas , Proteínas Serina-Treonina Quinasas , Humanos , Adenosina/análogos & derivados , Adenosina/metabolismo , Glucólisis/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adenocarcinoma del Pulmón/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Metiltransferasas/metabolismo , Metiltransferasas/genética , Animales , Regulación Neoplásica de la Expresión Génica , Ratones , Línea Celular Tumoral , Ratones Desnudos , Empalme del ARN/genética , Proteínas de Unión a Hormona Tiroide , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Proliferación Celular/genética
6.
Int J Med Sci ; 21(10): 1976-1989, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39113895

RESUMEN

Aortic aneurysm and dissection (AD) represent a critical cardiovascular emergency with an alarmingly high mortality rate. Recent research has spotlighted the overexpression of genes associated with the m6A modification in AD patients, linking them to the presence of inflammatory M1-type macrophages. Moreover, glycolysis is widely recognized as a key feature of inflammatory M1-type macrophages, but biomarkers linking glycolysis and macrophage function to promote disease progression in AD have not been reported. We conducted an analysis of aortic immune cell infiltration, macrophages, and m6A-related biomarkers in AD patients using bioinformatics techniques. Subsequently, we employed a combination of RT-PCR, WB, and immunofluorescence assays to elucidate the alterations in the expression of M1- and M2-type macrophages, as well as markers of glycolysis, following the overexpression of key biomarkers. These findings were further validated in vivo through the creation of a rat model of AD with knockdown of the aforementioned key biomarkers. The findings revealed that the m6A-modified related gene RBM15 exhibited heightened expression in AD samples and was correlated with macrophage polarization. Upon overexpression of RBM15 in macrophages, there was an observed increase in the expression of M1-type macrophage markers CXCL9 and CXCL10, alongside a decrease in the expression of M2-type macrophage markers CCL13 and MRC1. Furthermore, there was an elevation in the expression of glycolytic enzymes GLUT1 and Hexokinase, as well as HIF1α, GAPDH, and PFKFB3 after RBM15 overexpression. Moreover, in vivo knockdown of RBM15 led to an amelioration of aortic aneurysm in the rat AD model. This knockdown also resulted in a reduction of the M1-type macrophage marker iNOS, while significantly increasing the expression of the M2-type macrophage marker CD206. In conclusion, our findings demonstrate that RBM15 upregulates glycolysis in macrophages, thus contributing to the progression of AD through the promotion of M1-type macrophage polarization. Conversely, downregulation of RBM15 suppresses M1-type macrophage polarization, thereby decelerating the advancement of AD. These results unveil potential novel targets for the treatment of AD.


Asunto(s)
Aneurisma de la Aorta , Disección Aórtica , Progresión de la Enfermedad , Glucólisis , Macrófagos , Proteínas de Unión al ARN , Glucólisis/genética , Humanos , Animales , Macrófagos/metabolismo , Macrófagos/inmunología , Ratas , Disección Aórtica/patología , Disección Aórtica/genética , Disección Aórtica/metabolismo , Aneurisma de la Aorta/metabolismo , Aneurisma de la Aorta/genética , Aneurisma de la Aorta/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Masculino , Modelos Animales de Enfermedad , Quimiocina CXCL10/metabolismo , Quimiocina CXCL10/genética , Biomarcadores/metabolismo , Quimiocina CXCL9/metabolismo , Quimiocina CXCL9/genética , Femenino , Adenosina/análogos & derivados
7.
Nat Commun ; 15(1): 6755, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39117659

RESUMEN

Histone lysine methyltransferase 2D (KMT2D) is the most frequently mutated epigenetic modifier in head and neck squamous cell carcinoma (HNSCC). However, the role of KMT2D in HNSCC tumorigenesis and whether its mutations confer any therapeutic vulnerabilities remain unknown. Here we show that KMT2D deficiency promotes HNSCC growth through increasing glycolysis. Additionally, KMT2D loss decreases the expression of Fanconi Anemia (FA)/BRCA pathway genes under glycolytic inhibition. Mechanistically, glycolytic inhibition facilitates the occupancy of KMT2D to the promoter/enhancer regions of FA genes. KMT2D loss reprograms the epigenomic landscapes of FA genes by transiting their promoter/enhancer states from active to inactive under glycolytic inhibition. Therefore, combining the glycolysis inhibitor 2-DG with DNA crosslinking agents or poly (ADP-ribose) polymerase (PARP) inhibitors preferentially inhibits tumor growth of KMT2D-deficient mouse HNSCC and patient-derived xenografts (PDXs) harboring KMT2D-inactivating mutations. These findings provide an epigenomic basis for developing targeted therapies for HNSCC patients with KMT2D-inactivating mutations.


Asunto(s)
Glucólisis , Carcinoma de Células Escamosas de Cabeza y Cuello , Animales , Humanos , Ratones , Glucólisis/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Línea Celular Tumoral , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteína BRCA1/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/deficiencia , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proteína BRCA2/deficiencia , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/patología , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Regulación Neoplásica de la Expresión Génica , Ensayos Antitumor por Modelo de Xenoinjerto , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Femenino , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transducción de Señal , Regiones Promotoras Genéticas/genética , Proteína de la Leucemia Mieloide-Linfoide
8.
BMC Cancer ; 24(1): 979, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39118022

RESUMEN

BACKGROUND: Gastric cancer (GC) is a major contributor to cancer-related mortality. Glycolysis plays a pivotal role in tumor microenvironment (TME) reprogramming. In this research, the functions of glycolysis-associated genes (GRGs) were evaluated to predict the outcome and reveal the characteristics of the immune microenvironment in individuals with stomach cancer. METHODS: The Cancer Genome Atlas (TCGA)-stomach adenocarcinoma (STAD) cohort provided gene expression and clinical data for gastric cancer (GC) patients, which were further authenticated using datasets sourced from the Gene Expression Omnibus (GEO). By referencing the Molecular Signatures Database (MSigDB), a total of 326 GRGs were pinpointed. The various subtypes of GC were outlined through consensus clustering, derived from the expression patterns of these GRGs. Utilizing multivariate Cox regression analysis, a multigene risk score model was formulated. Both the CIBERSORT and ESTIMATE algorithms played a pivotal role in assessing the immune microenvironment. To delve into the biological functions of the key genes, wound healing, transwell invasion, and MTT assays were conducted. RESULTS: Based on the expression patterns of GRGs, patients were categorized into two distinct groups: the metabolic subtype, designated as cluster A, and the immune subtype, labeled as cluster B. Patients belonging to cluster B exhibited a poorer prognosis. A prognostic risk score model, formulated upon the expression levels of six key GRGs - ME1, PLOD2, NUP50, CXCR4, SLC35A3, and SRD35A3 - emerged as a viable tool for predicting patient outcomes. The downregulation of CXCR4 notably diminished the glycolytic capacity of gastric cancer (GC) cells, alongside their migratory, invasive, and proliferative capabilities. Intriguingly, despite the adverse prognostic implications associated with both the immune subtype (cluster B) and the high-risk cohort, these groups exhibited a favorable immune microenvironment coupled with elevated expression of immune checkpoint genes. Our investigations revealed a positive correlation between high CXCR4 expression and low ME1 expression with the infiltration of CD8+ T cells, as well as an enhanced responsiveness to treatment with an anti-PD-1 immune checkpoint inhibitor. CONCLUSIONS: In this study, we discovered that the expression profiles of GRGs hold the potential to forecast the prognosis of gastric cancer (GC) patients, thereby possibly aiding in clinical treatment decision-making.


Asunto(s)
Glucólisis , Neoplasias Gástricas , Microambiente Tumoral , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología , Neoplasias Gástricas/patología , Neoplasias Gástricas/mortalidad , Humanos , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Pronóstico , Glucólisis/genética , Regulación Neoplásica de la Expresión Génica , Masculino , Biomarcadores de Tumor/genética , Femenino , Perfilación de la Expresión Génica , Persona de Mediana Edad , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Línea Celular Tumoral
9.
PeerJ ; 12: e17861, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39119106

RESUMEN

Background: As a heterogeneous malignancy, breast cancer (BRCA) shows high incidence and mortality. Discovering novel molecular markers and developing reliable prognostic models may improve the survival of BCRA. Methods: The RNA-seq data of BRCA patients were collected from the training set The Cancer Genome Atlas (TCGA)-BRCA and validation set GSE20685 in the Gene Expression Omnibus (GEO) databases. The "GSVA" R package was used to calculate the glycolysis score for each patient, based on which all the patients were divided into different glycolysis groups. The "limma" package was employed to perform differentially expression genes (DEGs) analysis. Key signature genes were selected by performing un/multivariate and least absolute shrinkage and selection operator (LASSO) C regression and used to develop a RiskScore model. The ESTIMATE and MCP-Counter algorithms were used for quantifying immune infiltration level. The functions of the genes were validated using Western blot, colony formation, transwell and wound-healing assay. Results: The glycolysis score and prognostic analysis showed that high glycolysis score was related to tumorigenesis pathway and a poor prognosis in BRCA as overactive glycolysis inhibited the normal functions of immune cells. Subsequently, we screened five key prognostic genes using the LASSO Cox regression analysis and used them to establish a RiskScore with a high classification efficiency. Based on the results of the RiskScore, it was found that patients in the high-risk group had significantly unfavorable immune infiltration and prognostic outcomes. A nomogram integrating the RiskScore could well predict the prognosis for BRCA patients. Knockdown of PSCA suppressed cell proliferation, invasion and migration of BRCA cells. Conclusion: This study developed a glycolysis-related signature with five genes to distinguish between high-risk and low-risk BRCA patients. A nomogram developed on the basis of the RiskScore was reliable to predict BRCA survival. Our model provided clinical guidance for the treatment of BRCA patients.


Asunto(s)
Neoplasias de la Mama , Glucólisis , Humanos , Glucólisis/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/mortalidad , Femenino , Pronóstico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Perfilación de la Expresión Génica
10.
Front Biosci (Landmark Ed) ; 29(8): 308, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39206892

RESUMEN

BACKGROUND: Aerobic glycolysis and the cell cycle are well-established tumor hallmarks. Understanding their relationship could help to unravel the pathogenic mechanisms of breast cancer (BC) and suggest potential new strategies for treatment. METHODS: Glycolysis-related genes (GRGs) were downloaded from the Reactome database and screened using univariate Cox analysis. The consensus clustering method was employed to identify a glycolytic activity signature (GAS) using the Gene Expression Omnibus (GEO) dataset. A nomogram risk prediction model was constructed using coefficients from univariate Cox analysis. Immune cell infiltration was evaluated using single-sample gene set enrichment analysis (ssGSEA) and the ESTIMATE algorithm. Gene co-expression modules were created using weighted correlation network analysis (WGCNA) to identify hub genes. Gene expression in three BC cell lines was quantified using Quantitative Reverse Transcriptase Polymera (qRT-PCR). Single-cell RNA sequencing (scRNA-seq) data was used to examine the relationship between GAS and hub genes. The sensitivity of different groups to cell cycle-related clinical drugs was also examined. RESULTS: BC with high GAS (HGAS) showed high tumor grade and recurrence rate. HGAS was a prognostic indicator of worse overall survival (OS) in BC patients. HGAS BC showed more abundant immune cells and significantly higher expression of immunomodulators compared to BC with low GAS (LGAS). HGAS BC also showed enhanced cell cycle pathway, with high mRNA and protein expression levels of Cyclin B2 (CCNB2), a key component of the cell cycle pathway. Importantly, scRNA-seq analysis revealed that elevated CCNB2 expression was positively correlated with HGAS in triple-negative BC (TNBC). This was validated in clinical samples from TNBC patients. High expression of CCNB2 was found in three BC cell lines, and was also an indicator of poor prognosis. HGAS BC showed high sensitivity to several cell cycle-related clinical drugs, with 9 of these also showing activity in BC with high CCNB2 expression. CONCLUSIONS: HGAS was associated with enhanced cell cycle pathway and immune activity in BC. These results suggest that CCNB2 is a potential key therapeutic target in BC patients.


Asunto(s)
Ciclina B2 , Regulación Neoplásica de la Expresión Génica , Glucólisis , Neoplasias de la Mama Triple Negativas , Humanos , Glucólisis/genética , Femenino , Ciclina B2/genética , Ciclina B2/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Línea Celular Tumoral , Pronóstico , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Ciclo Celular/genética , Perfilación de la Expresión Génica/métodos , Nomogramas
11.
Nat Commun ; 15(1): 6915, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39134530

RESUMEN

Protein post-translational modifications (PTMs) are crucial for cancer cells to adapt to hypoxia; however, the functional significance of lysine crotonylation (Kcr) in hypoxia remains unclear. Herein we report a quantitative proteomics analysis of global crotonylome under normoxia and hypoxia, and demonstrate 128 Kcr site alterations across 101 proteins in MDA-MB231 cells. Specifically, we observe a significant decrease in K131cr, K156cr and K220cr of phosphoglycerate kinase 1 (PGK1) upon hypoxia. Enoyl-CoA hydratase 1 (ECHS1) is upregulated and interacts with PGK1, leading to the downregulation of PGK1 Kcr under hypoxia. Abolishment of PGK1 Kcr promotes glycolysis and suppresses mitochondrial pyruvate metabolism by activating pyruvate dehydrogenase kinase 1 (PDHK1). A low PGK1 K131cr level is correlated with malignancy and poor prognosis of breast cancer. Our findings show that PGK1 Kcr is a signal in coordinating glycolysis and the tricarboxylic acid (TCA) cycle and may serve as a diagnostic indicator for breast cancer.


Asunto(s)
Neoplasias de la Mama , Ciclo del Ácido Cítrico , Glucólisis , Fosfoglicerato Quinasa , Fosfoglicerato Quinasa/metabolismo , Fosfoglicerato Quinasa/genética , Humanos , Glucólisis/genética , Línea Celular Tumoral , Femenino , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Lisina/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Regulación hacia Abajo , Ratones , Proteómica/métodos , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica , Mitocondrias/metabolismo , Hipoxia de la Célula , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética
12.
Discov Med ; 36(187): 1692-1702, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39190384

RESUMEN

BACKGROUND: Cutaneous squamous cell carcinoma (cSCC) is a fatal disease characterized by metabolic dysregulation. The role of ephrin type-B receptor 2 (ephrin-B2), a crucial molecule in cancer cell biology, in regulating glycolysis and cell proliferation of cSCC is not well understood. This study aimed to investigate the biological pathways by which ephrin-B2 impacts the glycolysis and cell proliferation of cSCC. METHODS: Ephrin-B2 expression levels in cSCC were determined using quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting. Ephrin-B2 expression in cSCC cells was manipulated using overexpression and knockdown approaches. A series of in vitro assays, such as cell counting kit-8 (CCK-8), Transwell assay, immunofluorescence assay, enzyme-linked immunosorbent assay (ELISA), qRT-PCR, and Western blotting, were employed to delineate the biological roles of ephrin-B2/pyruvate kinase muscle isoenzyme 2 (PKM2)/hypoxia-inducible factor 1 alpha (HIF-1α) in proliferation, migration, invasion, and glucose metabolism of cSCC. RESULTS: This study highlights an upregulation of ephrin-B2 expression in cSCC. Knockdown of ephrin-B2 significantly suppressed the proliferation, migration, invasion, and glucose metabolism of cSCC cells. Moreover, ephrin-B2 expression was upregulated under hypoxic conditions. At the molecular level, ephrin-B2 knockdown resulted in the downregulation of PKM2 and HIF-1α expression. Additionally, the overexpression of PKM2 or HIF-1α successfully rescued the diminished proliferation, migration, invasion and glucose metabolism induced by ephrin-B2 knockdown in cSCC cells. CONCLUSION: These findings suggest that ephrin-B2 suppression may hinder cSCC cell proliferation and glycolytic metabolism, potentially via the PKM2/HIF-1α axis modulation.


Asunto(s)
Carcinoma de Células Escamosas , Proteínas Portadoras , Proliferación Celular , Efrina-B2 , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia , Proteínas de la Membrana , Neoplasias Cutáneas , Proteínas de Unión a Hormona Tiroide , Hormonas Tiroideas , Humanos , Masculino , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/genética , Proteínas Portadoras/metabolismo , Proteínas Portadoras/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Transducción de Señal , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/genética , Hormonas Tiroideas/metabolismo , Hormonas Tiroideas/genética , Efrina-B2/genética , Efrina-B2/metabolismo
13.
Signal Transduct Target Ther ; 9(1): 216, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39143065

RESUMEN

Third-generation EGFR tyrosine kinase inhibitors (TKIs), exemplified by osimertinib, have demonstrated promising clinical efficacy in the treatment of non-small cell lung cancer (NSCLC). Our previous work has identified ASK120067 as a novel third-generation EGFR TKI with remarkable antitumor effects that has undergone New Drug Application (NDA) submission in China. Despite substantial progress, acquired resistance to EGFR-TKIs remains a significant challenge, impeding the long-term effectiveness of therapeutic approaches. In this study, we conducted a comprehensive investigation utilizing high-throughput proteomics analysis on established TKI-resistant tumor models, and found a notable upregulation of branched-chain amino acid transaminase 1 (BCAT1) expression in both osimertinib- and ASK120067-resistant tumors compared with the parental TKI-sensitive NSCLC tumors. Genetic depletion or pharmacological inhibition of BCAT1 impaired the growth of resistant cells and partially re-sensitized tumor cells to EGFR TKIs. Mechanistically, upregulated BCAT1 in resistant cells reprogrammed branched-chain amino acid (BCAA) metabolism and promoted alpha ketoglutarate (α-KG)-dependent demethylation of lysine 27 on histone H3 (H3K27) and subsequent transcriptional derepression of glycolysis-related genes, thereby enhancing glycolysis and promoting tumor progression. Moreover, we identified WQQ-345 as a novel BCAT1 inhibitor exhibiting antitumor activity both in vitro and in vivo against TKI-resistant lung cancer with high BCAT1 expression. In summary, our study highlighted the crucial role of BCAT1 in mediating resistance to third-generation EGFR-TKIs through epigenetic activation of glycolysis in NSCLC, thereby supporting BCAT1 as a promising therapeutic target for the treatment of TKI-resistant NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Resistencia a Antineoplásicos , Epigénesis Genética , Receptores ErbB , Glucólisis , Neoplasias Pulmonares , Inhibidores de Proteínas Quinasas , Transaminasas , Humanos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Transaminasas/genética , Transaminasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Glucólisis/efectos de los fármacos , Glucólisis/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Ratones , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Acrilamidas/farmacología , Animales , Compuestos de Anilina/farmacología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Indoles , Pirimidinas
14.
Int J Mol Sci ; 25(16)2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39201646

RESUMEN

Accumulating evidence has indicated that stemness-related genes are associated with the aggressiveness of triple-negative breast cancer (TNBC). Because no universal markers for breast CSCs are available, we applied the density gradient centrifugation method to enrich breast CSCs. We demonstrated that the density centrifugation method allows for the isolation of cancer stem cells (CSCs) from adherent and non-adherent MCF7 (Luminal A), MDA-MB-231 (TNBC) and MDA-MB-468 (TNBC) breast cancer cells. The current study shows that the CSCs' enriched fraction from Luminal A and TNBC cells have an increased capacity to grow anchorage-independently. CSCs from adherent TNBC are mainly characterized by metabolic plasticity, whereas CSCs from Luminal A have an increased mitochondrial capacity. Moreover, we found that non-adherent growth CSCs isolated from large mammospheres have a higher ability to grow anchorage-independently compared to CSCs isolated from small mammospheres. In CSCs, a metabolic shift towards glycolysis was observed due to the hypoxic environment of the large mammosphere. Using a bioinformatic analysis, we indicate that hypoxia HYOU1 gene overexpression is associated with the aggressiveness, metastasis and poor prognosis of TNBC. An in vitro study demonstrated that HYOU1 overexpression increases breast cancer cells' stemness and hyperactivates their metabolic activity. In conclusion, we show that density gradient centrifugation is a non-marker-based approach to isolate metabolically flexible (normoxia) CSCs and glycolytic (hypoxic) CSCs from aggressive TNBC.


Asunto(s)
Centrifugación por Gradiente de Densidad , Células Madre Neoplásicas , Neoplasias de la Mama Triple Negativas , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Humanos , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Centrifugación por Gradiente de Densidad/métodos , Femenino , Línea Celular Tumoral , Separación Celular/métodos , Hipoxia de la Célula , Células MCF-7 , Glucólisis/genética
15.
Aging (Albany NY) ; 16(16): 11955-11969, 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39167430

RESUMEN

BACKGROUND: Clear cell renal carcinoma is a common urological malignancy with poor prognosis and treatment outcomes. lncRNAs are important in metabolic reprogramming and the tumor immune microenvironment, but their role in clear cell renal carcinoma is unclear. METHODS: Renal clear cell carcinoma sample data from The Cancer Genome Atlas was used to establish a new risk profile by glycolysis-associated lncRNAs via machine learning. Risk profile-associated column-line plots were constructed to provide a quantitative tool for clinical practice. Patients with renal clear cell carcinoma were divided into high- and low-risk groups. Clinical features, tumor immune microenvironments, and immunotherapy responses were systematically analyzed. We experimentally confirmed the role of LINC01138 and LINC01605 in renal clear cell carcinoma. RESULTS: The risk profile, consisting of LUCAT1, LINC01138, LINC01605, and HOTAIR, reliably predicted survival in patients with renal clear cell carcinoma and was validated in multiple external datasets. The high-risk group presented higher levels of immune cell infiltration and better immunotherapy responses than the low-risk group. LINC01138 and LINC01605 knockdown inhibited the proliferation of renal clear cell carcinoma. CONCLUSIONS: The identified risk profiles can accurately assess the prognosis of patients with clear cell renal carcinoma and identify patient populations that would benefit from immunotherapy, providing valuable insights and therapeutic targets for the clinical management of clear cell renal carcinoma.


Asunto(s)
Carcinoma de Células Renales , Glucólisis , Inmunoterapia , Neoplasias Renales , ARN Largo no Codificante , Microambiente Tumoral , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/terapia , Carcinoma de Células Renales/patología , Humanos , Neoplasias Renales/inmunología , Neoplasias Renales/genética , Neoplasias Renales/patología , Neoplasias Renales/terapia , Glucólisis/genética , Inmunoterapia/métodos , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Pronóstico , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , Proliferación Celular/genética , Multiómica
16.
J Cell Mol Med ; 28(14): e18465, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39022816

RESUMEN

Lung cancer (LC) is one of the malignancies with the highest incidence and mortality in the world, approximately 85% of which is non-small cell lung cancer (NSCLC). Circular RNAs (circRNAs) exert multiple roles in NSCLC occurrence and development. The sequencing results in previous literature have illustrated that multiple circRNAs exhibit upregulation in NSCLC. We attempted to figure out which circRNA exerts an oncogenic role in NSLCL progression. RT-qPCR evaluated circDHTKD1 level in NSCLC tissue specimens and cells. Reverse transcription as well as RNase R digestion assay evaluated circDHTKD1 circular characterization in NSCLC cells. FISH determined circDHTKD1 subcellular distribution in NSCLC cells. Loss- and gain-of-function assays clarified circDHTKD1 role in NSCLC cell growth, tumour growth and glycolysis. Bioinformatics and RIP and RNA pull-down assessed association of circDHTKD1 with upstream molecule Eukaryotic initiation factor 4A-III (EIF4A3) or downstream molecule phosphofructokinase-1 liver type (PFKL) and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) in NSCLC cells. Rescue assays assessed regulatory function of PFKL in circDHTKD1-meidated NSCLC cellular phenotypes. CircDHTKD1 exhibited upregulation and stable circular nature in NSCLC cells. EIF4A3 upregulated circDHTKD1 in NSCLC cells. CircDHTKD1 exerted a promoting influence on NSCLC cell malignant phenotypes and tumour growth. CircDHTKD1 exerted a promoting influence on NSCLC glucose metabolism. CircDHTKD1 exerts a promoting influence on NSCLC glucose metabolism through PFKL upregulation. RIP and RNA pull-down showed that circDHTKD1 could bind to IGF2BP, PFKL could bind to IGF2BP2, and circDHTKD1 promoted the binding of PFKL to IGF2BP2. In addition, RT-qPCR showed that IGF2BP2 knockdown promoted PFKL mRNA degradation, suggesting that IGF2BP2 stabilized PFKL in NSCLC cells. CircDHTKD1 exhibits upregulation in NSCLC. We innovatively validate that EIF4A3-triggered circDHTKD1 upregulation facilitates NSCLC glycolysis through recruiting m6A reader IGF2BP2 to stabilize PFKL, which may provide a new direction for seeking targeted therapy plans of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Factor 4A Eucariótico de Iniciación , Regulación Neoplásica de la Expresión Génica , Glucólisis , Neoplasias Pulmonares , ARN Circular , Proteínas de Unión al ARN , Humanos , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , ARN Circular/genética , ARN Circular/metabolismo , Glucólisis/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Factor 4A Eucariótico de Iniciación/metabolismo , Factor 4A Eucariótico de Iniciación/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Ratones , Ratones Desnudos , Masculino , Femenino , ARN Helicasas DEAD-box
17.
Genes (Basel) ; 15(7)2024 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-39062731

RESUMEN

The development and growth of cancer is fundamentally dependent on pro-tumor changes in metabolism. Cancer cells generally shift away from oxidative phosphorylation as the primary source of energy and rely more heavily on glycolysis. Receptor tyrosine kinases (RTKs) are a type of receptor that is implicated in this shift to pro-tumor metabolism. RTKs are important drivers of cancer growth and metastasis. One such family of RTKs is the MET family, which consists of MET and RON (MST1R). The overexpression of either MET or RON has been associated with worse cancer patient prognosis in a variety of tumor types. Both MET and RON signaling promote increased glycolysis by upregulating the expression of key glycolytic enzymes via increased MYC transcription factor activity. Additionally, both MET and RON signaling promote increased cholesterol biosynthesis downstream of glycolysis by upregulating the expression of SREBP2-induced cholesterol biosynthesis enzymes via CTTNB1. These changes in metabolism, driven by RTK activity, provide potential targets in limiting tumor growth and metastasis via pharmacological inhibition or modifications in diet. This review summarizes pro-tumor changes in metabolism driven by the MET family of RTKs. In doing so, we will offer our unique perspective on metabolic pathways that drive worse patient prognosis and provide suggestions for future study.


Asunto(s)
Glucólisis , Neoplasias , Proteínas Proto-Oncogénicas c-met , Proteínas Tirosina Quinasas Receptoras , Humanos , Proteínas Proto-Oncogénicas c-met/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Neoplasias/metabolismo , Neoplasias/genética , Neoplasias/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Glucólisis/genética , Transducción de Señal , Animales , Colesterol/metabolismo , Colesterol/biosíntesis , Regulación Neoplásica de la Expresión Génica
18.
BMC Cancer ; 24(1): 926, 2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39085784

RESUMEN

BACKGROUND: As the most malignant tumor of the female reproductive system, ovarian cancer (OC) has garnered increasing attention. The Warburg effect, driven by glycolysis, accounts for tumor cell proliferation under aerobic conditions. However, the metabolic heterogeneity linked to glycolysis in OC remains elusive. METHODS: We integrated single-cell data with OC to score glycolysis level in tumor cell subclusters. This led to the identification of a subcluster predominantly characterized by glycolysis, with a strong correlation to patient prognosis. Core transcription factors were pinpointed using hdWGCNA and metaVIPER. A specific transcription factor regulatory network was then constructed. A glycolysis-related prognostic model was developed and tested for estimating OC prognosis with a total of 85 machine-learning combinations, focusing on specific upregulated genes of two subtypes. We identified IGF2 as a key within the prognostic model and investigated its impact on OC progression and drug resistance through in vitro experiments, including the transwell assay, lactate production detection, and the CCK-8 assay. RESULTS: Analysis showed that the Malignant 7 subcluster was primarily related to glycolysis. Two OC molecular subtypes, CS1 and CS2, were identified with distinct clinical, biological, and microenvironmental traits. A prognostic model was built, and IGF2 emerged as a key gene linked to prognosis. Experiments have proven that IGF2 can promote the glycolysis pathway and the malignant biological progression of OC cells. CONCLUSIONS: We developed two novel OC subtypes based on glycolysis score, established a stable prognostic model, and identified IGF2 as the marker gene. These insights provided a new avenue for exploring OC's molecular mechanisms and personalized treatment approaches.


Asunto(s)
Glucólisis , Factor II del Crecimiento Similar a la Insulina , Neoplasias Ováricas , Femenino , Humanos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis/genética , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Ováricas/tratamiento farmacológico , Pronóstico , Análisis de Expresión Génica de una Sola Célula , Transcriptoma
19.
Cell Biol Toxicol ; 40(1): 58, 2024 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-39060874

RESUMEN

OBJECTIVE: Multiple myeloma (MM) is a deadly plasma cell malignancy with elusive pathogenesis. N6-methyladenosine (m6A) is critically engaged in hematological malignancies. The function of KIAA1429, the largest component of methyltransferases, is unknown. This study delved into the mechanism of KIAA1429 in MM, hoping to offer novel targets for MM therapy. METHODS: Bone marrow samples were attained from 55 MM patients and 15 controls. KIAA1429, YTHDF1, and FOXM1 mRNA levels were detected and their correlation was analyzed. Cell viability, proliferation, cell cycle, and apoptosis were testified. Glycolysis-enhancing genes (HK2, ENO1, and LDHA), lactate production, and glucose uptake were evaluated. The interaction between FOXM1 mRNA and YTHDF1, m6A-modified FOXM1 level, and FOXM1 stability were assayed. A transplantation tumor model was built to confirm the mechanism of KIAA1429. RESULTS: KIAA1429 was at high levels in MM patients and MM cells and linked to poor prognoses. KIAA1429 knockdown restrained MM cell viability, and proliferation, arrested G0/G1 phase, and increased apoptosis. KIAA1429 mRNA in plasma cells from MM patients was positively linked with to glycolysis-enhancing genes. The levels of glycolysis-enhancing genes, glucose uptake, and lactate production were repressed after KIAA1429 knockdown, along with reduced FOXM1 levels and stability. YTHDF1 recognized KIAA1429-methylated FOXM1 mRNA and raised FOXM1 stability. Knockdown of YTHDF1 curbed aerobic glycolysis and malignant behaviors in MM cells, which was nullified by FOXM1 overexpression. KIAA1429 knockdown also inhibited tumor growth in animal experiments. CONCLUSION: KIAA1429 knockdown reduces FOXM1 expression through YTHDF1-mediated m6A modification, thus inhibiting MM aerobic glycolysis and tumorigenesis.


Asunto(s)
Carcinogénesis , Proliferación Celular , Proteína Forkhead Box M1 , Glucólisis , Mieloma Múltiple , Proteínas de Unión al ARN , Humanos , Glucólisis/genética , Proteína Forkhead Box M1/metabolismo , Proteína Forkhead Box M1/genética , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Línea Celular Tumoral , Animales , Proliferación Celular/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , Masculino , Femenino , Ratones , Adenosina/análogos & derivados , Adenosina/metabolismo , Apoptosis/genética , Regulación Neoplásica de la Expresión Génica , Persona de Mediana Edad , Ratones Desnudos , Ratones Endogámicos BALB C
20.
Aging (Albany NY) ; 16(13): 11103-11116, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38990159

RESUMEN

Homeobox C4 (HOXC4) is a member of homeobox family and acts as a transcription factor in regulating morphological development. The current study aimed to determine its role in pancreatic cancer (PC). Bioinformatics analysis was employed to assess the expression and clinical significance of HOXC4 in PC, while the expression of HOXC4 was further confirmed in PC tissues through quantitative real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry (IHC). The impact of HOXC4 on PC cell proliferation was evaluated using various assays including Cell Counting Kit-8, colony formation, apoptosis detection, cell cycle analysis, and subcutaneous tumorigenesis. Extracellular acidification rate, glucose uptake, and lactate production measurements were detected to examine the impact of HOXC4 on glycolysis. The relationship between HOXC4 and lactate dehydrogenase A (LDHA) was investigated using CHIP assay, luciferase reporter assay, and western blot. Notably, there was a substantial increase in HOXC4 expression in PC, and patients with elevated HOXC4 levels exhibited shorter survival durations. HOXC4 knockdown resulted in significantly reduced proliferation and colony formation in PC cells, accompanied by increased apoptosis and G1 phase arrest. The overexpression of HOXC4 resulted in contrasting effects. In vivo, the proliferation of PC cells was diminished upon the knockdown of HOXC4. HOXC4 exhibited an increase in LDHA expression by binding to its promoter. The suppressive effects of HOXC4 knockdown on PC cells were counteracted upon the restoration of LDHA. In conclusion, HOXC4 promoted the proliferation of PC cells by increasing LDHA-mediated glycolysis. HOXC4 can act as a target for PC therapy.


Asunto(s)
Proliferación Celular , Glucólisis , Proteínas de Homeodominio , Neoplasias Pancreáticas , Animales , Femenino , Humanos , Masculino , Ratones , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Glucólisis/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , L-Lactato Deshidrogenasa/metabolismo , L-Lactato Deshidrogenasa/genética , Ratones Desnudos , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA