Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cereb Cortex ; 34(2)2024 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-38220573

RESUMEN

Diabetes mellitus (DM) causes damage to the central nervous system, resulting in cognitive impairment. Fibroblast growth factor 21 (FGF21) exhibits the potential to alleviate neurodegeneration. However, the therapeutic effect of intracerebroventricular (i.c.v) FGF21 infusion on diabetes-induced cognitive decline (DICD) and its potential mechanisms remain unclear. In this study, the impact of FGF21 on DICD was explored, and 1H nuclear magnetic resonance (NMR)-based metabolomics plus 13C NMR spectroscopy in combine with intravenous [1-13C]-glucose infusion were used to investigate the underlying metabolic mechanism. Results revealed that i.c.v FGF21 infusion effectively improved learning and memory performance of DICD mice; neuron loss and apoptosis in hippocampus and cortex were significantly blocked, suggesting a potential neuroprotective role of FGF21 in DICD. Metabolomics results revealed that FGF21 modulated DICD metabolic alterations related to glucose and neurotransmitter metabolism, which are characterized by distinct recovered enrichment of [3-13C]-lactate, [3-13C]-aspartate, [4-13C]-glutamine, [3-13C]-glutamine, [4-13C]-glutamate, and [4-13C]- γ-aminobutyric acid (GABA) from [1-13C]-glucose. Moreover, diabetes-induced neuron injury and metabolic dysfunctions might be mediated by PI3K/AKT/GSK-3ß signaling pathway inactivation in the hippocampus and cortex, which were activated by i.c.v injection of FGF21. These findings indicate that i.c.v FGF21 infusion exerts its neuroprotective effect on DICD by remodeling cerebral glucose and neurotransmitter metabolism by activating the PI3K/AKT/GSK-3ß signaling pathway.


Asunto(s)
Disfunción Cognitiva , Diabetes Mellitus , Factores de Crecimiento de Fibroblastos , Ratones , Animales , Glutamina/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas , Ácido Glutámico/metabolismo , Glucosa/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/etiología , Neurotransmisores
2.
FASEB J ; 37(9): e23134, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37561545

RESUMEN

3-Phosphoinositide-dependent protein kinase-1 (Pdk1) as a serine/threonine protein kinase plays a critical role in multiple signaling pathways. Analysis of the gene expression omnibus database showed that Pdk1 was significantly downregulated in patients with heart diseases. Gene set enrichment analysis of the proteomics dataset identified apoptotic- and metabolism-related signaling pathways directly targeted by Pdk1. Previously, our research indicated that Pdk1 deletion-induced metabolic changes might be involved in the pathogenesis of heart failure; however, the underlying mechanism remains elusive. Here, we demonstrated that deficiency of Pdk1 resulted in apoptosis, oxidative damage, and disturbed metabolism, both in vivo and in vitro. Furthermore, profiling of metabonomics by 1 H-NMR demonstrated that taurine was the major differential metabolite in the heart of Pdk1-knockout mice. Taurine treatment significantly reduced the reactive oxygen species production and apoptosis, improved cardiac function, and prolonged the survival time in Pdk1 deficient mice. Proteomic screening identified solute carrier family 6 member 6 (Slc6a6) as the downstream that altered taurine levels in Pdk1-expression cells. Consistently, cellular apoptosis and oxidative damage were rescued by Slc6a6 in abnormal Pdk1 expression cells. These findings collectively suggest that Pdk1 deficiency induces heart failure via disturbances in taurine homeostasis, triggered by Slc6a6.


Asunto(s)
Insuficiencia Cardíaca , Proteínas Quinasas , Animales , Ratones , Proteínas Quinasas Dependientes de 3-Fosfoinosítido , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Homeostasis , Ratones Noqueados , Proteómica , Taurina , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética
3.
Int J Cancer ; 150(9): 1525-1536, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-34985768

RESUMEN

The acquisition of ectopic type I fibroblast growth factor receptor (FGFR1) is a common feature of prostate cancer (PCa), the most frequently diagnostic cancer in men. However, how ectopic FGFR1 contributes to PCa progression is not well understood. In our study we showed that ablation of FGFR1 in DU145 human PCa cells changed the cell metabolite profile. Among the changes, the choline metabolism profile was the most significantly altered by FGFR1 ablation. Detailed characterization revealed that ablation of FGFR1 altered expression of multiple choline metabolism enzymes. Among the changes of FGFR1-regulated choline metabolic enzymes, downregulation of choline kinase α (CHKA) is the most prominent changes, which phosphorylates free choline to phosphocholine. Ablation of FGFR1 blunted the activity of choline to promote cell proliferation and survival. Furthermore, depletion of CHKA compromised FGF signaling activity in DU145 cells. We also first time demonstrated that FGFR1 formed complex with CHKA, suggesting that FGFR1 regulated CHKA at the posttranslational level. Together with the previous report that ectopic FGFR1 contributes to PCa progression and metastasis, our results here unravel a novel mechanism by which FGFR1 promotes PCa progression by dysregulating choline metabolism, and that the crosstalk between FGFR1-choline metabolism can be a potential target for managing PCa progression.


Asunto(s)
Colina , Neoplasias de la Próstata , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Línea Celular Tumoral , Proliferación Celular , Colina/metabolismo , Humanos , Masculino , Próstata/patología , Neoplasias de la Próstata/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
4.
J Proteome Res ; 20(8): 3900-3912, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34237942

RESUMEN

Sex differences in obesity have been well established, but the metabolic mechanism underlying these differences remains unclear. In the present study, we determined the expression levels of endogenous fibroblast growth factor 21 (FGF21) and its related receptors in male and female mice that were fed a high-fat diet (HFD) for 12 weeks. We also analyzed the metabolic changes in serum and livers using a nuclear magnetic resonance-based metabolomics approach. Reverse transcription polymerase chain reaction and western blotting results revealed that the levels of FGFR1, FGFR2, and co-factor ß-klotho were upregulated in female mice to alleviate FGF21 resistance induced by HFD. The metabolomics results demonstrated that the serum and liver metabolic patterns of HFD-fed male mice were significantly separated from those of the female HFD-fed group and the normal diet group. Furthermore, low-density lipoprotein/very low density lipoprotein and betaine levels were associated with the resistance of exogenous HFD in female mice. These findings imply that sex-based differences in metabolism and susceptibility to obesity might be mediated by the FGF21 signaling pathway.


Asunto(s)
Dieta Alta en Grasa , Factores de Crecimiento de Fibroblastos , Animales , Dieta Alta en Grasa/efectos adversos , Femenino , Factores de Crecimiento de Fibroblastos/genética , Hígado , Masculino , Metabolómica , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Transducción de Señal
5.
J Proteome Res ; 20(11): 5024-5035, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34699241

RESUMEN

Diabetic enteropathy (DE) is a diabetic complication and affects the quality of life for which there are limited therapies. In this study, db/db mice were administered with a basic fibroblast growth factor (bFGF) to explore its therapeutic effect on the intestine. 1H NMR-based metabolomics was applied to investigate the metabolic pattern. H&E and PAS staining were used to observe the morphological phenotypes related to intestinal barrier function. Tight junction proteins such as Zo-1 and Occluding were successively tested by immunofluorescence and real-time PCR. We found that bFGF treatment significantly restored intestinal barrier function. In addition, the administration of bFGF decreased the levels of inflammatory cytokines in the cecum. Metabolomic results show that bFGF remodeled metabolic phenotypes of the colon, cecum, and small intestine in db/db mice, including energy metabolism, short chain fatty acid metabolism, amino acid metabolism, and choline metabolism. Hence, this study indicates that the bFGF has a protective effect in diabetic bowel disease by restoring intestinal barrier function, reducing inflammatory infiltration, and remodeling metabolic function.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos , Calidad de Vida , Animales , Factor 2 de Crecimiento de Fibroblastos/genética , Intestinos , Metabolómica , Ratones , Espectroscopía de Protones por Resonancia Magnética
6.
FASEB J ; 34(10): 13333-13344, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32816365

RESUMEN

The deregulation of S100A2 has been implicated in the pathogenesis of several types of cancers. However, the molecular mechanisms underlying the protumorigenic capacities of S100A2 have not been fully elucidated. Here, we demonstrated the molecular mechanisms underlying the roles of S100A2 in glycolysis reprogramming and proliferation of colorectal cancer (CRC) cells. The results indicated that S100A2 overexpression raises glucose metabolism and proliferation. Mechanistically, S100A2 activated the PI3K/AKT signaling pathway, upregulated GLUT1 expression, induced glycolytic reprogramming, and consequently increased proliferation. Clinical data showed significantly increased S100A2 levels in CRC tissues and the Oncomine database. In addition, analysis revealed a positive correlation between S100A2 and GLUT1 mRNA expression in CRC tissues. Together, these results demonstrate that the S100A2/GLUT1 axis can promote the progression of CRC by modulating glycolytic reprogramming. Our results further suggest that targeting S100A2 could present a promising therapeutic avenue for the prevention of colorectal cancer progression.


Asunto(s)
Proliferación Celular , Factores Quimiotácticos/metabolismo , Neoplasias Colorrectales/metabolismo , Transportador de Glucosa de Tipo 1/metabolismo , Glucólisis , Proteínas S100/metabolismo , Animales , Transportador de Glucosa de Tipo 1/genética , Células HCT116 , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
7.
J Proteome Res ; 19(8): 3011-3021, 2020 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-32450697

RESUMEN

Diabetes mellitus (DM) can cause systemic metabolic disorders, but the impact of gender on DM-related metabolic changes is rarely reported. Herein, we analyzed metabolic alterations in the heart, liver, and kidney of male and female mice from normal to diabetes via a 1H NMR-based metabolomics method and aimed to investigate sex-specific metabolic mechanisms underlying the onset and development of diabetes and its complications. Our results demonstrate that male mice had more significant metabolic disorders from normal to diabetes than female mice. Moreover, the kidney was found as the major organ of metabolic disorders during the development of diabetes, followed by the liver and heart. These altered metabolites were mainly implicated in energy metabolism as well as amino acid, choline, and nucleotide metabolism. Therefore, this study suggests that the kidney is the primary organ affected by diabetes in a sex-specific manner, which provides a metabolic view on the pathogenesis of diabetic kidney diseases between genders.


Asunto(s)
Diabetes Mellitus Experimental , Nefropatías Diabéticas , Animales , Femenino , Hígado , Espectroscopía de Resonancia Magnética , Masculino , Metabolómica , Ratones
8.
Mol Cell Proteomics ; 17(12): 2335-2346, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30171160

RESUMEN

Diabetes mellitus causes brain structure changes and cognitive decline, and it has been estimated that diabetes doubles the risk for dementia. Until now, the pathogenic mechanism of diabetes-associated cognitive decline (DACD) has remained unclear. Using metabolomics, we show that lactate levels increased over time in the hippocampus of rats with streptozotocin-induced diabetes, as compared with age-matched control rats. Additionally, mRNA levels, protein levels, and enzymatic activity of lactate dehydrogenase-A (LDH-A) were significantly up-regulated, suggesting increased glycolysis activity. Importantly, by specifically blocking the glycolysis pathway through an LDH-A inhibitor, chronic diabetes-induced memory impairment was prevented. Analyzing the underlying mechanism, we show that the expression levels of cAMP-dependent protein kinase and of phosphorylated transcription factor cAMP response element-binding proteins were decreased in 12-week diabetic rats. We suggest that G protein-coupled receptor 81 mediates cognitive decline in the diabetic rat. In this study, we report that progressively increasing lactate levels is an important pathogenic factor in DACD, directly linking diabetes to cognitive dysfunction. LDH-A may be considered as a potential target for alleviating or treating DACD in the future.


Asunto(s)
Disfunción Cognitiva/metabolismo , Diabetes Mellitus Experimental/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Ácido Láctico/metabolismo , Análisis de Varianza , Animales , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/prevención & control , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/prevención & control , Glucólisis , Hipocampo/metabolismo , Inmunohistoquímica , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Lactato Deshidrogenasa 5 , Imagen por Resonancia Magnética , Masculino , Aprendizaje por Laberinto , Metabolómica/métodos , Análisis Multivariante , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Estreptozocina/farmacología
9.
Biochem J ; 476(13): 1943-1954, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31208986

RESUMEN

Metabolic remodeling plays an essential role in the pathophysiology of heart failure (HF). Many studies have shown that the disruption of phosphoinositide-dependent protein kinase-1 (PDK1) caused severe and lethal HF; however, the metabolic pattern of PDK1 deletion remains ambiguous. 1H nuclear magnetic resonance-based metabolomics was applied to explore the altered metabolic pattern in Pdk1-deficient mice. Principle component analysis showed significant separation as early as 4 weeks of age, and dysfunction of metabolism precedes a morphological change in Pdk1-deficient mice. A time trajectory plot indicated that disturbed metabolic patterns were related to the pathological process of the HF in Pdk1-deficient mice, rather than the age of mice. Metabolic profiles demonstrated significantly increased levels of acetate, glutamate, glutamine, and O-phosphocholine in Pdk1 deletion mice. Levels of lactate, alanine, glycine, taurine, choline, fumarate, IMP, AMP, and ATP were significantly decreased compared with controls. Furthermore, PDK1 knockdown decreased the oxygen consumption rate in H9C2 cells as determined using a Seahorse XF96 Analyzer. These findings imply that the disruption of metabolism and impaired mitochondrial activity might be involved in the pathogenesis of HF with PDK1 deletion.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/deficiencia , Insuficiencia Cardíaca/metabolismo , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Consumo de Oxígeno , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/deficiencia , Animales , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Ratones , Ratones Noqueados , Mitocondrias Cardíacas/genética , Mitocondrias Cardíacas/patología , Miocitos Cardíacos/patología
10.
Cell Physiol Biochem ; 48(3): 934-946, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30036879

RESUMEN

BACKGROUND/AIMS: Diabetic cardiomyopathy (DCM) is a serious complication of diabetes. It is therefore crucial to elucidate the characteristic metabolic changes that occur during the development of diabetes to gain an understanding the pathogenesis of this disease and identify potential drug targets involved. METHODS: 1H nuclear magnetic resonance-based metabonomics combined with HPLC measurements were used to determine the metabolic changes in isolated cardiac tissues after 5 weeks, 9 weeks, and 15 weeks in rats treated with streptozotocin. RESULTS: Pattern recognition analysis clearly discriminated the diabetic rats from time-matched control rats, suggesting that the metabolic profile of the diabetic group was markedly different from that of the controls. Quantitative analysis showed that the levels of energy metabolites, such as the high-energy phosphate pool (ATP and creatine), significantly decreased in a time-dependent manner. Correlation analysis revealed the inhibition of glycolysis and the tricarboxylic acid (TCA) cycle, enhanced lipid metabolism, and changes in some amino acids, which may have led to the decline in energy production in the DCM rats. CONCLUSIONS: The results indicated that the administration of energy substances or the manipulation of myocardial energy synthesis induced by increased glucose oxidation may contribute to the amelioration of cardiac dysfunction in diabetes.


Asunto(s)
Cardiomiopatías Diabéticas/patología , Metabolismo Energético , Metaboloma , Adenosina Trifosfato/análisis , Aminoácidos/análisis , Aminoácidos/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Creatina/análisis , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/patología , Cardiomiopatías Diabéticas/etiología , Análisis Discriminante , Análisis de los Mínimos Cuadrados , Espectroscopía de Resonancia Magnética , Masculino , Metabolómica , Miocardio/patología , Ratas , Ratas Sprague-Dawley
11.
Neural Plast ; 2018: 6473728, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29849562

RESUMEN

Diabetic patients often present with comorbid depression. However, the pathogenetic mechanisms underlying diabetic depression (DD) remain unclear. To explore the mechanisms underpinning the pathogenesis of the disease, we used ex vivo 1H nuclear magnetic resonance spectroscopy and immunohistochemistry to investigate the main metabolic and pathological changes in various rat brain areas in an animal model of DD. Compared with the control group, rats in the DD group showed significant decreases in neurotransmitter concentrations of glutamate (Glu) and glutamine (Gln) in the prefrontal cortex (PFC), hippocampus, and hypothalamus and aspartate and glycine in the PFC and hypothalamus. Gamma-aminobutyric acid (GABA) was decreased only in the hypothalamus. Levels of the energy product, lactate, were higher in the PFC, hippocampus, and hypothalamus of rats with DD than those in control rats, while creatine was lower in the PFC and hippocampus, and alanine was lower in the hypothalamus. The levels of other brain metabolites were altered, including N-acetyl aspartate, taurine, and choline. Immunohistochemistry analysis revealed that expressions of both glutamine synthetase and glutaminase were decreased in the PFC, hippocampus, and hypothalamus of rats with DD. The metabolic changes in levels of Glu, Gln, and GABA indicate an imbalance of the Glu-Gln metabolic cycle between astrocytes and neurons. Our results suggest that the development of DD in rats may be linked to brain metabolic changes, including inhibition of the Glu-Gln cycle, increases in anaerobic glycolysis, and disturbances in the lactate-alanine shuttle, and associated with dysfunction of neurons and astrocytes.


Asunto(s)
Depresión/metabolismo , Complicaciones de la Diabetes/metabolismo , Hipocampo/metabolismo , Hipotálamo/metabolismo , Corteza Prefrontal/metabolismo , Animales , Depresión/etiología , Modelos Animales de Enfermedad , Glutamato-Amoníaco Ligasa/metabolismo , Glutaminasa/metabolismo , Masculino , Espectroscopía de Protones por Resonancia Magnética , Ratas Wistar
12.
Molecules ; 23(4)2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29614759

RESUMEN

High glucose-induced cardiomyocyte death is a common symptom in advanced-stage diabetic patients, while its metabolic mechanism is still poorly understood. The aim of this study was to explore metabolic changes in high glucose-induced cardiomyocytes and the heart of streptozotocin-induced diabetic rats by ¹H-NMR-based metabolomics. We found that high glucose can promote cardiomyocyte death both in vitro and in vivo studies. Metabolomic results show that several metabolites exhibited inconsistent variations in vitro and in vivo. However, we also identified a series of common metabolic changes, including increases in branched-chain amino acids (BCAAs: leucine, isoleucine and valine) as well as decreases in aspartate and creatine under high glucose condition. Moreover, a reduced energy metabolism could also be a common metabolic characteristic, as indicated by decreases in ATP in vitro as well as AMP, fumarate and succinate in vivo. Therefore, this study reveals that a decrease in energy metabolism and an increase in BCAAs metabolism could be implicated in high glucose-induced cardiomyocyte death.


Asunto(s)
Aminoácidos de Cadena Ramificada/metabolismo , Metabolismo Energético/efectos de los fármacos , Glucosa/farmacología , Animales , Muerte Celular/efectos de los fármacos , Humanos , Isoleucina/metabolismo , Leucina/metabolismo , Metabolómica/métodos , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Valina/metabolismo
13.
Biochim Biophys Acta Mol Basis Dis ; 1863(1): 266-273, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27816519

RESUMEN

Type 2 diabetes has been associated with cognitive decline, but its metabolic mechanism remains unclear. In the present study, we attempted to investigate brain region-specific metabolic changes in db/db mice with cognitive decline and explore the potential metabolic mechanism linking type 2 diabetes and cognitive decline. We analyzed the metabolic changes in seven brain regions of two types of mice (wild-type mice and db/db mice with cognitive decline) using a 1H NMR-based metabolomic approach. Then, a mixed-model analysis was used to evaluate the effects of mice type, brain region, and their interaction on metabolic changes. Compared with the wild-type mice, the db/db mice with cognitive decline had significant increases in lactate, glutamine (Gln) and taurine as well as significant decreases in alanine, aspartate, choline, succinate, γ-Aminobutyric acid (GABA), glutamate (Glu), glycine, N-acetylaspartate, inosine monophosphate, adenosine monophosphate, adenosine diphosphate, and nicotinamide adenine dinucleotide. Brain region-specific metabolic differences were also observed between these two mouse types. In addition, we found significant interaction effects of mice type and brain region on creatine/phosphocreatine, lactate, aspartate, GABA, N-acetylaspartate and taurine. Based on metabolic pathway analysis, the present study suggests that cognitive decline in db/db mice might be linked to a series of brain region-specific metabolic changes, involving an increase in anaerobic glycolysis, a decrease in tricarboxylic acid (TCA) and Gln-Glu/GABA cycles as well as a disturbance in lactate-alanine shuttle and membrane metabolism.


Asunto(s)
Encefalopatías Metabólicas/complicaciones , Encéfalo/metabolismo , Disfunción Cognitiva/complicaciones , Diabetes Mellitus Tipo 2/complicaciones , Metaboloma , Animales , Encéfalo/patología , Encefalopatías Metabólicas/metabolismo , Encefalopatías Metabólicas/patología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Masculino , Metabolómica , Ratones Endogámicos C57BL
14.
Metab Brain Dis ; 32(2): 585-593, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28070703

RESUMEN

Diabetes mellitus (DM) can result in cognitive dysfunction, but its potential metabolic mechanisms remain unclear. In the present study, we analyzed the metabolite profiling in eight different brain regions of the normal rats and the streptozotocin (STZ)-induced diabetic rats accompanied by cognitive dysfunction using a 1H NMR-based metabolomic approach. A mixed linear model analysis was performed to assess the effects of DM, brain region and their interaction on metabolic changes. We found that different brain regions in rats displayed significant metabolic differences. In addition, the hippocampus was more susceptible to DM compared with other brain regions in rats. More interestingly, significant interaction effects of DM and brain region were observed on alanine, creatine/creatine-phosphate, lactate, succinate, aspartate, glutamate, glutamine, γ-aminobutyric acid, glycine, choline, N-acetylaspartate, myo-inositol and taurine. Based on metabolic pathway analysis, we speculate that cognitive dysfunction in the STZ-induced diabetic rats may be associated with brain region-specific metabolic alterations involving energy metabolism, neurotransmitters, membrane metabolism and osmoregulation.


Asunto(s)
Química Encefálica/efectos de los fármacos , Trastornos del Conocimiento/metabolismo , Trastornos del Conocimiento/psicología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/psicología , Espectroscopía de Resonancia Magnética/métodos , Metabolómica/métodos , Animales , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Redes y Vías Metabólicas , Ratas Sprague-Dawley
15.
Int J Biol Macromol ; 271(Pt 2): 132667, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38801850

RESUMEN

Fibroblast growth factor 21 (FGF21) is one endogenous metabolic molecule that functions as a regulator in glucose and lipid homeostasis. However, the effect of FGF21 on L-lactate homeostasis and its mechanism remains unclear until now. Forty-five Six-week-old male C57BL/6 mice were divided into three groups: control, L-lactate, and FGF21 (1.5 mg/kg) groups. At the end of the treatment, nuclear magnetic resonance-based metabolomics, and key proteins related to L-lactate homeostasis were determined respectively to evaluate the efficacy of FGF21 and its mechanisms. The results showed that, compared to the vehicle group, the L-lactate-treated mice displayed learning and memory performance impairments, as well as reduced hippocampal ATP and NADH levels, but increased oxidative stress, mitochondrial dysfunction, and apoptosis, which suggesting inhibited L-lactate-pyruvate conversion in the brain. Conversely, FGF21 treatment ameliorated the L-lactate accumulation state, accompanied by restoration of the learning and memory defects, indicating enhanced L-lactate uptake and utilization in hippocampal neurons. We demonstrated that maintaining constant L-lactate-pyruvate flux is essential for preserving neuronal bioenergetic and redox levels. FGF21 contributed to preparing the brain for situations of high availability of L-lactate, thus preventing neuronal vulnerability in metabolic reprogramming.


Asunto(s)
Factores de Crecimiento de Fibroblastos , Hipocampo , Homeostasis , Ácido Láctico , Memoria , Ratones Endogámicos C57BL , Animales , Factores de Crecimiento de Fibroblastos/metabolismo , Hipocampo/metabolismo , Hipocampo/efectos de los fármacos , Ratones , Memoria/efectos de los fármacos , Ácido Láctico/metabolismo , Masculino , Homeostasis/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Apoptosis/efectos de los fármacos
16.
CNS Neurosci Ther ; 29(12): 3815-3828, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37334756

RESUMEN

AIMS: The effects of FGF21 on Parkinson's disease (PD) and its relationship with gut microbiota have not been elucidated. This study aimed to investigate whether FGF21 would attenuate behavioral impairment through microbiota-gut-brain metabolic axis in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced PD mice model. METHODS: Male C57BL/6 mice were rendomized into 3 groups: vehicle (CON); MPTP 30 mg/kg/day i.p. injection (MPTP); FGF21 1.5 mg/kg/d i.p. injection plus MPTP 30 mg/kg/day i.p. injection (FGF21 + MPTP). The behavioral features, metabolimics profiling, and 16 s rRNA sequencing were performed after FGF21 treatment for 7 days. RESULTS: MPTP-induced PD mice showed motor and cognitive deficits accompanied by gut microbiota dysbiosis and brain-region-specific metabolic abnormalities. FGF21 treatment dramatically attenuated motor and cognitive dysfunction in PD mice. FGF21 produced a region-specific alteration in the metabolic profile in the brain in ways indicative of greater ability in neurotransmitter metabolism and choline production. In addition, FGF21 also re-structured the gut microbiota profile and increased the relative abundance of Clostridiales, Ruminococcaceae, and Lachnospiraceae, thereby rescuing the PD-induced metabolic disorders in the colon. CONCLUSION: These findings indicate that FGF21 could affect behavior and brain metabolic homeostasis in ways that promote a favorable colonic microbiota composition and through effects on the microbiota-gut-brain metabolic axis.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad de Parkinson , Masculino , Animales , Ratones , Ratones Endogámicos C57BL , Homeostasis , Modelos Animales de Enfermedad , 1-Metil-4-fenil-1,2,3,6-Tetrahidropiridina/farmacología
17.
Biomed Pharmacother ; 168: 115737, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37862975

RESUMEN

AIMS/HYPOTHESIS: Fibroblast growth factor 21 (FGF21) is a hepatokine with pleiotropic effects on glucose and lipid metabolic homeostasis. Here, we aimed to elucidate the mechanisms underlying the protective effects of FGF21 on L-lactate homeostasis and liver lesions in a type 1 diabetes mellitus (T1DM) mice model. METHODS: Six-week-old male C57BL/6 mice were divided into control, T1DM, and FGF21 groups. We also examined hepatic apoptotic signaling and functional indices in wild-type and hydroxycarboxylic acid receptor 1 (HCA1) knockout mice with T1DM or long-term L-lactate exposure. After preincubation of high glucose- or L-lactate treated hepatic AML12 cells, L-lactate uptake, apoptosis, and monocarboxylic acid transporter 2 (MCT2) expression were investigated. RESULTS: In a mouse model of T1DM, hepatic FGF21 expression was downregulated by approximately 1.5-fold at 13 weeks after the hyperglycemic insult. In vivo administration of exogenous FGF21 (2 mg/kg) to diabetic or L-lactate-infused mice significantly prevented hepatic oxidative stress and apoptosis by activating extracellular signal-regulated kinase (ERK)1/2, p38 mitogen-activated protein kinase (MAPK) and AMP-activated protein kinase (AMPK) pathways. HCA1-KO mice were less susceptible to diabetes- and L-lactate-induced hepatic apoptosis and dysfunction. In addition, inhibition of PI3K-mTOR activity revealed that FGF21 prevented L-lactate-induced Cori cycle alterations and hepatic apoptosis by upregulating MCT2 protein translation. CONCLUSIONS/INTERPRETATION: These results demonstrate that L-lactate homeostasis may be a therapeutic target for T1DM-related hepatic dysfunction. The protective effects of FGF21 on hepatic damage were associated with its ability to ameliorate MCT2-dependent Cori cycle alterations and prevent HCA1-mediated inhibition of ERK1/2, p38 MAPK, and AMPK signaling.


Asunto(s)
Diabetes Mellitus Tipo 1 , Ratones , Masculino , Animales , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Ratones Endogámicos C57BL , Hígado , Factores de Crecimiento de Fibroblastos/farmacología , Factores de Crecimiento de Fibroblastos/metabolismo , Modelos Animales de Enfermedad , Glucosa/metabolismo , Homeostasis , Apoptosis , Ratones Noqueados
18.
J Adv Res ; 43: 233-245, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36585111

RESUMEN

INTRODUCTION: Type 1 diabetes (T1D) causes cognitive decline and has been associated with brain metabolic disorders, but its potential molecular mechanisms remain unclear. OBJECTIVES: The purpose of this study was to explore the molecular mechanisms underlying T1D-induced cognitive impairment using metabolomics and lipidomics. METHODS: We developed an optimized integration approach of metabolomics and lipidomics for brain tissue based on UPLC-Q-TOF-MS and analyzed a comprehensive characterization of metabolite and lipid profiles in the hippocampus and frontal cortex of T1D male mice with cognitive decline (T1DCD) and age-matched control (CONT) mice. RESULTS: The results show that T1DCD mice had brain metabolic disorders in a region-specific manner relative to CONT mice, and the frontal cortex exhibited a higher lipid peroxidation than the hippocampus in T1DCD mice. Based on metabolic changes, we found that microglia was activated under diabetic condition and thereby promoted oxidative stress and neuroinflammation, leading to neuronal injury, and this event was more pronounced in the frontal cortex than the hippocampus. CONCLUSION: Our results suggest that brain region-specific shifts in oxidative stress and neuroinflammation may contribute to diabetic cognitive decline, and the frontal cortex could be the more vulnerable brain region than the hippocampus.


Asunto(s)
Encefalopatías Metabólicas , Disfunción Cognitiva , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Masculino , Ratones , Animales , Lipidómica , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/psicología , Enfermedades Neuroinflamatorias , Encéfalo/metabolismo , Metabolómica/métodos , Estrés Oxidativo , Disfunción Cognitiva/metabolismo , Encefalopatías Metabólicas/metabolismo
19.
J Interv Med ; 5(2): 57-63, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35936663

RESUMEN

Background: To investigate the underlying molecular mechanisms of radiofrequency hyperthermia (RFH)-enhanced direct chemotherapy of pancreatic cancers. Method: Rat ductal PaCa cell line DSL-6A/C1 and orthotopic pancreatic cancers of Lewis rats were divided into four study groups with various treatments: i) phosphate-buffered saline (PBS) as a control; ii) RFH alone; iii) intratumoral chemotherapy alone (gemcitabine); and (iv) combination therapy of gemcitabine plus intratumoral RFH at 42 â€‹°C for 30 â€‹min. In the in-vitro confirmation experiments, the viability and apoptosis of DSL-6A/C1 cells in each treatment group were evaluated using cell live/dead staining, flow cytometry, and Western blot. In the in vivo validation experiments, related proteins were evaluated by immunohistochemistry (IHC) staining of tumors. Results: Of the in-vitro experiments, the lowest cell viability and more apoptotic cells were shown in the group with combination therapy compared to other treatments. Western blot data showed elevated Bax/Bcl-2, Caspase-3, and HSP70 expressions in DSL cells with combination therapy, compared to other treatments. Of the in vivo experiments, IHC staining detected the significantly increased expressions of HSP70, IL-1ß, TNF-ɑ, Bax, and Caspase-3 in pancreatic cancer tissues of the animal group treated by combination therapy of gemcitabine with RFH. Conclusion: Molecular imaging-guided interventional RFH can significantly enhance the chemotherapeutic effect on pancreatic cancers via potential molecular mechanisms of up-regulating Bax/caspase-3-dependent apoptosis pathways.

20.
J Ethnopharmacol ; 295: 115379, 2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-35595221

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Xiao-Xu-Ming Decoction (XXMD) is a classical Chinese medicinal compound for the treatment of ischemic stroke, which has good efficacy in clinical studies and also plays a neuroprotective role in pharmacological studies. AIM OF THE STUDY: The purpose of this study is to investigate the potential and integral interventional effects of XXMD on cerebral ischemia/reperfusion rat model. MATERIALS AND METHODS: In this study, 1H NMR metabolomics was used, combined with neurological functional assessments, cerebral infarct area measurements, and pathological staining including Nissl staining, immunofluorescence staining of NeuN and TUNEL, and immunohistochemical staining of MCT2, to analyze the metabolic effects of XXMD in the treatment of an ischemia/reperfusion rat model. RESULTS: It's observed that XXMD treatment could improve the neurological deficit scores and reduce the cerebral infarct areas on cerebral ischemia/reperfusion rat model. The pathological staining results performed that XXMD treatment could improve the decrease of Nissl bodies and the expression of NeuN and MCT2, reduce the high expression of TUNEL. In 1H NMR study, it revealed that the metabolic patterns among three experimental groups were different, the level of lactate, acetate, NAA, glutamate, and GABA were improved to varying degrees in different brain area. CONCLUSION: Our findings indicated that XXMD has positive effect on neuroprotection and improvement of metabolism targeting cerebral ischemic injury in rats, which showed great potential for ischemic stroke.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Fármacos Neuroprotectores , Daño por Reperfusión , Animales , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Infarto Cerebral/tratamiento farmacológico , Medicamentos Herbarios Chinos , Isquemia/tratamiento farmacológico , Metabolómica , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Ratas , Reperfusión , Daño por Reperfusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA