Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 10.472
Filtrar
1.
Immun Inflamm Dis ; 12(10): e70045, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39422344

RESUMEN

BACKGROUND: Osteoclasts are integral to the advancement of osteoporosis (OP), and their generation under conditions of oxidative stress (OS) involves various pathways. However, the specific mechanism through which the natural antioxidant kaempferol (KAE) mitigates the influence of OS on osteoclasts remains somewhat uncertain. This study aims to evaluate the effect of KAE on osteoclast formation under OS and explore its possible mechanism. METHODS: Zebrafish were used to observe the effects of KAE on OP and OS. OP and OS "double disease targets" network pharmacology were used to predict the action target and mechanism of KAE on OP under OS. The effects of KAE on osteoclast differentiation induced by OS were evaluated using RWA264.7 cells induced by LPS. To elucidate the potential mechanism, we detected the expression of related factors and target genes during induction. RESULTS: The presence of KAE exhibited potential in improving the conditions of OP and OS in zebrafish. KAE can reduce the OS of RAW 264.7 cells stimulated by LPS, inhibit the formation of osteoclasts, and change the level of related factors of OS, and reduce the increase of TRAP. The utilization of network pharmacology and target gene expression assay revealed that KAE exerted a down-regulatory effect on the expression of proto-oncogene tyrosine protein kinase (SRC), nuclear factor kappa-B (NF-κB), Serine/Threonine Kinase-1 (AKT1), Nitric Oxide Synthase 3 (NOS3) and Matrix Metallopeptidase-2 (MMP2). CONCLUSION: Based on the results of this study, KAE may effectively mitigate OS and impede the formation of osteoclasts through the SRC/NF-κB-AKT/NOS3 axis.


Asunto(s)
Quempferoles , FN-kappa B , Osteoclastos , Osteogénesis , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Pez Cebra , Animales , Quempferoles/farmacología , FN-kappa B/metabolismo , Ratones , Estrés Oxidativo/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Osteoporosis/metabolismo , Osteoporosis/tratamiento farmacológico , Diferenciación Celular/efectos de los fármacos , Familia-src Quinasas/metabolismo , Antioxidantes/farmacología
2.
Sci Rep ; 14(1): 23942, 2024 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-39397108

RESUMEN

Nontransformed cells form heterotypic cadherin junctions with adjacent transformed cells to inhibit tumor cell growth and motility. Transformed cells must override this form of growth control, called "contact normalization", to invade and metastasize during cancer progression. Heterocellular cadherin junctions between transformed and nontransformed cells are needed for this process. However, specific mechanisms downstream of cadherin signaling have not been clearly elucidated. Here, we utilized a ß-catenin reporter construct to determine if contact normalization affects Wnt signaling in transformed cells. ß-catenin driven GFP expression in Src transformed mouse embryonic cells was decreased when cultured with cadherin competent nontransformed cells compared to transformed cells cultured with themselves, but not when cultured with cadherin deficient nontransformed cells. We also utilized a layered culture system to investigate the effects of oncogenic transformation and contact normalization on gene expression and oncogenic Src kinase mediated phosphorylation events. RNA-Seq analysis found that cadherin dependent contact normalization inhibited the expression of 22 transcripts that were induced by Src transformation, and increased the expression of 78 transcripts that were suppressed by Src transformation. Phosphoproteomic analysis of cells expressing a temperature sensitive Src kinase construct found that contact normalization decreased phosphorylation of 10 proteins on tyrosine residues that were phosphorylated within 1 h of Src kinase activation in transformed cells. Taken together, these results indicate that cadherin dependent contact normalization inhibits Wnt signaling to regulate oncogenic kinase activity and gene expression, particularly PDPN expression, in transformed cells in order to control tumor progression.


Asunto(s)
Cadherinas , Transformación Celular Neoplásica , beta Catenina , Familia-src Quinasas , Cadherinas/metabolismo , Cadherinas/genética , Animales , Fosforilación , Ratones , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , beta Catenina/metabolismo , beta Catenina/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/genética , Vía de Señalización Wnt
3.
Front Immunol ; 15: 1375781, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39391314

RESUMEN

Introduction: Ulcerative colitis, a subtype of chronic inflammatory bowel disease (IBD), is characterized by relapsing colonic inflammation and ulcers. The traditional Chinese herbal formulation Huang Lian Jie Du (HLJD) decoction is used clinically to treat diarrhea and colitis. However, the mechanisms associated with the effects of treatment remain unclear. This study aims to elucidate the molecular mechanistic effects of HLJD formulation on colitis. Methods: Chronic colitis in mice was induced by adding 1% dextran sulfate sodium (DSS) to their drinking water continuously for 8 weeks, and HLJD decoction at the doses of 2 and 4 g/kg was administered orally to mice daily from the second week until experimental endpoint. Stool consistency scores, blood stool scores, and body weights were recorded weekly. Disease activity index (DAI) was determined before necropsy, where colon tissues were collected for biochemical analyses. In addition, the fecal microbiome of treated mice was characterized using 16S rRNA amplicon sequencing. Results: HLJD decoction at doses of 2 and 4 g/kg relieved DSS-induced chronic colitis in mice by suppressing inflammation through compromised macrophage activity in colonic tissues associated with the colony-stimulating factor 1 receptor (Csf1r)/Src pathway. Furthermore, the HLJD formula could modify the gut microbiota profile by decreasing the abundance of Bacteroides, Odoribacter, Clostridium_sensu_stricto_1, and Parasutterella. In addition, close correlations between DAI, colon length, spleen weight, and gut microbiota were identified. Discussion: Our findings revealed that the HLJD formula attenuated DSS-induced chronic colitis by reducing inflammation via Csf1r/Src-mediated macrophage infiltration, as well as modulating the gut microbiota profile.


Asunto(s)
Colitis , Sulfato de Dextran , Medicamentos Herbarios Chinos , Microbioma Gastrointestinal , Macrófagos , Transducción de Señal , Familia-src Quinasas , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Transducción de Señal/efectos de los fármacos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/microbiología , Familia-src Quinasas/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Masculino , Colon/patología , Colon/efectos de los fármacos , Colon/microbiología
4.
Egypt J Immunol ; 31(4): 36-45, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39417739

RESUMEN

This study examined the genotype, allelic frequencies (polymorphisms) in a sample of Egyptian patients and examined the relationship between disease activity in systemic lupus erythematosus (SLE) and the B lymphoid tyrosine kinase (BLK) and B-cell scaffold protein with ankyrin repeats 1 (BANK1) gene. This case control study involved 70 SLE patients and 40 subjects matched for age and sex as a control group. Clinical data were gathered from each participant, including SLE-related clinical activity indicators. Utilizing the restriction fragment length polymorphism (RFLP)-polymerase chain reaction (PCR), the single nucleotide polymorphisms (SNPs) BLK rs13277113G/A and BANK1 rs10516487G/A were assessed. The most prevalent genotype among the study participants was BLK rs13277113; G/G (57.1%), and BANK rs10516487; GG/ genotype (74.3%). There were no substantial variations in the incidence of genotype and allelic polymorphism between patients and controls (p>0.05). In the studied SLE patients, however, there was no significant association between both alleles (BANK rs10516487 gene alleles, G/G and G/A and BLK rs13277113, G/A, G/G and A/A) and the SLE disease activity score. While there was a significant association between BLK rs13277113 genotype alleles and age. In conclusion, BLK rs13277113 G/A and BANK1 rs10516487 G/A alleles showed no difference between SLE Egyptian patients compared to controls with no link between both genes and SLE disease activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Lupus Eritematoso Sistémico , Proteínas de la Membrana , Polimorfismo de Nucleótido Simple , Humanos , Lupus Eritematoso Sistémico/genética , Femenino , Masculino , Polimorfismo de Nucleótido Simple/genética , Egipto/epidemiología , Proteínas Adaptadoras Transductoras de Señales/genética , Adulto , Predisposición Genética a la Enfermedad/genética , Estudios de Casos y Controles , Proteínas de la Membrana/genética , Alelos , Persona de Mediana Edad , Familia-src Quinasas
5.
J Headache Pain ; 25(1): 175, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39390364

RESUMEN

BACKGROUND: Src family kinases (SFKs) contribute to migraine pathogenesis, yet its role in regulating photophobia behaviour, one of the most common forms of migraine, remains unknown. Here, we addressed whether SFKs antagonism alleviates photophobia behavior and explored the underlying mechanism involving hypothalamus and trigeminal ganglion activity, as measured by the alteration of neuropeptide levels and transcriptome respectively. METHODS: A rapid-onset and injury-free mouse model of photophobia was developed following intranasal injection of the TRPA1 activator, umbellulone. The role of SFKs antagonism on light aversion was assessed by the total time the mouse stays in the light and transition times between the dark and light compartments. To gain insight to the preventive mechanism of SFKs antagonism, hypothalamic neuropeptides levels were assessed using enzyme linked immunofluorescent assay and trigeminal ganglion activity were assessed using RNA-sequencing and qPCR analysis. RESULTS: SFKs antagonism by a clinically relevant SFKs inhibitor saracatinib reduced the total time in light and transition times in male mice, but not in females, suggesting SFKs play a crucial role in photophobia progressing and exhibit a male-only effect. SFKs antagonism had no effect on hypothalamic calcitonin gene-related peptide and pituitary adenylate cyclase-activating polypeptide levels of all mice investigated, suggesting the gender-different effect of saracatinib on light aversion appears to be independent of these hypothalamic neuropeptide levels. In trigeminal ganglion of male mice, photophobia is associated with profound alteration of differentially expressed genes, part of which were reversed by SFKs antagonism. Subsequent qPCR analysis showed SFKs antagonism displayed gender-different modulation of expression in some candidate genes, particularly noteworthy those encoding ion channels (trpm3, Scn8a), ATPase signaling (crebbp, Atp5α1) and kinase receptors (Zmynd8, Akt1). CONCLUSIONS: In conclusion, our data revealed that SFKs antagonism reduced photophobia processing in male mice and exhibited gender-different modulation of trigeminal ganglion activity, primarily manifesting as alterations in the transcriptome profile. These findings underscore the potential of SFKs antagonism for allieving photophobia in males, highlighting its value in the emerging field of precision medicine.


Asunto(s)
Fotofobia , Ganglio del Trigémino , Familia-src Quinasas , Animales , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/metabolismo , Masculino , Fotofobia/etiología , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Femenino , Ratones , Modelos Animales de Enfermedad , Factores Sexuales , Ratones Endogámicos C57BL , Hipotálamo/efectos de los fármacos , Hipotálamo/metabolismo , Quinazolinas , Benzodioxoles
6.
Int J Mol Sci ; 25(19)2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39408805

RESUMEN

c-Src is involved in multiple signaling pathways and serves as a critical target in various cancers. Growing evidence suggests that prolonging a drug's residence time (RT) can enhance its efficacy and selectivity. Thus, the development of c-Src antagonists with longer residence time could potentially improve therapeutic outcomes. In this study, we employed molecular dynamics simulations to explore the binding modes and dissociation processes of c-Src with antagonists characterized by either long or short RTs. Our results reveal that the long RT compound DAS-DFGO-I (DFGO) occupies an allosteric site, forming hydrogen bonds with residues E310 and D404 and engaging in hydrophobic interactions with residues such as L322 and V377. These interactions significantly contribute to the long RT of DFGO. However, the hydrogen bonds between the amide group of DFGO and residues E310 and D404 are unstable. Substituting the amide group with a sulfonamide yielded a new compound, DFOGS, which exhibited more stable hydrogen bonds with E310 and D404, thereby increasing its binding stability with c-Src. These results provide theoretical guidance for the rational design of long residence time c-Src inhibitors to improve selectivity and efficacy.


Asunto(s)
Enlace de Hidrógeno , Simulación de Dinámica Molecular , Inhibidores de Proteínas Quinasas , Humanos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteína Tirosina Quinasa CSK/química , Proteína Tirosina Quinasa CSK/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/química , Familia-src Quinasas/metabolismo , Unión Proteica , Interacciones Hidrofóbicas e Hidrofílicas , Sitio Alostérico , Sitios de Unión
7.
Mol Biol Rep ; 51(1): 950, 2024 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-39222158

RESUMEN

BACKGROUND: Hepatic fibrosis, a prevalent chronic liver condition, involves excessive extracellular matrix production associated with aberrant wound healing. Hepatic stellate cells (HSCs) play a pivotal role in liver fibrosis, activated by inflammatory factors such as sphingosine 1-phosphate (S1P). Despite S1P's involvement in fibrosis, its specific role and downstream pathway in HSCs remain controversial. METHODS: In this study, we investigated the regulatory role of S1P/S1P receptor (S1PR) in Hippo-YAP activation in both LX-2 cell lines and primary HSCs. Real-time PCR, western blot, pharmacological inhibitors, siRNAs, and Rho activity assays were adopted to address the molecular mechanisms of S1P mediated YAP activation. RESULTS: Serum and exogenous S1P significantly increased the expression of YAP target genes in HSCs. Pharmacologic inhibitors and siRNA-mediated knockdowns of S1P receptors showed S1P receptor 2 (S1PR2) as the primary mediator for S1P-induced CTGF expression in HSCs. Results using siRNA-mediated knockdown, Verteporfin, and Phospho-Tag immunoblots showed that S1P-S1PR2 signaling effectively suppressed the Hippo kinases cascade, thereby activating YAP. Furthermore, S1P increased RhoA activities in cells and ROCK inhibitors effectively blocked CTGF induction. Cytoskeletal-perturbing reagents were shown to greatly modulate CTGF induction, suggesting the important role of actin cytoskeleton in S1P-induced YAP activation. Exogeneous S1P treatment was enough to increase the expression of COL1A1 and α-SMA, that were blocked by YAP specific inhibitor. CONCLUSIONS: Our data demonstrate that S1P/S1PR2-Src-RhoA-ROCK axis leads to Hippo-YAP activation, resulting in the up-regulation of CTGF, COL1A1 and α-SMA expression in HSCs. Therefore, S1PR2 may represent a potential therapeutic target for hepatic fibrosis.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo , Células Estrelladas Hepáticas , Lisofosfolípidos , Transducción de Señal , Esfingosina , Factores de Transcripción , Proteínas Señalizadoras YAP , Quinasas Asociadas a rho , Proteína de Unión al GTP rhoA , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/efectos de los fármacos , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Factor de Crecimiento del Tejido Conjuntivo/genética , Lisofosfolípidos/metabolismo , Lisofosfolípidos/farmacología , Humanos , Quinasas Asociadas a rho/metabolismo , Quinasas Asociadas a rho/genética , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Proteínas Señalizadoras YAP/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Receptores de Esfingosina-1-Fosfato/metabolismo , Receptores de Esfingosina-1-Fosfato/genética , Línea Celular , Cirrosis Hepática/metabolismo , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Familia-src Quinasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Receptores de Lisoesfingolípidos/metabolismo , Receptores de Lisoesfingolípidos/genética , Colágeno Tipo I/metabolismo , Colágeno Tipo I/genética , Vía de Señalización Hippo
8.
Nat Commun ; 15(1): 7885, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251588

RESUMEN

The IL6-GP130-STAT3 pathway facilitates lung cancer progression and resistance to tyrosine kinase inhibitors. Although glycosylation alters the stability of GP130, its effect on the ligand IL6 remains unclear. We herein find that N-glycosylated IL6, especially at Asn73, primarily stimulates JAK-STAT3 signaling and prolongs STAT3 phosphorylation, whereas N-glycosylation-defective IL6 (deNG-IL6) induces shortened STAT3 activation and alters the downstream signaling preference for the SRC-YAP-SOX2 axis. This signaling shift induces epithelial-mesenchymal transition (EMT) and migration in vitro and metastasis in vivo, which are suppressed by targeted inhibitors and shRNAs against SRC, YAP, and SOX2. Osimertinib-resistant lung cancer cells secrete a large amount of deNG-IL6 through reduced N-glycosyltransferase gene expression, leading to clear SRC-YAP activation. deNG-IL6 contributes to drug resistance, as confirmed by in silico analysis of cellular and clinical transcriptomes and signal expression in patient specimens. Therefore, the N-glycosylation status of IL6 not only affects cell behaviors but also shows promise in monitoring the dynamics of lung cancer evolution.


Asunto(s)
Acrilamidas , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Interleucina-6 , Neoplasias Pulmonares , Inhibidores de Proteínas Quinasas , Factor de Transcripción STAT3 , Humanos , Glicosilación , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Interleucina-6/metabolismo , Interleucina-6/genética , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Inhibidores de Proteínas Quinasas/farmacología , Animales , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/genética , Línea Celular Tumoral , Acrilamidas/farmacología , Ratones , Transducción de Señal/efectos de los fármacos , Proteínas Señalizadoras YAP/metabolismo , Proteínas Señalizadoras YAP/genética , Compuestos de Anilina/farmacología , Receptor gp130 de Citocinas/metabolismo , Receptor gp130 de Citocinas/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXB1/genética , Fosforilación , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Ratones Desnudos , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Metástasis de la Neoplasia , Regulación Neoplásica de la Expresión Génica , Femenino , Indoles , Pirimidinas
9.
Cell Death Dis ; 15(9): 678, 2024 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-39284838

RESUMEN

SPHK1 (sphingosine kinase type 1) is characterized as a rate-limiting enzyme in sphingolipid metabolism to phosphorylate sphingosine into sphingosine-1-phosphate (S1P) that can bind to S1P receptors (S1PRs) to initiate several signal transductions leading to cell proliferation and survival of normal cell. Many studies have indicated that SPHK1 is involved in several types of cancer development, however, a little is known in bladder cancer. The TCGA database analysis was utilized for analyzing the clinical relevance of SPHK1 in bladder cancer. Through CRISPR/Cas9 knockout (KO) and constitutive activation (CA) strategies on SPHK1 in the bladder cancer cells, we demonstrated the potential downstream target could be programmed cell death 1 ligand 2 (PD-L2). On the other hand, we demonstrated that FDA-approved SPHK1 inhibitor Gilenya® (FTY720) can successfully suppress bladder cancer metastasis by in vitro and in vivo approaches. This finding indicated that SPHK1 as a potent therapeutic target for metastatic bladder cancer by dissecting the mechanism of action, SPHK1/S1P-elicited Akt/ß-catenin activation promoted the induction of PD-L2 that is a downstream effector in facilitating bladder cancer invasion and migration. Notably, PD-L2 interacted with c-Src that further activates FAK. Here, we unveil the clinical relevance of SPHK1 in bladder cancer progression and the driver role in bladder cancer metastasis. Moreover, we demonstrated the inhibitory effect of FDA-approved SPHK1 inhibitor FTY720 on bladder cancer metastasis from both in vitro and in vivo models.


Asunto(s)
Fosfotransferasas (Aceptor de Grupo Alcohol) , Transducción de Señal , Neoplasias de la Vejiga Urinaria , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/genética , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Animales , Línea Celular Tumoral , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/genética , Metástasis de la Neoplasia , Ratones , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Familia-src Quinasas/metabolismo , Movimiento Celular , Ratones Desnudos , Lisofosfolípidos/metabolismo , Proteína Tirosina Quinasa CSK/metabolismo , Clorhidrato de Fingolimod/farmacología , Proliferación Celular
10.
Cell Commun Signal ; 22(1): 454, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39327614

RESUMEN

BACKGROUND: Treatment options for the Triple-Negative Breast Cancer (TNBC) subtype remain limited and the outcome for patients with advanced TNBC is very poor. The standard of care is chemotherapy, but approximately 50% of tumors develop resistance. METHODS: We performed gene expression profiling of 58 TNBC tumor samples by microarray, comparing chemosensitive with chemoresistant tumors, which revealed that one of the top upregulated genes was TGFß2. A connectivity mapping bioinformatics analysis predicted that the SRC inhibitor Dasatinib was a potential pharmacological inhibitor of chemoresistant TNBCs. Claudin-low TNBC cell lines were selected to represent poor-outcome, chemoresistant TNBC, for in vitro experiments and in vivo models. RESULTS: In vitro, we identified a signaling axis linking SRC, AKT and ERK2, which in turn upregulated the stability of the transcription factors, Slug and Snail. Slug was shown to repress TGFß2-antisense 1 to promote TGFß2 signaling, upregulating cell survival via apoptosis and DNA-damage responses. Additionally, an orthotopic allograft in vivo model demonstrated that the SRC inhibitor Dasatinib reduced tumor growth as a single agent, and enhanced responses to the TNBC mainstay drug, Epirubicin. CONCLUSION: Targeting the SRC-Slug-TGFß2 axis may therefore lead to better treatment options and improve patient outcomes in this highly aggressive subpopulation of TNBCs.


In our study, we focused on a particular subtype of aggressive breast cancer called Triple-Negative Breast Cancer (TNBC). We investigated a complex series of events that contribute to poor outcomes in this disease and uncovered a crucial signaling cascade driving tumor growth and progression.At the core of this signaling cascade are three key proteins: SRC, AKT, and ERK2. Together, they form a pathway that activates a transcription factor called Slug. Transcription factors act like molecular switches, controlling the expression of genes. Once Slug is activated, it strongly suppresses genes that would normally restrict cell growth and cell spread.One of the genes downregulated by Slug is TGFB2-AS1. This product of the TGFB2-AS1 gene normally controls levels of its target protein called TGF-beta2 (TGFB2), a protein which has roles in cell growth, cell migration and differentiation. Slug downregulation of TGFB2-AS1 results in higher TGFB2 levels, and this in turn contributes to the uncontrolled growth and spread of cancer cells. TGFB2, and other proteins in this pathway (SRC, AKT, ERK2, and a Slug interactor called LSD1) all maintain the stability of Slug, meaning that Slug levels remain high and drive the aggressive features of this subtype of breast cancer.Overall, our research sheds light on the intricate molecular mechanisms driving aggressive TNBC. It also identifies potential targets for future therapies, aimed at disrupting this harmful signaling pathway and potentially improving patient outcomes for this disease.


Asunto(s)
Dasatinib , Transducción de Señal , Factores de Transcripción de la Familia Snail , Factor de Crecimiento Transformador beta2 , Neoplasias de la Mama Triple Negativas , Familia-src Quinasas , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Humanos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta2/genética , Familia-src Quinasas/metabolismo , Línea Celular Tumoral , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción de la Familia Snail/genética , Femenino , Animales , Dasatinib/farmacología , Dasatinib/uso terapéutico , Ratones , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/efectos de los fármacos
11.
FASEB J ; 38(17): e70046, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39259502

RESUMEN

Large-conductance, calcium-activated potassium channels (BK channels) and the Na/K-ATPase are expressed universally in vascular smooth muscle. The Na/K-ATPase may act via changes in the intracellular Ca2+ concentration mediated by the Na/Ca exchanger (NCX) and via Src kinase. Both pathways are known to regulate BK channels. Whether BK channels functionally interact in vascular smooth muscle cells with the Na/K-ATPase remains to be elucidated. Thus, this study addressed the hypothesis that BK channels limit ouabain-induced vasocontraction. Rat mesenteric arteries were studied using isometric myography, FURA-2 fluorimetry and proximity ligation assay. The BK channel blocker iberiotoxin potentiated methoxamine-induced contractions. The cardiotonic steroid, ouabain (10-5 M), induced a contractile effect of IBTX at basal tension prior to methoxamine administration and enhanced the pro-contractile effect of IBTX on methoxamine-induced contractions. These facilitating effects of ouabain were prevented by the inhibition of either NCX or Src kinase. Furthermore, inhibition of NCX or Src kinase reduced the BK channel-mediated negative feedback regulation of arterial contraction. The effects of NCX and Src kinase inhibition were independent of each other. Co-localization of the Na/K-ATPase and the BK channel was evident. Our data suggest that BK channels limit ouabain-induced vasocontraction by a dual mechanism involving the NCX and Src kinase signaling. The data propose that the NCX and the Src kinase pathways, mediating the ouabain-induced activation of the BK channel, act in an independent manner.


Asunto(s)
Canales de Potasio de Gran Conductancia Activados por el Calcio , Arterias Mesentéricas , Músculo Liso Vascular , Ouabaína , Intercambiador de Sodio-Calcio , ATPasa Intercambiadora de Sodio-Potasio , Familia-src Quinasas , Animales , Ouabaína/farmacología , Familia-src Quinasas/metabolismo , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/efectos de los fármacos , Ratas , Masculino , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Arterias Mesentéricas/efectos de los fármacos , Arterias Mesentéricas/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Vasoconstricción/efectos de los fármacos , Ratas Wistar , Contracción Muscular/efectos de los fármacos
12.
Sci Rep ; 14(1): 21444, 2024 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-39271790

RESUMEN

Mesenchymal stromal/stem cells (MSC) play a crucial role in promoting neovascularization, which is essential for wound healing. They are commonly utilized as an autologous source of progenitor cells in various stem cell-based therapies. However, incomplete MSC differentiation towards a vascular endothelial cell phenotype questions their involvement in an alternative process to angiogenesis, namely vasculogenic mimicry (VM), and the signal transducing events that regulate their in vitro priming into capillary-like structures. Here, human MSC were primed on top of Cultrex matrix to recapitulate an in vitro phenotype of VM. Total RNA was extracted, and differential gene expression assessed through RNA-Seq analysis and RT-qPCR. Transient gene silencing was achieved using specific siRNA. AG490, Tofacitinib, and PP2 pharmacological effects on VM structures were analyzed using the Wimasis software. In vitro VM occurred within 4 h and was prevented by the JAK/STAT3 inhibitors AG490 and Tofacitinib, as well as by the Src inhibitor PP2. RNA-Seq highlighted STAT3 as a signaling hub contributing to VM when transcripts from capillary-like structures were compared to those from cell monolayers. Concomitant increases in IL6, IL1b, CSF1, CSF2, STAT3, FOXC2, RPSA, FN1, and SNAI1 transcript levels suggest the acquisition of a combined angiogenic, inflammatory and epithelial-to-mesenchymal transition phenotype in VM cultures. Increases in STAT3, FOXC2, RPSA, Fibronectin, and Snail protein expression were confirmed during VM. STAT3 and RPSA gene silencing abrogated in vitro VM. In conclusion, in vitro priming of MSC into VM structures requires Src/JAK/STAT3 signaling. This molecular evidence indicates that a clinically viable MSC-mediated pseudo-vasculature process could temporarily support grafts through VM, allowing time for the host vasculature to infiltrate and remodel the injured tissues.


Asunto(s)
Quinasas Janus , Células Madre Mesenquimatosas , Neovascularización Fisiológica , Factor de Transcripción STAT3 , Transducción de Señal , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Factor de Transcripción STAT3/metabolismo , Quinasas Janus/metabolismo , Familia-src Quinasas/metabolismo , Células Cultivadas , Diferenciación Celular
13.
J Transl Med ; 22(1): 859, 2024 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-39334374

RESUMEN

This study aimed to investigate the effect and potential mechanism of evodiamine (EVO) on proliferation and apoptosis of nasopharyngeal carcinoma (NPC) cells. EVO inhibited proliferation, blocked cell cycle progression, and induced apoptosis of NPC cells. There are 27 known anti-NPC targets of EVO, of which eight are core targets, namely SRC, ERBB2, STAT3, MAPK8, NOS3, CXCL8, APP, and HDAC1. Molecular docking analysis showed that the binding of EVO with its key targets (SRC, ERBB2) was good. EVO also reduced the expression of SRC and ERBB2, the key proteins p-MEK and p-ERK1/2 of the MAPK/ERK signaling pathway, and the downstream proteins PCNA and XIAP. EVO inhibited the growth of NPC xenografts in nude mice and reduced the expression levels of SRC, ERBB2, ERK1/2, p-ERK1/2, PCNA and XIAP in NPC tissue. When the MAPK/ERK signaling pathway was activated by epidermal growth factor (EGF), the expression levels of PCNA and XIAP increased, the cell proliferation index increased, and the apoptosis rate decreased in the EGF + EVO treatment group compared to treatment with EVO alone. These changes indicated that the inhibitory effect of EVO on proliferation and apoptosis of NPC cells was related to the down-regulation of SRC and ERBB2 expression, and further inhibition of the MAPK/ERK signaling pathway.


Asunto(s)
Apoptosis , Proliferación Celular , Sistema de Señalización de MAP Quinasas , Ratones Desnudos , Carcinoma Nasofaríngeo , Quinazolinas , Receptor ErbB-2 , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Carcinoma Nasofaríngeo/patología , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/tratamiento farmacológico , Humanos , Animales , Receptor ErbB-2/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Línea Celular Tumoral , Quinazolinas/farmacología , Quinazolinas/uso terapéutico , Familia-src Quinasas/metabolismo , Neoplasias Nasofaríngeas/patología , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , Simulación del Acoplamiento Molecular , Ratones Endogámicos BALB C , Ratones
14.
ACS Chem Biol ; 19(10): 2220-2231, 2024 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-39289808

RESUMEN

The differentiation of placental cytotrophoblasts (CTBs) into the syncytiotrophoblast (STB) layer results in a significant remodeling of the plasma membrane proteome. Here, we use a peroxidase-catalyzed proximity labeling strategy to map the dynamic plasma membrane proteomes of CTBs and STBs. Coupled with mass-spectrometry-based proteomics, we identify hundreds of plasma membrane proteins and observe relative changes in protein abundance throughout differentiation, including the upregulation of the plasma-membrane-localized nonreceptor tyrosine kinase LYN. We show that both siRNA-mediated knockdown and small molecule inhibition of LYN kinase function impairs CTB fusion and reduces the expression of syncytialization markers, presenting a function for LYN outside of its canonical role in immunological signaling. Our results demonstrate the use of the proximity labeling platform to discover functional regulators within the plasma membrane and provide new avenues to regulate trophoblast differentiation.


Asunto(s)
Diferenciación Celular , Membrana Celular , Trofoblastos , Familia-src Quinasas , Trofoblastos/metabolismo , Trofoblastos/citología , Familia-src Quinasas/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Humanos , Membrana Celular/metabolismo , Femenino , Embarazo , Proteómica/métodos
15.
Genes (Basel) ; 15(9)2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39336808

RESUMEN

BACKGROUND/OBJECTIVES: Glass, bone, and dentin are commonly applied substrates for osteoclast cultures; however, the impact of these substrates on osteoclastogenesis remains underexplored. This study aimed to address a significant gap in understanding how different substrates influence the process of osteoclastogenesis. METHODS: RAW 264.7 cells were cultured and induced with RANKL on glass, bone, and dentin slides. Histological and molecular techniques were used to identify patterns and differences in osteoclast behavior on each substrate. RESULTS: Osteoclasts cultured on glass slides possessed the greatest number of nuclei and the highest expression levels of ACP5 (TRAP) and CTSK, with osteoclasts on bone and dentin slides displaying progressively lower levels. Src expression was also most pronounced in osteoclasts on glass slides, with decreased levels observed on bone and dentin. This variation in Src expression likely contributed to differences in cytoskeletal remodeling and oxidative phosphorylation (OXPHOS), resulting in substrate-dependent divergences in osteoclastogenesis. CONCLUSIONS: Glass slides were the most favorable substrate for inducing osteoclastogenesis, while bone and dentin slides were less effective. The substrate-induced expression of Src played a fundamental role in shaping the phenotypic divergence of osteoclasts. These insights fill important knowledge gaps and have significant implications for the development and selection of in vitro models for bone-related diseases and drug screening platforms.


Asunto(s)
Osteoclastos , Osteogénesis , Animales , Ratones , Osteoclastos/metabolismo , Osteoclastos/citología , Células RAW 264.7 , Osteogénesis/genética , Ligando RANK/metabolismo , Ligando RANK/genética , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Diferenciación Celular , Dentina/metabolismo , Vidrio , Huesos/metabolismo , Huesos/citología
16.
Biomed Pharmacother ; 179: 117325, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39226729

RESUMEN

Direct-acting antivirals ledipasvir (LDV) and daclatasvir (DCV) are widely used as part of combination therapies to treat Hepatitis C infections. Here we show that these compounds inhibit the proliferation, invasion, and colony formation of triple-negative MDA-MB-231 breast cancer cells, SRC-transduced SW620 colon cancer cells and SRC- transduced NIH3T3 fibroblasts. DCV also inhibits the expression of PDL-1, which is responsible for resistance to immunotherapy in breast cancer cells. The demonstrated low toxicity in many Hepatitis C patients suggests LDV and DCV could be used in combination therapies for cancer patients. At the molecular level, these direct-acting antivirals inhibit the phosphorylation of Akt and the ephrin type A receptor 2 (EPHA2) by destabilizing a Src-EPHA2 complex, although they do not affect the general kinase activity of Src. Thus, LDV and DCV could be effective drugs for Src-associated cancers without the inherent toxicity of classical Src inhibitors.


Asunto(s)
Antivirales , Bencimidazoles , Carbamatos , Neoplasias Colorrectales , Regulación hacia Abajo , Fluorenos , Imidazoles , Proteínas Proto-Oncogénicas c-akt , Pirrolidinas , Transducción de Señal , Neoplasias de la Mama Triple Negativas , Valina , Familia-src Quinasas , Humanos , Bencimidazoles/farmacología , Animales , Pirrolidinas/farmacología , Imidazoles/farmacología , Ratones , Proteínas Proto-Oncogénicas c-akt/metabolismo , Familia-src Quinasas/metabolismo , Fluorenos/farmacología , Línea Celular Tumoral , Antivirales/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/genética , Carbamatos/farmacología , Regulación hacia Abajo/efectos de los fármacos , Valina/análogos & derivados , Valina/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/genética , Transducción de Señal/efectos de los fármacos , Células 3T3 NIH , Femenino , Proliferación Celular/efectos de los fármacos , United States Food and Drug Administration , Aprobación de Drogas , Estados Unidos
17.
Eur J Pharmacol ; 983: 176983, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39243926

RESUMEN

Chronic kidney disease (CKD) is a global public health problem, involving about 10% of the global population. Unfortunately, there are currently no effective drugs. Kidney fibrosis is the main pathology of CKD, where integrins play crucial roles in renal fibrogenesis. Recently, Bexotegrast (PLN-74809) as a dual integrin αvß1/αvß6 inhibitor could reduce the degree of lung fibrosis in patients with idiopathic pulmonary fibrosis. However, the role of PLN-74809 remains unclear in fibrotic kidney disease. Here, we have revealed that PLN-74809 administration dose-dependently delayed the progression of renal fibrosis in both adenine diet- and unilateral ureteral obstruction (UUO)-induced mice. Mechanistically, PLN-74809 targeted integrin αvß1/αvß6 to inhibit FAK/Src/Akt/ß-catenin cascade in fibrotic kidneys. In summary, our results for the first time highlighted the αvß1/αvß6 inhibitor PLN-74809 exerted potential therapeutic against kidney fibrosis.


Asunto(s)
Fibrosis , Integrinas , Animales , Masculino , Ratones , Adenina/análogos & derivados , Adenina/farmacología , Antígenos de Neoplasias/metabolismo , beta Catenina/metabolismo , beta Catenina/antagonistas & inhibidores , Modelos Animales de Enfermedad , Quinasa 1 de Adhesión Focal/metabolismo , Quinasa 1 de Adhesión Focal/antagonistas & inhibidores , Integrinas/antagonistas & inhibidores , Integrinas/metabolismo , Riñón/patología , Riñón/efectos de los fármacos , Riñón/metabolismo , Enfermedades Renales/patología , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/metabolismo , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/metabolismo , Transducción de Señal/efectos de los fármacos , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo , Obstrucción Ureteral/patología , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/tratamiento farmacológico
18.
Int J Biol Sci ; 20(12): 4731-4749, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39309445

RESUMEN

Methyltransferase-like (METTL)18 has histidine methyltransferase activity on the RPL3 protein and is involved in ribosome biosynthesis and translation elongations. Several studies have reported that actin polymerization serves as a Src regulator, and HSP90 is involved in forming polymerized actin bundles. To understand the role of METTL18 in breast cancer and to demonstrate the importance of METTL18 in HER-2 negative breast cancer metastasis, we used biochemical, molecular biological, and immunological approaches in vitro (breast tumor cell lines), in vivo (tumor xenograft model), and in samples of human breast tumors. A gene expression comparison of 31 METTL series genes and 22 methyltransferases in breast cancer patients revealed that METTL18 is highly amplified in human HER2-negative breast cancer. In addition, elevated levels of METTL18 expression in patients with HER2-negative breast cancer are associated with poor prognosis. Loss of METTL18 significantly reduced the metastatic responses of breast tumor cells in vitro and in vivo. Mechanistically, METTL18 indirectly regulates the phosphorylation of the proto-oncogene tyrosine-protein kinase Src and its downstream molecules in MDA-MB-231 cells via METTL18-mediated RPL3 methylation, which is also involved in determining HSP90 integrity and protein levels. In confocal microscopy and F/G-actin assays, METTL18 was found to induce actin polymerization via HSP90. Molecular events involving METTL18, RPL3, HSP90, and actin polymerization yielded Src phosphorylated at both tyrosine 419 and tyrosine 530 with kinase activity and oncogenic functions. Therefore, it is suggested that the METTL18-HSP90-Actin-Src regulatory axis plays critical oncogenic roles in the metastatic responses of HER2-negative breast cancer and could be a promising therapeutic target.


Asunto(s)
Neoplasias de la Mama , Metiltransferasas , Proto-Oncogenes Mas , Receptor ErbB-2 , Familia-src Quinasas , Humanos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Femenino , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Línea Celular Tumoral , Animales , Metiltransferasas/metabolismo , Metiltransferasas/genética , Familia-src Quinasas/metabolismo , Ratones , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/genética , Ratones Desnudos , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/genética , Fosforilación
19.
Cancer Genomics Proteomics ; 21(5): 485-501, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39191501

RESUMEN

BACKGROUND/AIM: Glioblastomas (GBM) are infiltrative malignant brain tumors which mostly recur within a year's time following surgical resection and chemo-radiation therapy. Studies on glioblastoma cells following radio-chemotherapy, have been demonstrated to induce trans-differentiation, cellular plasticity, activation of DNA damage response and stemness. As glioblastomas are heterogenous tumors that develop treatment resistance and plasticity, we investigated if there exist genome-wide DNA methylation changes in recurrent tumors. MATERIALS AND METHODS: Utilizing genome-wide DNA methylation arrays, we compared the DNA methylation profile of 11 primary (first occurrence) tumors with 13 recurrent (relapsed) GBM, to delineate the contribution of epigenetic changes associated with therapy exposure, therapy resistance, and relapse of disease. RESULTS: Our data revealed 1,224 hypermethylated- and 526 hypomethylated-probes in recurrent glioblastomas compared to primary disease. We found differential methylation of solute carrier and ion channel genes, interleukin receptor/ligand genes, tumor-suppressor genes and genes associated with metastasis. We functionally characterized one such hypomethylated-up-regulated gene, namely anthrax toxin receptor 1/tumor endothelial marker 8 (ANTXR1/TEM8), whose expression was validated to be significantly up-regulated in recurrent glioblastomas compared to primary tumors and confirmed by immunohistochemistry. Using overexpression and knockdown approaches, we showed that TEM8 induces proliferation, invasion, migration, and chemo-radioresistance in glioblastoma cells. Additionally, we demonstrated a novel mechanism of ß-catenin stabilization and activation of the ß-catenin transcriptional program due to TEM8 overexpression via a Src/PI3K/AKT/GSK3ß/ß-catenin pathway. CONCLUSION: We report genome-wide DNA methylation changes in recurrent GBM and suggest involvement of the TEM8 gene in GBM recurrence and progression.


Asunto(s)
Neoplasias Encefálicas , Metilación de ADN , Glioblastoma , Glucógeno Sintasa Quinasa 3 beta , Recurrencia Local de Neoplasia , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , beta Catenina , Humanos , Glioblastoma/genética , Glioblastoma/patología , Glioblastoma/metabolismo , beta Catenina/metabolismo , beta Catenina/genética , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Glucógeno Sintasa Quinasa 3 beta/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Femenino , Transducción de Señal , Regulación Neoplásica de la Expresión Génica , Masculino , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Persona de Mediana Edad , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Anciano , Línea Celular Tumoral , Adulto , Proteínas de Microfilamentos
20.
Nat Commun ; 15(1): 7455, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39198451

RESUMEN

Increased fatty acid synthesis benefits glioblastoma malignancy. However, the coordinated regulation of cytosolic acetyl-CoA production, the exclusive substrate for fatty acid synthesis, remains unclear. Here, we show that proto-oncogene tyrosine kinase c-SRC is activated in glioblastoma and remodels cytosolic acetyl-CoA production for fatty acid synthesis. Firstly, acetate is an important substrate for fatty acid synthesis in glioblastoma. c-SRC phosphorylates acetyl-CoA synthetase ACSS2 at Tyr530 and Tyr562 to stimulate the conversion of acetate to acetyl-CoA in cytosol. Secondly, c-SRC inhibits citrate-derived acetyl-CoA synthesis by phosphorylating ATP-citrate lyase ACLY at Tyr682. ACLY phosphorylation shunts citrate to IDH1-catalyzed NADPH production to provide reducing equivalent for fatty acid synthesis. The c-SRC-unresponsive double-mutation of ACSS2 and ACLY significantly reduces fatty acid synthesis and hampers glioblastoma progression. In conclusion, this remodeling fulfills the dual needs of glioblastoma cells for both acetyl-CoA and NADPH in fatty acid synthesis and provides evidence for glioma treatment by c-SRC inhibition.


Asunto(s)
Acetilcoenzima A , Ácidos Grasos , Glioblastoma , Proto-Oncogenes Mas , Glioblastoma/metabolismo , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ácidos Grasos/metabolismo , Ácidos Grasos/biosíntesis , Línea Celular Tumoral , Fosforilación , Acetilcoenzima A/metabolismo , Animales , Proteína Tirosina Quinasa CSK/metabolismo , Proteína Tirosina Quinasa CSK/genética , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Progresión de la Enfermedad , Ratones , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , NADP/metabolismo , Ratones Desnudos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...