Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Redox Biol ; 71: 103108, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38457903

RESUMEN

High-risk human papillomaviruses (HPVs) are the causative agents of cervical cancer. Here, we report that HPV16 E6E7 promotes cervical cancer cell proliferation by activating the pentose phosphate pathway (PPP). We found that HPV16 E6 activates the PPP primarily by increasing glucose-6-phosphate dehydrogenase (G6PD) enzyme activity. Mechanistically, HPV16 E6 promoted G6PD dimer formation by inhibiting its lactylation. Importantly, we suggest that G6PD K45 was lactylated during G6PD-mediated antioxidant stress. In primary human keratinocytes and an HPV-negative cervical cancer C33A cells line ectopically expressing HPV16 E6, the transduction of G6PD K45A (unable to be lactylated) increased GSH and NADPH levels and, correspondingly, decreasing ROS levels. Conversely, the re-expression of G6PD K45T (mimicking constitutive lactylation) in HPV16-positive SiHa cells line inhibited cell proliferation. In vivo, the inhibition of G6PD enzyme activity with 6-aminonicotinamide (6-An) or the re-expression of G6PD K45T inhibited tumor proliferation. In conclusion, we have revealed a novel mechanism of HPV oncoprotein-mediated malignant transformation. These findings might provide effective strategies for treating cervical and HPV-associated cancers.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Femenino , Humanos , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Línea Celular Tumoral , Neoplasias del Cuello Uterino/metabolismo , Glucosafosfato Deshidrogenasa/genética , Glucosafosfato Deshidrogenasa/metabolismo , Vía de Pentosa Fosfato , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Proliferación Celular
2.
J Exp Clin Cancer Res ; 43(1): 36, 2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38291438

RESUMEN

BACKGROUND: Discoidin, CUB, and LCCL domain-containing type I (DCBLD1) is identified as an oncogene involved in multiple regulation of tumor progression, but specific mechanisms remain unclear in cervical cancer. Lactate-mediated lactylation modulates protein function. Whether DCBLD1 can be modified by lactylation and the function of DCBLD1 lactylation are unknown. Therefore, this study aims to investigate the lactylation of DCBLD1 and identify its specific lactylation sites. Herein, we elucidated the mechanism by which lactylation modification stabilizes the DCBLD1 protein. Furthermore, we investigated DCBLD1 overexpression activating pentose phosphate pathway (PPP) to promote the progression of cervical cancer. METHODS: DCBLD1 expression was examined in human cervical cancer cells and adjacent non-tumorous tissues using quantitative reverse transcription-polymerase chain reaction, western blotting, and immunohistochemistry. In vitro and in vivo studies were conducted to investigate the impact of DCBLD1 on the progression of cervical cancer. Untargeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomics studies were used to characterize DCBLD1-induced metabolite alterations. Western blot, immunofuorescence and transmission electron microscopy were performed to detect DCBLD1 degradation of G6PD by activating autophagy. Chromatin immunoprecipitation, dual luciferase reporter assay for detecting the mechanism by which lactate increases DCBLD1 transcription. LC-MS/MS was employed to verify specific modification sites within the DCBLD1 protein. RESULTS: We found that lactate increased DCBLD1 expression, activating the PPP to facilitate the proliferation and metastasis of cervical cancer cells. DCBLD1 primarily stimulated PPP by upregulating glucose-6-phosphate dehydrogenase (G6PD) expression and enzyme activity. The mechanism involved the increased enrichment of HIF-1α in the DCBLD1 promoter region, enhancing the DCBLD1 mRNA expression. Additionally, lactate-induced DCBLD1 lactylation stabilized DCBLD1 expression. We identified DCBLD1 as a lactylation substrate, with a predominant lactylation site at K172. DCBLD1 overexpression inhibited G6PD autophagic degradation, activating PPP to promote cervical cancer progression. In vivo, 6-An mediated inhibition of G6PD enzyme activity, inhibiting tumor proliferation. CONCLUSIONS: Our findings revealed a novel post-translational modification type of DCBDL1, emphasizing the significance of lactylation-driven DCBDL1-mediated PPP in promoting the progression of cervical cancer.


Asunto(s)
Neoplasias del Cuello Uterino , Femenino , Humanos , Cromatografía Liquida , Lactatos , Vía de Pentosa Fosfato , Espectrometría de Masas en Tándem , Neoplasias del Cuello Uterino/genética
3.
Mol Carcinog ; 63(2): 339-355, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37988232

RESUMEN

Over 99% of precancerous cervical lesions are associated with human papillomavirus (HPV) infection, with HPV types 16 and 18 (especially type 16) found in over 70% of cervical cancer cases globally. E6, a critical HPV gene, triggers malignant proliferation by degrading p53; however, this mechanism alone cannot fully explain the oncogenic effects of HPV16 E6. Therefore, we aimed to investigate new targets of HPV oncogenic mechanisms. Our results revealed significant changes in nonoxidative pentose phosphate pathway (PPP) metabolites in HPV16-positive cells. However, the role of nonoxidative PPP in HPV-associated cell transformation and tumor development remained unexplored. In this study, we investigated the impact and mechanisms of HPV16 E6 on cervical cancer proliferation using the HPV-negative cervical cancer cell line (C33A). HPV16 E6 was found to promote cervical cancer cell proliferation both in vitro and in vivo, activating the nonoxidative PPP. Transketolase (TKT), a key enzyme in the nonoxidative PPP, is highly expressed in cervical cancer tissues and associated with poor prognosis. HPV16 E6 promotes cervical cancer cell proliferation by upregulating TKT activity through the activation of AKT. In addition, oxythiamine (OT), a TKT inhibitor, hindered tumor growth, with enhanced effects when combined with cisplatin (DDP). In conclusion, HPV16 E6 promotes cervical cancer proliferation by upregulating TKT activity through the activation of AKT. OT demonstrates the potential to inhibit HPV16-positive cervical cancer growth, and when combined with DDP, could further enhance the tumor-suppressive effect of DDP.


Asunto(s)
Proteínas Oncogénicas Virales , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Femenino , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Papillomavirus Humano 16/metabolismo , Transcetolasa/metabolismo , Neoplasias del Cuello Uterino/genética , Infecciones por Papillomavirus/genética , Proteínas Oncogénicas Virales/metabolismo , Proliferación Celular , Línea Celular Tumoral
4.
Front Nutr ; 10: 1157352, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37680899

RESUMEN

Senescence is an inevitable biological process. Disturbances in glucose and lipid metabolism are essential features of cellular senescence. Given the important roles of these types of metabolism, we review the evidence for how key metabolic enzymes influence senescence and how senescence-related secretory phenotypes, autophagy, apoptosis, insulin signaling pathways, and environmental factors modulate glucose and lipid homeostasis. We also discuss the metabolic alterations in abnormal senescence diseases and anti-cancer therapies that target senescence through metabolic interventions. Our work offers insights for developing pharmacological strategies to combat senescence and cancer.

5.
Front Pharmacol ; 13: 932154, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091812

RESUMEN

Glucose-6-phosphate dehydrogenase (G6PD) is the only rate-limiting enzyme in the pentose phosphate pathway (PPP). Rapidly proliferating cells require metabolites from PPP to synthesize ribonucleotides and maintain intracellular redox homeostasis. G6PD expression can be abnormally elevated in a variety of cancers. In addition, G6PD may act as a regulator of viral replication and vascular smooth muscle function. Therefore, G6PD-mediated activation of PPP may promote tumor and non-neoplastic disease progression. Recently, studies have identified post-translational modifications (PTMs) as an important mechanism for regulating G6PD function. Here, we provide a comprehensive review of various PTMs (e.g., phosphorylation, acetylation, glycosylation, ubiquitination, and glutarylation), which are identified in the regulation of G6PD structure, expression and enzymatic activity. In addition, we review signaling pathways that regulate G6PD and evaluate the role of oncogenic signals that lead to the reprogramming of PPP in tumor and non-neoplastic diseases as well as summarize the inhibitors that target G6PD.

6.
Biomed Pharmacother ; 154: 113607, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36030587

RESUMEN

Transketolase (TKT) is an enzyme that is ubiquitously expressed in all living organisms and has been identified as an important regulator of cancer. Recent studies have shown that the TKT family includes the TKT gene and two TKT-like (TKTL) genes; TKTL1 and TKTL2. TKT and TKTL1 have been reported to be involved in the regulation of multiple cancer-related events, such as cancer cell proliferation, metastasis, invasion, epithelial-mesenchymal transition, chemoradiotherapy resistance, and patient survival and prognosis. Therefore, TKT may be an ideal target for cancer treatment. More importantly, the levels of TKTL1 were detected using EDIM technology for the early detection of some malignancies, and TKTL1 was more sensitive and specific than traditional tumor markers. Detecting TKTL1 levels before and after surgery could be used to evaluate the surgery's effect. While targeted TKT suppresses cancer in multiple ways, in some cases, it has detrimental effects on the organism. In this review, we discuss the role of TKT in different tumors and the detailed mechanisms while evaluating its value and limitations in clinical applications. Therefore, this review provides a basis for the clinical application of targeted therapy for TKT in the future, and a strategy for subsequent cancer-related research.


Asunto(s)
Neoplasias , Transcetolasa , Biomarcadores de Tumor/genética , Proliferación Celular , Humanos , Neoplasias/terapia , Transcetolasa/genética
7.
Clin Transl Oncol ; 24(11): 2045-2054, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35896782

RESUMEN

The somatic mutation of liver kinase B1 (LKB1) has been implicated in various tumors, which is reflected in the survival, proliferation, and metastasis of tumor cells. However, the regulation of LKB1 in lipid metabolism, a process that is involved in tumor progression is not completely clear. We conclude that LKB1 deficiency results in abnormal expression and activation of multiple molecules related to lipid metabolism which locate downstream of AMP-activated protein kinase (AMPK) or salt-induced kinase (SIK). Abnormal lipid metabolism induced by LKB1 deficiency contributes to the proliferation and metastasis of tumor cells through energy regulation.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Metabolismo de los Lípidos , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/metabolismo , Humanos , Hígado
8.
Mol Metab ; 44: 101131, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33278637

RESUMEN

BACKGROUND: Live kinase B1 (LKB1) is a tumor suppressor that is mutated in Peutz-Jeghers syndrome (PJS) and a variety of cancers. Lkb1 encodes serine-threonine kinase (STK) 11 that activates AMP-activated protein kinase (AMPK) and its 13 superfamily members, regulating multiple biological processes, such as cell polarity, cell cycle arrest, embryo development, apoptosis, and bioenergetics metabolism. Increasing evidence has highlighted that deficiency of LKB1 in cancer cells induces extensive metabolic alterations that promote tumorigenesis and development. LKB1 also participates in the maintenance of phenotypes and functions of normal cells through metabolic regulation. SCOPE OF REVIEW: Given the important role of LKB1 in metabolic regulation, we provide an overview of the association of metabolic alterations in glycolysis, aerobic oxidation, the pentose phosphate pathway (PPP), gluconeogenesis, glutamine, lipid, and serine induced by aberrant LKB1 signals in tumor progression, non-neoplastic diseases, and functions of immune cells. MAJOR CONCLUSIONS: In this review, we summarize layers of evidence demonstrating that disordered metabolisms in glucose, glutamine, lipid, and serine caused by LKB1 deficiency promote carcinogenesis and non-neoplastic diseases. The metabolic reprogramming resulting from the loss of LKB1 confers cancer cells with growth or survival advantages. Nevertheless, it also causes a metabolic frangibility for LKB1-deficient cancer cells. The metabolic regulation of LKB1 also plays a vital role in maintaining cellular phenotype in the progression of non-neoplastic diseases. In addition, lipid metabolic regulation of LKB1 plays an important role in controlling the function, activity, proliferation, and differentiation of several types of immune cells. We conclude that in-depth knowledge of metabolic pathways regulated by LKB1 is conducive to identifying therapeutic targets and developing drug combinations to treat cancers and metabolic diseases and achieve immunoregulation.


Asunto(s)
Carcinogénesis/genética , Carcinogénesis/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Quinasas de la Proteína-Quinasa Activada por el AMP , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis/fisiología , Metabolismo Energético/fisiología , Glucosa/metabolismo , Glutamina/metabolismo , Humanos , Metabolismo de los Lípidos , Lípidos , Ingeniería Metabólica , Redes y Vías Metabólicas/genética , Mutación , Neoplasias/genética , Neoplasias/metabolismo , Síndrome de Peutz-Jeghers/genética , Síndrome de Peutz-Jeghers/metabolismo , Serina/metabolismo , Transducción de Señal
9.
Shanghai Kou Qiang Yi Xue ; 28(1): 81-84, 2019 Feb.
Artículo en Chino | MEDLINE | ID: mdl-31081006

RESUMEN

PRUPOSE: To evaluate the clinical efficacy of prosthesis-like applicators with radioactive seeds in treatment of palatal glandular malignancy. METHODS: Eleven patients with palatal glandular malignant tumors were treated with surgical resection and postoperative 125I radioactive seed brachytherapy. After resection of the palatal tumors, a prosthesis denture was fabricated for each patient. According to the design of treatment plan system, several 125I radioactive seeds were embedded in the tissue surface of the prosthesis at the same time. All the patients were followed-up for 6 to 24 months and the results were evaluated. SPSS 21.0 software package was used for statistical analysis. RESULTS: Eleven patients could wear prosthesis-like applicators all the time and neither tumor recurrence nor metastasis were found around the target area during the follow-up period. Furthermore, significant improvement was shown in terms of speech, mastication and facial appearance for all patients after prosthesis-like applicator restorations. CONCLUSIONS: For patients with palatal glandular malignant tumors, prosthesis-like applicators with 125I radioactive seed brachytherapy may be effective for preventing recurrence and metastasis of the malignancies and improving the patients'quality of life.


Asunto(s)
Braquiterapia , Radioisótopos de Yodo , Neoplasias Glandulares y Epiteliales , Braquiterapia/métodos , Humanos , Radioisótopos de Yodo/uso terapéutico , Recurrencia Local de Neoplasia , Neoplasias Glandulares y Epiteliales/radioterapia , Calidad de Vida
10.
Cell Biochem Biophys ; 68(1): 181-4, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23733674

RESUMEN

The present study aimed to evaluate efficacy and adverse effects of Nimotuzumab combined with docetaxel-cisplatin-fluorouracil regimen in the treatment of advanced oral carcinoma. Nine patients with advanced oral carcinoma were treated with Nimotuzumab combined with docetaxel-cisplatin-fluorouracil regimen (test group). The treatment was given as follows: Nimotuzumab 200 mg, given as intravenous infusion once a week for 6 weeks; docetaxel and cisplatin, 75 mg/m(2) each, on day 1 only; 5-fluorouracil, 750 mg/m(2) infused continually for 8 h, used from day 1 to 5; the total cycle was for 21 days. Another eight patients comprised control group (docetaxel-cisplatin-fluorouracil regimen alone). Study patients from both groups were evaluated for objective response. The response rate was significantly (p = 0.044) higher in test group (88.9 vs. 37.5 % in control group). The disease control rate also tended to be higher in test group (100 vs. 62.5 % in control group; p = 0.083). The major adverse effects were bone marrow suppression, nausea, vomiting, and alopecia. The incidence of adverse effects was similar between both study groups. In conclusion, Nimotuzumab combined with docetaxel-cisplatin-fluorouracil regimen is effective and safe in the treatment of advanced oral carcinoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Anciano , Anciano de 80 o más Años , Alopecia/etiología , Anticuerpos Monoclonales Humanizados/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Recuento de Células Sanguíneas , Cisplatino/administración & dosificación , Docetaxel , Esquema de Medicación , Femenino , Fluorouracilo/administración & dosificación , Humanos , Inyecciones Intravenosas , Masculino , Persona de Mediana Edad , Náusea/etiología , Estadificación de Neoplasias , Taxoides/administración & dosificación , Resultado del Tratamiento
11.
Cell Biochem Biophys ; 67(3): 1529-32, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23709313

RESUMEN

We sought to evaluate the effect of (125)I radioactive seed implantation combined with prosthesis denture on the treatment of oral and maxillofacial malignancy. For this purpose, 10 patients with glandule palatine malignancy were selected and subjected to the treatment plan of radioactive seed implantation during CT examination. All patients were treated as follow. The tumor tissues were extracted first. After 2 weeks, radioactive seeds were implanted in the palatine tissue and the prosthesis denture was made and worn for the tissue defect. Several radioactive seeds were still embedded in the tissue surface of the prosthesis at the same time; 24-36 seeds (average: 28) were used for each patient. All patients were followed up for 3-16 months and the results were evaluated. We found no tumor recurrence or metastasis around the target area in all patients. Significant improvement was shown in terms of speech, mastication, and facial appearance in all cases. Therefore, we concluded that in patients with glandule palatine malignancy, tumorectomy followed by radioactive seed implantation and prosthesis denture are effective for preventing the recurrence and metastasis of malignancy and improving the quality of life.


Asunto(s)
Prótesis Dental , Neoplasias de la Boca/radioterapia , Radiofármacos/uso terapéutico , Adulto , Braquiterapia , Carcinoma/radioterapia , Carcinoma/cirugía , Femenino , Estudios de Seguimiento , Humanos , Radioisótopos de Yodo/química , Radioisótopos de Yodo/uso terapéutico , Masculino , Implantación de Prótesis Maxilofacial , Persona de Mediana Edad , Neoplasias de la Boca/cirugía , Radiofármacos/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA