Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cureus ; 16(8): e67176, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39295726

RESUMEN

This study aimed to assess the alterations in the temporomandibular joint among adult patients undergoing orthodontic treatment involving the extraction of four premolars. A cohort of 44 adults, with a mean age of 24.2 years, underwent orthodontic therapy that included quadruple premolar extractions. Cone-beam computed tomography scans were performed before and after the treatment to evaluate the temporomandibular joints. The three-dimensional assessment focused on the condylar position relative to the cranial base and the articular fossa, the axial condylar rotation, and the joint spaces. Notably, a significant posterior shift of the condyle was detected (P≤0.01), averaging a 0.41mm retraction. The posterior joint space narrowed by 0.32mm post-treatment. Additionally, a medial tilt of 0.62° in the condyle's long axis was observed in the frontal plane. No significant changes were recorded for the other condylar positions, rotations, or joint space dimensions. The findings suggest that orthodontic treatment with four premolar extractions may instigate condylar repositioning and rotation. These insights can inform refinements in treatment protocols.

2.
Gynecol Oncol ; 185: 148-155, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38422947

RESUMEN

OBJECTIVE: To evaluate the effects of DEAR weight management in overweight patients undergoing fertility-sparing treatment for endometrial cancer or atypical hyperplasia. METHODS: Women with endometrial cancer or atypical hyperplasia who received fertility-sparing treatment and had a body mass index of >25 kg/m2 were randomly allocated to the DEAR (DEAR weight management) and control (self weight management) groups. Body morphology and composition, glycolipid metabolism, and tumor outcomes were assessed in both groups before and at 3 and 6 months after intervention. RESULTS: Overall, 72 subjects were included (36 in each group). Following intervention, the DEAR group showed significantly lower median body weight (69.45 vs. 78.05), body mass index (26.19 vs. 29.15), lipid accumulation index (29.21 vs. 57.86), body fat mass (24.00 vs. 29.30), visceral fat area (112.5 vs. 133.3), and glycolipid metabolic indices (except high density lipoprotein) than the control group (P < 0.05) and showed a decreasing trend. The test group achieved significantly higher complete remission (88.46% vs. 57.14%; P < 0.05); the time to complete remission did not differ significantly (P > 0.05). CONCLUSIONS: DEAR weight management can improve the studied parameters and complete remission rates in this population. REGISTRATION: NCT06169449.


Asunto(s)
Neoplasias Endometriales , Preservación de la Fertilidad , Sobrepeso , Humanos , Femenino , Sobrepeso/complicaciones , Sobrepeso/metabolismo , Adulto , Neoplasias Endometriales/patología , Preservación de la Fertilidad/métodos , Índice de Masa Corporal , Hiperplasia Endometrial
3.
Brain Sci ; 13(12)2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-38137092

RESUMEN

Astrocytes are morphologically intricate cells and actively modulate the function of the brain. Through numerous fine processes, astrocytes come into contact with neurons, blood vessels, and other glia cells. Emerging evidence has shown that astrocytes exhibit brain regional diversity in their morphology, transcriptome, calcium signaling, and functions. However, little is known about the brain regional heterogeneity of astrocyte-astrocyte structural interaction. So far, the visualization and characterization of the morphological features of adjacent astrocytes have been difficult, and as a result, it is still well-accepted that astrocytes in the adult brain share non-overlapped territory. In contrast, employing an approach that combines viral labeling with dual-fluorescent dyes iontophoresis under brightfield and imaging using confocal microscopy allows for the efficient and specific labeling of adjacent astrocytes, enabling a comprehensive visualization of their fine processes and the degree of their territorial overlap. Our study in the hypothalamic regions of the brain revealed a marked spatial overlap among adjacent astrocytes, which differs from the conventional understanding based on more extensively studied regions, like the hippocampus. Additionally, we revealed the heterogeneity of the astrocyte-neuron ratio across brain regions and conducted an assessment of the photostability and labeling efficiency of fluorescent dyes used for labeling adjacent astrocytes. Our study provides new insights for studying the morphological heterogeneity of astrocytes across the central nervous system.

5.
iScience ; 26(1): 105597, 2023 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-36654857

RESUMEN

Behavioral responses to environmental risks create gains and losses. We use high-frequency datasets to elucidate such behavior responses against air pollution and find a "double-peaked" time pattern in reducing outdoor exposure and in increasing electricity consumption. Despite that one standard deviation increase in the Air Quality Index induces 2% less outdoor population and 6% more household electricity consumption at peak, most responses fail to match with the intra-day pollution peaks, implying ineffective exposure avoidance. We find an unbalanced trade-off between health benefits and energy co-damages. The behavior-induced change in annual residential power consumption (+1.01% to +1.20%) is estimated to be 20 times more than that in the population-based exposure (-0.02% to -0.05%), and generates 0.13-0.15 million more metric tons of citywide carbon emissions. Our results imply that by targeting peak pollution periods, policies can shrink the trade-off imbalance and achieve mutual improvements in exposure reduction and energy conservation.

6.
BMC Immunol ; 22(1): 11, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33509093

RESUMEN

BACKGROUND: Interleukin-17A (IL17A) is a proinflammatory cytokine critically involved in autoimmune diseases, and monoclonal antibodies of IL17A have been approved for clinical treatment of psoriasis. However, a usable psoriatic animal model has been always required for preclinical evaluation of IL17A antagonists. Imiquimod (IMQ)-induced psoriasis model is widely used in fundamental research, but it's not able to accurately show anti-psoriatic effect of IL17A antagonists with conventional modelling condition. RESULTS: On female C57BL/6 mice, with optimization on the usage of IMQ, positive control reagent and anti-mIL17A antibody, a 7-day model with proper testing window, acceptable disease severity as well as high repeatability was developed, and the efficacy of IL17A antagonist can be objectively evaluated by several qualitative and quantitative indices. Meanwhile, we validated the detailed involvement of IL17A signaling in disease progression, confirmed that the expression levels of IL17A and its related cytokines were induced by IMQ application, and its downstream cytokines can be inhibited by IL17A antagonist treatment. In further study, we revealed that IL17A was transient induced by IMQ and directly caused downstream signaling activation. This finding on the kinetical change of IL17A signaling will manifest the pharmacokinetics-pharmacodynamics investigation of IL17A antagonists. CONCLUSIONS: Our work presents the application of a convenient psoriatic animal model in the research and development of IL17A antagonists, meanwhile providing extra evidence for understanding IL17A's role in the progression of IMQ-induced psoriasis model, which manifest the research and development of IL17A antagonists.


Asunto(s)
Modelos Animales de Enfermedad , Interleucina-17/antagonistas & inhibidores , Psoriasis/tratamiento farmacológico , Animales , Citocinas/metabolismo , Evaluación Preclínica de Medicamentos , Femenino , Imiquimod/efectos adversos , Ratones , Ratones Endogámicos C57BL , Psoriasis/inducido químicamente , Psoriasis/inmunología , Transducción de Señal/efectos de los fármacos
7.
Cancer Sci ; 109(10): 3294-3304, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30151975

RESUMEN

Bevacizumab (Bv) can be used synergistically with fluoropyrimidine-based chemotherapy to treat colorectal cancer. Whether and how it affects the delivery of fluoropyrimidine drugs is unknown. The present study aimed to explore the effect of Bv on the delivery of 5-fluorouracil (5-FU) to tumors and the underlying mechanism from metabolic perspective. Bv enhanced the anti-tumor effects of 5-FU in LoVo colon cancer xenograft mice and increased the 5-FU concentration in tumors without affecting hepatic 5-FU metabolism. Interestingly, Bv remarkably upregulated thymidine phosphorylase (TP) in tumors, which mediated the metabolic activation of 5-FU. Although TP is reported to promote angiogenesis and resistance, the combination of Bv and 5-FU resulted in anti-angiogenesis and vessel normalization in tumors, indicating that the elevated TP mainly contributed to the enhanced response to 5-FU. Bv also induced TP upregulation in LoVo cancer cells. Treatment with vascular endothelial growth factor receptor 2 (VEGFR2) antagonist apatinib and VEGFR2 silencing further confirmed TP upregulation. Bv and apatinib both enhanced the cytotoxicity of 5-FU in LoVo cells, but there was no synergism with adriamycin and paclitaxel. We further demonstrated that the effect of Bv was dependent on VEGFR2 blockade and specificity protein 1 activation via MDM2 inhibition. In summary, Bv enhanced the accumulation of 5-FU in tumors and the cytotoxicity of 5-FU via TP upregulation. We provide data to better understand how Bv synergizes with 5-FU from metabolic perspective, and it may give clues to the superiority of Bv in combination with fluoropyrimidine drugs compared to other chemotherapeutic drugs in colon cancer.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bevacizumab/farmacología , Neoplasias del Colon/tratamiento farmacológico , Fluorouracilo/farmacología , Transducción de Señal/efectos de los fármacos , Timidina Fosforilasa/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bevacizumab/uso terapéutico , Línea Celular Tumoral , Neoplasias del Colon/patología , Sinergismo Farmacológico , Fluorouracilo/uso terapéutico , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/patología , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Piridinas/farmacología , Piridinas/uso terapéutico , Factor de Transcripción Sp1/metabolismo , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Cell Proteomics ; 17(8): 1531-1545, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29739823

RESUMEN

Butyrate is a short chain fatty acid present in a high concentration in the gut lumen. It has been well documented that butyrate, by serving as an energetic metabolite, promotes the proliferation of normal colonocytes while, by serving as a histone deacetylase inhibitor, epigenetically suppressing the proliferation of cancerous counterparts undergoing the Warburg effect. However, how butyrate interrupts the metabolism of colorectal cancer cells and ultimately leads to the suppression of cell proliferation remains unclear. Here, we employed a metabolomics-proteomics combined approach to explore the link between butyrate-mediated proliferation arrest and cell metabolism. A metabolomics study revealed a remodeled metabolic profile with pronounced accumulation of pyruvate, decreased glycolytic intermediates upstream of pyruvate and reduced levels of nucleotides in butyrate-treated HCT-116 cells. Supplementation of key metabolite intermediates directly affected cancer-cell metabolism and modulated the suppressive effect of butyrate in HCT-116 cells. By a Drug Affinity Responsive Target Stability (DARTS)-based quantitative proteomics approach, we revealed the M2 isoform of a pyruvate kinase, PKM2, as a direct binding target of butyrate. Butyrate activates PKM2 via promoting its dephosphorylation and tetramerization and thereby reprograms the metabolism of colorectal cancer cells, inhibiting the Warburg effect while favoring energetic metabolism. Our study thus provides a mechanistic link between PKM2-induced metabolic remodeling and the antitumorigenic function of butyrate and demonstrates a widely applicable approach to uncovering unknown protein targets for small molecules with biological functions.


Asunto(s)
Butiratos/farmacología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Piruvato Quinasa/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/enzimología , Activación Enzimática/efectos de los fármacos , Glucólisis/efectos de los fármacos , Humanos , Modelos Biológicos , Fosforilación/efectos de los fármacos , Multimerización de Proteína , Proteómica
9.
Acta Pharmacol Sin ; 39(3): 449-458, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28836581

RESUMEN

The use of ginseng extract as an adjuvant for cancer treatment has been reported in both animal models and clinical applications, but its molecular mechanisms have not been fully elucidated. Mitomycin C (MMC), an anticancer antibiotic used as a first- or second-line regimen in the treatment for non-small cell lung carcinoma (NSCLC), causes serious adverse reactions when used alone. Here, by using both in vitro and in vivo experiments, we provide evidence for an optimal therapy for NSCLC with total ginsenosides extract (TGS), which significantly enhanced the MMC-induced cytotoxicity against NSCLC A549 and PC-9 cells in vitro when used in combination with relatively low concentrations of MMC. A NSCLC xenograft mouse model was used to confirm the in vivo synergistic effects of the combination of TGS with MMC. Further investigation revealed that TGS could significantly reverse MMC-induced S-phase cell cycle arrest and inhibit Rad51-mediated DNA damage repair, which was evidenced by the inhibitory effects of TGS on the levels of phospho-MEK1/2, phospho-ERK1/2 and Rad51 protein and the translocation of Rad51 from the cytoplasm to the nucleus in response to MMC. In summary, our results demonstrate that TGS could effectively enhance the cytotoxicity of MMC against NSCLC cells in vitro and in vivo, thereby revealing a novel adjuvant anticancer mechanism of TGS. Combined treatment with TGS and MMC can significantly lower the required concentration of MMC and can further reduce the risk of side effects, suggesting a better treatment option for NSCLC patients.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Ginsenósidos/farmacología , Mitomicina/farmacología , Recombinasa Rad51/antagonistas & inhibidores , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Fosforilación/efectos de los fármacos , Recombinasa Rad51/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Pharmacol Sci ; 135(4): 148-155, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29233468

RESUMEN

Butyrate is a typical short chain fatty acid produced by gut microbiota of which the dysmetabolism has been consistently associated with colorectal diseases. However, whether butyrate affects metastatic colorectal cancer is not clear. In this study we investigated in vitro the effect of butyrate on motility, a significant metastatic factor of colorectal cancer cells and explored the potential mechanism. By using wound healing and transwell-based invasion models, we demonstrated that pretreatment of butyrate significantly inhibited motility of HCT116, HT29, LOVO and HCT8 cells, this activity was further attributed to deactivation of Akt1 and ERK1/2. Suberanilohydroxamic acid (SAHA), another HDAC inhibitor, mimicked the inhibitory effect of butyrate on cell motility and deactivation of Akt/ERK. Furthermore, by silencing of HDAC3 with siRNA, we confirmed dependence of butyrate's effect on HDAC3, the similar reduced cell motility observed under HDAC3 silencing also indicates the significance of HDAC itself in cell motility. In conclusion, we confirmed the HDAC3-relied activity of butyrate on inhibiting motility of colorectal cancer cells via deactivating Akt/ERK signaling. Our data indicate that modulating butyrate metabolism is an effective therapeutic strategy of metastatic colorectal cancer; and HDAC3 might be a novel target for management of colorectal cancer metastasis.


Asunto(s)
Butiratos/farmacología , Movimiento Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Histona Desacetilasas/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Butiratos/metabolismo , Butiratos/uso terapéutico , Línea Celular Tumoral , Inhibidores de Histona Desacetilasas , Histona Desacetilasas/fisiología , Humanos , Ácidos Hidroxámicos/farmacología , Terapia Molecular Dirigida , Metástasis de la Neoplasia , Vorinostat
11.
J Proteome Res ; 16(6): 2250-2261, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28467092

RESUMEN

Apoptosis and senescence are two types of cell fates in response to chemotherapy. Besides canonical pathways that mediate cell fates, cancer cell metabolism has been revealed as a crucial factor affecting cell fate decisions and thus represents a new target for antitumor therapy. Therefore, a comprehensive description of metabolic pathways underlying cell senescence and apoptosis in response to chemotherapy is highly demanded for therapeutic exploitation of both processes. Herein we employed a metabolomics-proteomics combined approach to identify metabolism-associated molecular events that mediate cellular responses to senescence and apoptosis using doxorubicin-treated human breast cancer cells MCF7 as models. Such biomics approach revealed that tricarboxylic acid cycle, pentose phosphate pathway, and nucleotide synthesis pathways were significantly upregulated in the senescent model, whereas fatty acid synthesis was reduced. In apoptotic cells, an overall reduced activity of major metabolic pathways was observed except for the arginine and proline pathway. Combinatorially, these data show the utility of biomics in exploring biochemical mechanism-based differences between apoptosis and senescence and reveal an unprecedented finding of the metabolic events that were induced for survival by facilitating ROS elimination and DNA damage repair in senescent cells, while they were downregulated in apoptotic cells when DNA damage was irreparable.


Asunto(s)
Apoptosis/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Metabolómica/métodos , Proteómica/métodos , Ciclo del Ácido Cítrico , Daño del ADN , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Ácidos Grasos/biosíntesis , Humanos , Células MCF-7 , Nucleótidos/biosíntesis , Vía de Pentosa Fosfato , Especies Reactivas de Oxígeno/metabolismo
12.
Acta Pharmacol Sin ; 38(2): 264-276, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27867186

RESUMEN

Shenmai injection (SMI) is a Chinese patent-protected injection, which was mainly made of Red Ginseng and Radix Ophiopogonis and widely used for treating coronary heart disease and tumors by boosting Qi and nourishing Yin. In this study we examined whether SMI could produce direct synergetic effects on the cytoxicity of adriamycin (ADR) and paclitaxel (PTX) in colorectal cancers in vivo and in vitro, and explored the underlying pharmacokinetic mechanisms. BALB/c nude mice with LoVo colon cancer xenografts were intraperitoneally injected with ADR (2 mg·kg-1·3d-1) or PTX (7.5 mg·kg-1·3d-1) with or without SMI (0.01 mL·g-1·d-1) for 13 d. Co-administration of SMI significantly enhanced the chemotherapeutic efficacy of ADR and PTX, whereas administration of SMI alone at the given dosage did not produce visible anti-cancer effects, The chemosensitizing action of SMI was associated with increased concentrations of ADR and PTX in the plasma and tumors. In Caco-2 and LoVo cells in vitro, co-treatment with SMI (2 µL/mL) significantly enhanced the cytotoxicity of ADR and PTX, and resulted in some favorable pharmacokinetic changes in the subcellular distribution of ADR and PTX. In addition, SMI-induced intracellular accumulation of ADR was closely correlated with the increased expression levels of P-glycoprotein in 4 colon cancer cell lines (r2=+0.8558). SMI enhances the anti-cancer effects of ADR and PTX in colon cancers in vivo and in vitro by improving the subcellular distributions of ADR and PTX.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Doxorrubicina/farmacología , Doxorrubicina/farmacocinética , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/farmacocinética , Paclitaxel/farmacología , Paclitaxel/farmacocinética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Doxorrubicina/sangre , Combinación de Medicamentos , Sinergismo Farmacológico , Medicamentos Herbarios Chinos/análisis , Humanos , Ratones , Paclitaxel/sangre , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Chin J Nat Med ; 14(8): 582-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27608947

RESUMEN

Nicotinamide phosphoribosyltransferase (NAMPT) catalyzes the first rate-limiting step in converting nicotinamide to NAD(+), essential for a number of enzymes and regulatory proteins involved in a variety of cellular processes, including deacetylation enzyme SIRT1 which modulates several tumor suppressors such as p53 and FOXO. Herein we report that NQO1 substrates Tanshione IIA (TSA) and ß-lapachone (ß-lap) induced a rapid depletion of NAD(+) pool but adaptively a significant upregulation of NAMPT. NAMPT inhibition by FK866 at a nontoxic dose significantly enhanced NQO1-targeting agent-induced apoptotic cell death. Compared with TSA or ß-lap treatment alone, co-treatment with FK866 induced a more dramatic depletion of NAD(+), repression of SIRT1 activity, and thereby the increased accumulation of acetylated FOXO1 and the activation of apoptotic pathway. In conclusion, the results from the present study support that NAMPT inhibition can synergize with NQO1 activation to induce apoptotic cell death, thereby providing a new rationale for the development of combinative therapeutic drugs in combating non-small lung cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Citocinas/metabolismo , Inhibidores Enzimáticos/farmacología , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Abietanos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/fisiopatología , Línea Celular Tumoral , Citocinas/antagonistas & inhibidores , Citocinas/genética , Humanos , NAD/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/genética , Naftoquinonas/farmacología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/genética
14.
Oncotarget ; 7(38): 62503-62519, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27566573

RESUMEN

Tryptophan metabolism is essential in diverse kinds of tumors via regulating tumor immunology. However, the direct role of tryptophan metabolism and its signaling pathway in cancer cells remain largely elusive. Here, we establish a mechanistic link from L-type amino acid transporter 1 (LAT1) mediated transport of tryptophan and the subsequent de-novo NAD+ synthesis to SIRT1-FOXO1 regulated apoptotic signaling in A549 cells in response to NQO1 activation. In response to NQO1 activation, SIRT1 is repressed leading to the increased cellular accumulation of acetylated FOXO1 that transcriptionally activates apoptotic signaling. Decreased uptake of tryptophan due to the downregulation of LAT1 coordinates with PARP-1 hyperactivation to induce rapid depletion of NAD+ pool. Particularly, the LAT1-NAD+-SIRT1 signaling is activated in tumor tissues of patients with non-small cell lung cancer. Because NQO1 activation is characterized with oxidative challenge induced DNA damage, these results suggest that LAT1 and de-novo NAD+ synthesis in NSCLC cells may play essential roles in sensing excessive oxidative stress.


Asunto(s)
Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteína Forkhead Box O1/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Neoplasias Pulmonares/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , NAD/biosíntesis , Sirtuina 1/metabolismo , Células A549 , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Estrés Oxidativo , Oxígeno/metabolismo , Transducción de Señal , Triptófano/metabolismo
15.
PLoS One ; 10(2): e0117051, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25692465

RESUMEN

ß-lapachone (ß-lap), an NAD(P)H: quinone oxidoreductase 1 (NQO1) targeting antitumor drug candidate in phase II clinical trials, is metabolically eliminated via NQO1 mediated quinone reduction and subsequent UDP-glucuronosyltransferases (UGTs) catalyzed glucuronidation. This study intends to explore the inner link between the cellular glucuronidation and pharmacokinetics of ß-lap and its apoptotic effect in human colon cancer cells. HT29 cells S9 fractions exhibited high glucuronidation activity towards ß-lap, which can be inhibited by UGT1A9 competitive inhibitor propofol. UGT1A siRNA treated HT29 cells S9 fractions displayed an apparent low glucuronidation activity. Intracellular accumulation of ß-lap in HCT116 cells was much higher than that in HT29 cells, correlated with the absence of UGT1A in HCT116 cells. The cytotoxic and apoptotic effect of ß-lap in HT29 cells were much lower than that in HCT116 cells; moreover, ß-lap triggered activation of SIRT1-FOXO1 apoptotic pathway was observed in HCT116 cells but not in HT29 cells. Pretreatment of HT29 cells with UGT1A siRNA or propofol significantly decreased ß-lap's cytotoxic and apoptotic effects, due to the repression of glucuronidation and the resultant intracellular accumulation. In conclusion, UGT1A is an important determinant, via switching NQO1-triggered redox cycle to metabolic elimination, in the intracellular accumulation of ß-lap and thereafter its cytotoxicity in human colon cancer cells. Together with our previous works, we propose that UGTs determined cellular pharmacokinetics is an important determinant in the apoptotic effects of NQO1 targeting substrates serving as chemotherapeutic drugs.


Asunto(s)
Antineoplásicos/metabolismo , Neoplasias del Colon/patología , Glucuronosiltransferasa/metabolismo , Espacio Intracelular/metabolismo , Naftoquinonas/metabolismo , Naftoquinonas/farmacología , Apoptosis/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/metabolismo , Ácido Glucurónico/metabolismo , Células HCT116 , Células HT29 , Humanos , Espacio Intracelular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Sirtuina 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA