Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 589
Filtrar
1.
Sci Rep ; 12(1): 304, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34997135

RESUMEN

To evaluate the effects of fructose diphosphate (FDP) on routine coagulation tests in vitro, we added FDP into the mixed normal plasma to obtain the final concentration of 0, 1, 2, 3, 4, 5, 6, 10, 15, 20, 25, 30 and 35 mg/mL of drug. Prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen (FBG) and thrombin time (TT) of samples were analyzed with blood coagulation analyzers from four different manufacturers(Sysmex, Stago, SEKISUI and Werfen) and their corresponding reagents, respectively. Before the experiment, we also observed whether there were significant differences in coagulation test results of different lots of reagents produced by each manufacturer. At the same time as the four routine clotting tests, the Sysmex blood coagulation analyzer and its proprietary analysis software were used to detect the change of maximum platelet aggregation rate in platelet-rich plasma after adding FDP (0, 1, 2, 3, 4, 5 and 6 mg/mL). The results of PT, aPTT and TT showed a FDP (0-35 mg/mL) concentration-dependent increase and a FBG concentration-dependent decrease. The degree of change (increase or decrease) varied depending on the assay system, with PT and aPTT being more affected by the Sysmex blood coagulation testing instrument reagent system and less affected by CEKISUI, TT less affected by CEKISUI and more affected by Stago, and FBG less affected by Stago and more affected by Sysmex. The results of PT, aPTT and TT were statistically positively correlated with their FDP concentrations, while FBG was negatively correlated. The correlation coefficients between FDP and the coagulation testing systems of Sysmex, Stago, Werfen and SEKISUI were 0.975, 0.988, 0.967, 0.986 for PT, and 0.993, 0.989, 0.990 and 0.962 for aPTT, 0.994, 0.960, 0.977 and 0.982 for TT, - 0.990, - 0.983, - 0.989 and - 0.954 for FBG, respectively. Different concentrations of FDP (0, 1, 2, 3, 4, 5 and 6 mg/mL) had different effects on the maximum aggregation rate of platelet induced by the agonists of adenosine diphosphate (ADP, 5 µmol/L), arachidonic acid (Ara, 1 mmol/L), collagen (Col, 2.5 µg/mL) and epinephrine (Epi,10 µmol/L), but the overall downward trend was consistent, that is, with the increase of FDP concentration, the platelet aggregation rate decreased significantly. Our experimental study demonstrated a possible effect of FDP on the assays of coagulation and Platelet aggregation, which may arise because the drug interferes with the coagulation and platelet aggregation detection system, or it may affect our in vivo coagulation system and Platelet aggregation function, the real mechanism of which remains to be further verified and studied.


Asunto(s)
Pruebas de Coagulación Sanguínea , Coagulación Sanguínea/efectos de los fármacos , Fructosadifosfatos/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Tiempo de Tromboplastina Parcial , Agregación Plaquetaria/efectos de los fármacos , Pruebas de Función Plaquetaria , Tiempo de Protrombina , Tiempo de Trombina
2.
Int J Mol Sci ; 22(3)2021 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-33540748

RESUMEN

Tuberculosis (TB) remains one of the major health concerns worldwide. Mycobacterium tuberculosis (Mtb), the causative agent of TB, can flexibly change its metabolic processes during different life stages. Regulation of key metabolic enzyme activities by intracellular conditions, allosteric inhibition or feedback control can effectively contribute to Mtb survival under different conditions. Phosphofructokinase (Pfk) is one of the key enzymes regulating glycolysis. Mtb encodes two Pfk isoenzymes, Pfk A/Rv3010c and Pfk B/Rv2029c, which are differently expressed upon transition to the hypoxia-induced non-replicating state of the bacteria. While pfkB gene and protein expression are upregulated under hypoxic conditions, Pfk A levels decrease. Here, we present biochemical characterization of both Pfk isoenzymes, revealing that Pfk A and Pfk B display different kinetic properties. Although the glycolytic activity of Pfk A is higher than that of Pfk B, it is markedly inhibited by an excess of both substrates (fructose-6-phosphate and ATP), reaction products (fructose-1,6-bisphosphate and ADP) and common metabolic allosteric regulators. In contrast, synthesis of fructose-1,6-bisphosphatase catalyzed by Pfk B is not regulated by higher levels of substrates, and metabolites. Importantly, we found that only Pfk B can catalyze the reverse gluconeogenic reaction. Pfk B thus can support glycolysis under conditions inhibiting Pfk A function.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mycobacterium tuberculosis/enzimología , Fosfofructoquinasas/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Difosfato/farmacología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Regulación Alostérica , Proteínas Bacterianas/antagonistas & inhibidores , Catálisis , Inducción Enzimática , Retroalimentación Fisiológica , Fructosadifosfatos/biosíntesis , Fructosadifosfatos/farmacología , Fructosafosfatos/metabolismo , Fructosafosfatos/farmacología , Gluconeogénesis , Glucólisis , Hexosafosfatos/metabolismo , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Cinética , L-Lactato Deshidrogenasa/metabolismo , Mycobacterium tuberculosis/efectos de los fármacos , Oxígeno/farmacología , Fosfofructoquinasas/antagonistas & inhibidores , Piruvato Quinasa/metabolismo , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato
3.
J Appl Toxicol ; 41(7): 1050-1062, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33078453

RESUMEN

Fructose-1,6-bisphosphate (F1,6BP), an intermediate of the glycolytic pathway, has been found to play a promising anticancer effect; nevertheless, the mechanisms involved remain poorly understood. The present study aimed to evaluate the effect and mechanisms of F1,6BP in a human endometrial cancer cell line (Ishikawa). F1,6BP showed an antiproliferative and non-cytotoxic effect on endometrial cancer cells. These effects are related to the increase in reactive oxygen species (ROS) levels and mitochondrial membrane potential (ΔΨm). These harmful stimuli trigger the upregulation of the expression of pro-apoptotic genes (p53 and Bax), leading to the reduction of cell proliferation through inducing programmed cell death by apoptosis. Furthermore, F1,6BP-treated cells had the formation of autophagosomes induced, as well as a decrease in their proliferative capacity after withdrawing the treatment. Our results demonstrate that F1,6BP acts as an anticancer agent through the generation of mitochondrial instability, loss of cell function, and p53-dependent cell death. Thus, F1,6BP proves to be a potential molecule for use in the treatment against endometrial cancer.


Asunto(s)
Antineoplásicos/farmacología , Fructosadifosfatos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/genética , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Endometriales , Femenino , Fructosa/farmacología , Humanos , Mitocondrias/efectos de los fármacos
4.
Can J Physiol Pharmacol ; 98(10): 733-740, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32551885

RESUMEN

This study aims to investigate the mechanisms through which fructose diphosphate (FDP) causes anti-hypoxia and anti-fatigue effects and improves learning and memory. Mice were divided into three groups: low-dose FDP (FDP-L), high-dose FDP (FDP-H), and a control group. Acute toxic hypoxia induced by carbon monoxide, sodium nitrite, and potassium cyanide and acute cerebral ischemic hypoxia were used to investigate the anti-hypoxia ability of FDP. The tests of rod-rotating, mouse tail suspension, and swimming endurance were used to explore the anti-fatigue effects of FDP. The Morris water maze experiment was used to determine the impact of FDP on learning and memory ability. Poisoning-induced hypoxic tests showed that mouse survival time was significantly prolonged in the FDP-L and FDP-H groups compared with the control group (p < 0.05). In the exhaustive swimming test, FDP significantly shortened struggling time and prolonged the time of mass-loaded swimming; the rod-rotating test showed that endurance time was significantly prolonged by using FDP (p < 0.05). FDP significantly decreased lactate and urea nitrogen levels and increased hepatic and muscle glycogen and glucose transporter-4 and Na+-K+-ATPase (p < 0.05). To conclude, FDP enhances hypoxia tolerance and fatigue resistance and improves learning and memory ability through regulating glucose and energy metabolism.


Asunto(s)
Conducta Animal/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Fatiga/prevención & control , Fructosadifosfatos/farmacología , Hipoxia-Isquemia Encefálica/prevención & control , Hipoxia/prevención & control , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Fatiga/metabolismo , Fatiga/fisiopatología , Fatiga/psicología , Hipoxia/metabolismo , Hipoxia/fisiopatología , Hipoxia/psicología , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/fisiopatología , Hipoxia-Isquemia Encefálica/psicología , Locomoción/efectos de los fármacos , Ratones , Prueba del Laberinto Acuático de Morris/efectos de los fármacos , Prueba de Desempeño de Rotación con Aceleración Constante , Natación
5.
J Cell Mol Med ; 23(10): 6846-6858, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31343107

RESUMEN

Pyruvate kinase M2 (PKM2), playing a central role in regulating aerobic glycolysis, was considered as a promising target for cancer therapy. However, its role in cancer metastasis is rarely known. Here, we found a tight relationship between PKM2 and breast cancer metastasis, demonstrated by the findings that beta-elemene (ß-elemene), an approved drug for complementary cancer therapy, exerted distinct anti-metastatic activity dependent on PKM2. The results indicated that ß-elemene inhibited breast cancer cell migration, invasion in vitro as well as metastases in vivo. ß-Elemene further inhibited the process of aerobic glycolysis and decreased the utilization of glucose and the production of pyruvate and lactate through suppressing pyruvate kinase activity by modulating the transformation of dimeric and tetrameric forms of PKM2. Further analysis revealed that ß-elemene suppressed aerobic glycolysis by blocking PKM2 nuclear translocation and the expression of EGFR, GLUT1 and LDHA by influencing the expression of importin α5. Furthermore, the effect of ß-elemene on migration, invasion, PKM2 transformation, and nuclear translocation could be reversed in part by fructose-1,6-bisphosphate (FBP) and L-cysteine. Taken together, tetrameric transformation and nuclear translocation of PKM2 are essential for cancer metastasis, and ß-elemene inhibited breast cancer metastasis via blocking aerobic glycolysis mediated by dimeric PKM2 transformation and nuclear translocation, being a promising anti-metastatic agent from natural compounds.


Asunto(s)
Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Núcleo Celular/metabolismo , Multimerización de Proteína , Piruvato Quinasa/metabolismo , Sesquiterpenos/farmacología , Aerobiosis , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Cisteína/farmacología , Receptores ErbB/metabolismo , Femenino , Fructosadifosfatos/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Transportador de Glucosa de Tipo 1/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Multimerización de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos
6.
Inflamm Res ; 68(5): 415-421, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30927049

RESUMEN

BACKGROUND: Although some glycolytic intermediates have been shown to modulate several cell type formation and activation, the functional role of fructose 1,6-bisphosphate (FBP) on osteoclastogenesis is still unknown. METHODS: Osteoclastogenesis was evaluated on bone marrow preosteoclasts cultured with M-CSF - 30 ng/ml, RANKL - 10 ng/ml, and two concentrations of FBP (100 and 300 µM). TRAP-positive stained cells were counted, and osteoclastogenic marker genes expression were evaluated by qPCR. Osteoclasts resorption capacity was evaluated by the expression of specific enzymes and capacity to resorb a mineralized matrix. The NF-κB activation was detected using RAW 264.7, stably expressing luciferase on the NF-κB responsive promoter. RESULTS: We show that FBP, the product of the first stage of glycolysis, inhibited RANKL-induced osteoclasts differentiation and TRAP activity. The treatment of preosteoclasts with FBP attenuated osteoclast fusion and formation, without affecting cell viability. Moreover, the inhibition of several osteoclastogenic marker genes expression (TRAP, OSCAR, DC-STAMP, Integrin αv, NFATc1) by FBP correlates with a reduction of mineralized matrix resorption capacity. The mechanism underlying FBP-inhibition of osteoclastogenesis involves NF-κB/NFATc1 signaling pathway inhibition. CONCLUSION: Altogether these data show a protective role of a natural glycolytic intermediate in bone homeostasis that may have therapeutic benefit for osteolytic diseases.


Asunto(s)
Fructosadifosfatos/farmacología , FN-kappa B/metabolismo , Factores de Transcripción NFATC/genética , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Ligando RANK/farmacología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Fémur/citología , Masculino , Ratones Endogámicos C57BL , Osteoclastos/citología , Tibia/citología
7.
Stem Cells Transl Med ; 8(6): 606-616, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30779327

RESUMEN

Fat grafting procedures are considered to be a promising regenerative, cell-directed therapy; however, their survival is mainly influenced by ischemia condition. Fructose 1,6-bisphosphate (FBP), as an intermediate in energy metabolism, has the potential to rescue cells and tissues from hypoxic-ischemic circumstances. In the present study, human lipoaspirates were grafted subcutaneously into nude mice followed by a daily intraperitoneal injection of FBP at different doses for 7 days. Next, the grafts were harvested at different time points till 12 weeks postimplantation and were evaluated for cell viability and function, tissue revascularization and inflammatory cell infiltration using histological analysis, whole-mount living tissue imaging, glycerol 3-phosphate dehydrogenase activity assays, and quantitative analysis of gene expression. The results demonstrated that exogenous FBP administration could attenuate the volume and weight reduction of fat graft; meanwhile, FBP enhanced adipocyte viability and function, increased blood vessel formation, and decreased inflammation. Moreover, in vitro cell experiments showed that FBP could promote adipose-derived stem cell viability and vascular endothelial growth factor (VEGF) mRNA expression in ischemia conditions. Our study indicates that FBP can be used as a protective agent for fat grafting and may be applied in stem cell-based regenerative medicine. Stem Cells Translational Medicine 2019;8:606-616.


Asunto(s)
Tejido Adiposo/trasplante , Fructosadifosfatos/farmacología , Supervivencia de Injerto/efectos de los fármacos , Sustancias Protectoras/farmacología , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/genética , Antígenos de Diferenciación Mielomonocítica/metabolismo , Hipoxia de la Célula , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Expresión Génica/efectos de los fármacos , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/metabolismo , Supervivencia de Injerto/fisiología , Humanos , Inyecciones Intraperitoneales , Ratones , Ratones Desnudos , Células Madre/citología , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo
8.
Inflammation ; 41(5): 1987-2001, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29995294

RESUMEN

Pulmonary fibrosis is a specific form of interstitial pneumonia. In addition to the idiopathic cause, it may be caused by drugs such as bleomycin (BLM)-used in the treatment of tumors. Fructose-1,6-bisphosphate (FBP) is a high-energy endogenous glycolytic compound that has antifibrotic, anti-inflammatory, and immunomodulatory effects. The aim of this study was to investigate the effects of FBP on both BLM-induced pulmonary fibrosis in mice and in a human embryonic lung fibroblast (MRC-5) culture system. C57BL/6 mice were divided into four groups: control, FBP, BLM, and BLM plus FBP. A single dose of bleomycin (7.5 U/kg) was administered intratracheally, and survival, body weight, Ashcroft score, and histological analysis were evaluated. Pulmonary function and bronchoalveolar lavage fluid (BALF) were also evaluated after a single dose of bleomycin (1.2 U/kg-intratracheally). Treatment with FBP (500 mg/kg) was given on day 0 intraperitoneally. Fibroblasts (MRC-5 cells) were used to access the effect of FBP in vitro. In vivo, FBP increased the survival rate and reduced body weight loss (BLM vs. BLM plus FBP-p < 0.05). FBP also prevented BLM-induced loss of pulmonary function and decreased BALF inflammatory cells, level of fibrosis, and superficial collagen density (p < 0.05). In vitro, FBP (0.62 and 1.25 mM) had inhibitory activity on MRC-5 cells and was able to induce senescence in fibroblasts. These results showed that FBP has the potential of reducing the toxic effects of BLM and may provide supportive therapy for conventional methods used for the treatment of cancer.


Asunto(s)
Fibroblastos/patología , Fructosadifosfatos/farmacología , Fibrosis Pulmonar/prevención & control , Animales , Bleomicina/farmacología , Línea Celular , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fructosadifosfatos/uso terapéutico , Humanos , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Fibrosis Pulmonar/inducido químicamente , Tasa de Supervivencia , Pérdida de Peso/efectos de los fármacos
9.
Brain Res ; 1698: 54-61, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-29932894

RESUMEN

Sepsis is one of the main causes of hospitalization and mortality in Intensive Care Units. One of the first manifestations of sepsis is encephalopathy, reported in up to 70% of patients, being associated with higher mortality and morbidity. The factors that cause sepsis-associated encephalopathy (SAE) are still not well known, and may be multifactorial, as perfusion changes, neuroinflammation, oxidative stress and glycolytic metabolism alterations. Fructose-1,6-bisphosphate (FBP), a metabolite of the glycolytic route, has been reported as neuroprotective agent. The present study used an experimental sepsis model in C57BL/6 mice. We used in vivo brain imaging to evaluate glycolytic metabolism through microPET scans and the radiopharmaceutical 18F-fluoro-2-deoxy-D-glucose (18F-FDG). Brain images were obtained before and 12 h after the induction of sepsis in animals with and without FBP treatment. We also evaluated the treatment effects in the brain oxidative stress by measuring the production of reactive oxygen species (ROS), the activity of catalase (CAT) and glutathione peroxidase (GPx), and the levels of fluorescent marker 2'7'-dichlorofluorescein diacetate (DCF). There was a significant decrease in brain glucose metabolism due to experimental sepsis. A significant protective effect of FBP treatment was observed in the cerebral metabolic outcomes. FBP also modulated the production of ROS, evidenced by reduced CAT activity and lower levels of DCF. Our results suggest that FBP may be a possible candidate in the treatment of SAE.


Asunto(s)
Fructosadifosfatos/farmacología , Glucosa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sepsis/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encefalopatías/tratamiento farmacológico , Modelos Animales de Enfermedad , Fluorodesoxiglucosa F18 , Fructosa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Sepsis/tratamiento farmacológico
10.
Sci Rep ; 7(1): 15052, 2017 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-29118438

RESUMEN

Lactate/lactic acid is an important chemical compound for the manufacturing of bioplastics. The unicellular cyanobacterium Synechocystis sp. PCC 6803 can produce lactate from carbon dioxide and possesses D-lactate dehydrogenase (Ddh). Here, we performed a biochemical analysis of the Ddh from this cyanobacterium (SyDdh) using recombinant proteins. SyDdh was classified into a cyanobacterial clade similar to those from Gram-negative bacteria, although it was distinct from them. SyDdh can use both pyruvate and oxaloacetate as a substrate and is activated by fructose-1,6-bisphosphate and repressed by divalent cations. An amino acid substitution based on multiple sequence alignment data revealed that the glutamine at position 14 and serine at position 234 are important for the allosteric regulation by Mg2+ and substrate specificity of SyDdh, respectively. These results reveal the characteristic biochemical properties of Ddh in a unicellular cyanobacterium, which are different from those of other bacterial Ddhs.


Asunto(s)
Sustitución de Aminoácidos , Proteínas Bacterianas/genética , Lactato Deshidrogenasas/genética , Synechocystis/genética , Regulación Alostérica , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Biocatálisis/efectos de los fármacos , Cationes Bivalentes/farmacología , Fructosadifosfatos/farmacología , Cinética , Lactato Deshidrogenasas/clasificación , Lactato Deshidrogenasas/metabolismo , Modelos Moleculares , Filogenia , Conformación Proteica , Homología de Secuencia de Aminoácido , Especificidad por Sustrato , Synechocystis/enzimología , Synechocystis/metabolismo
11.
Eur J Pharmacol ; 815: 274-281, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28943104

RESUMEN

Fructose 1, 6-diphosphate (FDP), a glycolytic intermediate,has been identified to possess antioxidant activities. Here we show the protective effect of FDP against alcohol-induced liver injury in mice and the underlying mechanisms. The in vivo experiments demonstrated that FDP, orally administered to mice, dose-dependently suppressed alcohol (50%, v/v, 12ml/kg)-induced increase of serum activities of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), serum triglyceride (TG) level and hepatic malondialdehyde (MDA) level. FDP also inhibited liver histological lesions induced by seven-day administration of alcohol to mice. In vitro study indicated that FDP inhibited ethanol-induced L02 cell apoptosis via reducing pro-caspase3 protein level and increasing poly ADP-ribose polymerase (PARP) cleavage. The mechanism analysis showed that FDP prevented ethanol-induced decrease of mouse antioxidant capability through inhibiting the reducion of the level of glutathione (GSH) and activities of superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR) and glutathione peroxidase (GSH-PX) in mouse livers, and suppressing the reducion of GSH level and SOD activity in L02 cells. FDP also enhanced alcohol metabolic rate through increasing alcohol dehydrogenase (ADH) activity and acetaldehyde dehydrogenase (ALDH) protein level, and down-regulating cytochrome p450 2E1 (CYP2E1). These results displayed that FDP protected mice from alcohol-induced liver injury, suggesting the potential activity of FDP in preventing alcoholic liver disease (ALD).


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Etanol/efectos adversos , Etanol/metabolismo , Fructosadifosfatos/farmacología , Estrés Oxidativo/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Masculino , Ratones , Ratones Endogámicos ICR
12.
Biometals ; 30(4): 549-558, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28639108

RESUMEN

Hepatic fibrosis is an extracellular matrix deposition by hepatic stellate cells (HSC). Fibrosis can be caused by iron, which will lead to hydroxyl radical production and cell damage. Fructose-1,6-bisphosphate (FBP) has been shown to deliver therapeutic effects in many pathological situations. In this work, we aimed to test the effects of FBP in HSC cell line, GRX, exposed to an excess of iron (Fe). The Fe-treatment increased cell proliferation and FBP reversed this effect, which was not due to increased necrosis, apoptosis or changes in cell cycle. Oil Red-O staining showed that FBP successfully increased lipid content and lead GRX cells to present characteristics of quiescent HSC. Fe-treatment decreased PPAR-γ expression and increased Col-1 expression. Both effects were reversed by FBP which also decreased TGF-ß1 levels in comparison to both control and Fe groups. FBP, also, did not present scavenger activity in the DPPH assay. The treatment with FBP resulted in decreased proliferation rate, Col-1 expression and TGF-ß1 release by HSC cells. Furthermore, activated PPAR-γ and increased lipid droplets induce cells to become quiescent, which is a key event to reversion of hepatic fibrosis. FBP also chelates iron showing potential to improve Cell redox state.


Asunto(s)
Compuestos Ferrosos/antagonistas & inhibidores , Fructosadifosfatos/farmacología , Células Estrelladas Hepáticas/efectos de los fármacos , Quelantes del Hierro/farmacología , Animales , Compuestos de Bifenilo/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Compuestos Ferrosos/farmacología , Regulación de la Expresión Génica , Células Estrelladas Hepáticas/citología , Células Estrelladas Hepáticas/metabolismo , Gotas Lipídicas/efectos de los fármacos , Gotas Lipídicas/metabolismo , Ratones , Oxidación-Reducción , PPAR gamma/genética , PPAR gamma/metabolismo , Picratos/química , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
13.
Hum Mutat ; 38(9): 1132-1143, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28407397

RESUMEN

Many studies of allosteric mechanisms use limited numbers of mutations to test whether residues play "key" roles. However, if a large percentage of the protein contributes to allosteric function, mutating any residue would have a high probability of modifying allostery. Thus, a predicted mechanism that is dependent on only a few residues could erroneously appear to be supported. We used whole-protein alanine-scanning mutagenesis to determine which amino acid sidechains of human liver pyruvate kinase (hL-PYK; approved symbol PKLR) contribute to regulation by fructose-1,6-bisphosphate (Fru-1,6-BP; activator) and alanine (inhibitor). Each nonalanine/nonglycine residue of hL-PYK was mutated to alanine to generate 431 mutant proteins. Allosteric functions in active proteins were quantified by following substrate affinity over a concentration range of effectors. Results show that different residues contribute to the two allosteric functions. Only a small fraction of mutated residues perturbed inhibition by alanine. In contrast, a large percentage of mutated residues influenced activation by Fru-1,6-BP; inhibition by alanine is not simply the reverse of activation by Fru-1,6-BP. Moreover, the results show that Fru-1,6-BP activation would be extremely difficult to elucidate using a limited number of mutations. Additionally, this large mutational data set will be useful to train and test computational algorithms aiming to predict allosteric mechanisms.


Asunto(s)
Alanina/farmacología , Fructosadifosfatos/farmacología , Mutación , Piruvato Quinasa/química , Piruvato Quinasa/genética , Algoritmos , Regulación Alostérica , Biología Computacional , Cristalografía por Rayos X , Activación Enzimática , Humanos , Modelos Moleculares , Piruvato Quinasa/metabolismo
14.
Biomed Pharmacother ; 89: 358-365, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28242545

RESUMEN

Hepatocellular carcinoma (HCC) is the most prevalent primary liver tumor that affects the world population. Liver cancer inevitably causes great harms and its treatment is extremely difficult. Its development is related to the existence of chronic liver injury, such as in cirrhosis. Cancer is a disease related to the process of inflammation so, research with anti-inflammatory agents has been performed for the development of anti-tumor drugs. Fructose-1,6-bisphosphate (FBP), a metabolite of the glycolytic route, has shown anti-inflammatory actions. The purpose of this study is to investigate the effect of FBP on HepG2 cells growth and inflammatory parameters. Results showed that FBP decreased the proliferation of HepG2 cells through trypan blue assay, without causing necrosis, shown by the intracellular release of LDH. By flow cytometry, we observed a significant IL-8 decrease which is closely related to the tumoral progression and chemotherapeutic resistance, especially in HCC. Then, we found, by RT-PCR, a high expression level of pro-apoptotic protein, such as Bax and p53, and decreased the expression levels of anti-apoptotic proteins, like Bcl-2 suggesting apoptosis. Finally, our results showed that FBP can be a potential therapeutic agent to slow the progress of HCC.


Asunto(s)
Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Fructosadifosfatos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-8/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Hep G2 , Humanos , Inflamación/metabolismo , Interleucina-8/genética , Estrés Oxidativo/efectos de los fármacos
15.
Plant Cell Physiol ; 58(1): 145-155, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28011870

RESUMEN

Glucitol, also known as sorbitol, is a major photosynthetic product in plants from the Rosaceae family. This sugar alcohol is synthesized from glucose-6-phosphate by the combined activities of aldose-6-phosphate reductase (Ald6PRase) and glucitol-6-phosphatase. In this work we show the purification and characterization of recombinant Ald6PRase from peach leaves. The recombinant enzyme was inhibited by glucose-1-phosphate, fructose-6-phosphate, fructose-1,6-bisphosphate and orthophosphate. Oxidizing agents irreversibly inhibited the enzyme and produced protein precipitation. Enzyme thiolation with oxidized glutathione protected the enzyme from insolubilization caused by diamide, while incubation with NADP+ (one of the substrates) completely prevented enzyme precipitation. Our results suggest that Ald6PRase is finely regulated to control carbon partitioning in peach leaves.


Asunto(s)
Aldehído Reductasa/metabolismo , Hojas de la Planta/enzimología , Proteínas de Plantas/metabolismo , Prunus domestica/enzimología , Aldehído Reductasa/antagonistas & inhibidores , Aldehído Reductasa/genética , Fructosadifosfatos/metabolismo , Fructosadifosfatos/farmacología , Fructosafosfatos/metabolismo , Fructosafosfatos/farmacología , Glucofosfatos/metabolismo , Glucofosfatos/farmacología , Disulfuro de Glutatión/metabolismo , Hexosafosfatos/metabolismo , Hexosafosfatos/farmacología , Immunoblotting , Cinética , Modelos Biológicos , NADP/metabolismo , Oxidantes/metabolismo , Oxidantes/farmacología , Fosfatos/metabolismo , Fosfatos/farmacología , Filogenia , Hojas de la Planta/genética , Proteínas de Plantas/clasificación , Proteínas de Plantas/genética , Prunus domestica/genética , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Compuestos de Sulfhidrilo/metabolismo
16.
Curr Med Chem ; 23(39): 4396-4417, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27758716

RESUMEN

In clinical and experimental settings, a great deal of effort is being made to protect cells and tissues against harmful conditions and to facilitate metabolic recovery following these insults. Much of the recent attention has focused on the protective role of a natural form of sugar, fructose 1,6-bisphosphate (F16bP). F16bP is a high-energy glycolytic intermediate that has been shown to exert a protective action in different cell types and tissues (including the brain, kidney, intestine, liver and heart) against various harmful conditions. For example, there is much evidence that it prevents neuronal damage due to hypoxia and ischemia. Furthermore, the cytoprotective effects of F16bP have been documented in lesions caused by chemicals or cold storage, in a decrease in mortality during sepsis shock and even in the prevention of bone loss in experimental osteoporosis. Intriguingly, protection in such a variety of targets and animal models suggests that the mechanisms induced by F16bP are complex and involve different pathways. In this review we will discuss the most recent theories concerning the molecular model of action of F16bP inside cells. These include its incorporation as an energy substrate, the mechanism for the improvement of ATP availability, and for preservation of organelle membrane stability and functionality. In addition we will present new evidences regarding the capacity of F16bP to decrease oxidative stress by limiting free radical production and improving antioxidant systems, including the role of nitric oxide in the protective mechanism induced by F16bP. Finally we will review the proposed mechanisms for explaining its anti-inflammatory, immunomodulatory and neuroprotective properties.


Asunto(s)
Células/efectos de los fármacos , Citoprotección/efectos de los fármacos , Fructosadifosfatos/farmacología , Animales , Células/citología , Células/inmunología , Células/metabolismo , Fructosadifosfatos/uso terapéutico , Humanos
17.
FEMS Microbiol Lett ; 363(17)2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27481705

RESUMEN

The effect of fructose 1,6-bisphosphate (Fru 1,6-P2) on the regulatory enzymes of pentose phosphate pathway of Escherichia coli was examined. Fru 1,6-P2 inhibited E. coli transaldolase (EC 2.2.1.2) competitively against fructose 6-phosphate and uncompetitively against erythrose 4-phosphate, whereas Fru 1,6-P2 did not affect glucose 6-phosphate dehydrogenase (EC 1.1.1.49) and 6-phosphogluconate dehydrogenase (EC 1.1.1.44). Kinetic results can be explained by assuming that transaldolase has two kinds of binding sites for Fru 1,6-P2: a competitive binding site for fructose 6-phosphate and a second binding site on the enzyme-erythrose 4-phosphate complex. Fru 1,6-P2 increased resulting from the stimulation of glycolysis, can inhibit transaldolase and further participates in the elevation of the concentration of ribose 5-phosphate that can be preferentially utilized for anabolic reaction in exponential phase of E. coli.


Asunto(s)
Escherichia coli/metabolismo , Fructosadifosfatos/metabolismo , Vía de Pentosa Fosfato/efectos de los fármacos , Transaldolasa/antagonistas & inhibidores , Sitios de Unión , Unión Competitiva , Escherichia coli/efectos de los fármacos , Escherichia coli/enzimología , Escherichia coli/crecimiento & desarrollo , Fructosadifosfatos/farmacología , Fructosafosfatos/farmacología , Glucosafosfato Deshidrogenasa/metabolismo , Glucólisis/efectos de los fármacos , Cinética , Fosfogluconato Deshidrogenasa/metabolismo , Ribosamonofosfatos/metabolismo , Fosfatos de Azúcar/farmacología , Transaldolasa/metabolismo
18.
J Chem Inf Model ; 56(6): 1184-1192, 2016 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-27227511

RESUMEN

Pyruvate kinase M2 (PKM2) plays a key role in tumor metabolism and regulates the rate-limiting final step of glycolysis. In tumor cells, there are two allosteric effectors for PKM2: fructose-1,6-bisphosphate (FBP) and serine. However, the relationship between FBP and serine for allosteric regulation of PKM2 is unknown. Here we constructed residue/residue fluctuation correlation network based on all-atom molecular dynamics simulations to reveal the regulation mechanism. The results suggest that the correlation network in bound PKM2 is distinctly different from that in the free state, FBP/PKM2, or Ser/PKM2. The community network analysis indicates that the information can freely transfer from the allosteric sites of FBP and serine to the substrate site in bound PKM2, while there exists a bottleneck for information transfer in the network of the free state. Furthermore, the binding free energy between the substrate and PKM2 for bound PKM2 is significantly lower than either of FBP/PKM2 or Ser/PKM2. Thus, a hypothesis of "synergistic allosteric mechanism" is proposed for the allosteric regulation of FBP and serine. This hypothesis was further confirmed by the perturbational and mutational analyses of community networks and binding free energies. Finally, two possible synergistic allosteric pathways of FBP-K433-T459-R461-A109-V71-R73-MG2-OXL and Ser-I47-C49-R73-MG2-OXL were identified based on the shortest path algorithm and were confirmed by the network perturbation analysis. Interestingly, no similar pathways could be found in the free state. The process targeting on the allosteric pathways can better regulate the glycolysis of PKM2 and significantly inhibit the progression of tumor.


Asunto(s)
Fructosadifosfatos/farmacología , Piruvato Quinasa/química , Piruvato Quinasa/metabolismo , Serina/farmacología , Regulación Alostérica/efectos de los fármacos , Sinergismo Farmacológico , Estabilidad de Enzimas/efectos de los fármacos , Simulación de Dinámica Molecular , Mutación , Conformación Proteica , Piruvato Quinasa/genética , Termodinámica
19.
Sci Rep ; 5: 15171, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26478088

RESUMEN

Fructose 1,6-bisphosphate (FBP) is an endogenous intermediate of the glycolytic pathway. Exogenous administration of FBP has been shown to exert protective effects in a variety of ischemic injury models, which are attributed to its ability to sustain glycolysis and increase ATP production. Here, we demonstrated that a single treatment with FBP markedly attenuated arthritis, assessed by reduction of articular hyperalgesia, joint swelling, neutrophil infiltration and production of inflammatory cytokines, TNF and IL-6, while enhancing IL-10 production in two mouse models of arthritis. Our mechanistic studies showed that FBP reduces joint inflammation through the systemic generation of extracellular adenosine and subsequent activation of adenosine receptor A2a (A2aR). Moreover, we showed that FBP-induced adenosine generation requires hydrolysis of extracellular ATP through the activity of the ectonucleosides triphosphate diphosphohydrolase-1 (ENTPD1, also known as CD39) and ecto-5'-nucleotidase (E5NT, also known as CD73). In accordance, inhibition of CD39 and CD73 abolished anti-arthritic effects of FBP. Taken together, our findings provide a new insight into the molecular mechanism underlying the anti-inflammatory effect of FBP, showing that it effectively attenuates experimental arthritis by activating the anti-inflammatory adenosinergic pathway. Therefore, FBP may represent a new therapeutic strategy for treatment of rheumatoid arthritis (RA).


Asunto(s)
Adenosina/metabolismo , Antiinflamatorios/farmacología , Artritis Experimental/metabolismo , Fructosadifosfatos/farmacología , Redes y Vías Metabólicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , 5'-Nucleotidasa/antagonistas & inhibidores , Antagonistas del Receptor de Adenosina A2/farmacología , Animales , Antígenos CD , Apirasa/antagonistas & inhibidores , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/etiología , Artritis Experimental/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Espacio Extracelular/metabolismo , Glucólisis , Masculino , Ratones , Receptor de Adenosina A2A/metabolismo , Fiebre Reumática/tratamiento farmacológico , Fiebre Reumática/metabolismo
20.
Int Immunopharmacol ; 26(1): 203-11, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25843256

RESUMEN

Fructose-1,6-bisphosphate (FBP) is a glycolytic intermediate with salutary effects in various brain injury models, but its neuroprotective mechanism is incompletely understood. In this study, we examined the effects of FBP on the expression of adhesion molecules in cerebrovascular endothelial cells and explored the possible mechanisms therein involved. FBP significantly down-regulated lipopolysaccharide (LPS)-induced expression of adhesion molecules and leukocyte adhesion to brain endothelial cells and inhibited NF-κB activity, which is implicated in the expression of adhesion molecules. FBP abrogated ICAM-1 expression and NF-κB activation induced by macrophage-activating lipopeptide 2-kDa (MALP-2) or overexpression of MyD88 or TRAF6. FBP suppressed TRAF6-induced phosphorylation of TAK1, IKKß and IκBα, but fail to affect NF-κB activity induced by ectopic expression of IKKß. In addition, LPS-induced IRAK-1 phosphorylation was inhibited by FBP, suggesting the presence of multiple molecular targets of FBP in MyD88-dependent signaling pathway. FBP significantly attenuated ICAM-1 expression and NF-κB activity induced by poly[I:C] or overexpression of TRIF or TBK1. FBP significantly repressed the expression of interferon-ß (IFN-ß) and the activation of IFN regulatory factor 3 (IRF3) induced by LPS, poly[I:C] or overexpression of TRIF or TBK1, but fail to affect IRF3 activity induced by ectopic expression of constitutively active IRF3. Overall, our results demonstrate that FBP modulates both MyD88- and TRIF-dependent signaling pathways of TLR4 and subsequent inflammatory responses in brain endothelial cells, providing insight into its neuroprotective mechanism in brain injury associated with inflammation.


Asunto(s)
Encéfalo/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Fructosadifosfatos/farmacología , Molécula 1 de Adhesión Intercelular/biosíntesis , Fármacos Neuroprotectores/farmacología , Receptor Toll-Like 4/metabolismo , Animales , Encéfalo/irrigación sanguínea , Encéfalo/inmunología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/inmunología , Línea Celular , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Ensayo de Inmunoadsorción Enzimática , Fructosadifosfatos/administración & dosificación , Humanos , Lipopolisacáridos/farmacología , Ratones , Fármacos Neuroprotectores/administración & dosificación , Transducción de Señal/efectos de los fármacos , Células U937
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA