Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Int J Biol Macromol ; 271(Pt 2): 132667, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38801850

RESUMO

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.


Assuntos
Fatores de Crescimento de Fibroblastos , Hipocampo , Homeostase , Ácido Láctico , Memória , Camundongos Endogâmicos C57BL , Animais , Fatores de Crescimento de Fibroblastos/metabolismo , Hipocampo/metabolismo , Hipocampo/efeitos dos fármacos , Camundongos , Memória/efeitos dos fármacos , Ácido Láctico/metabolismo , Masculino , Homeostase/efeitos dos fármacos , Aprendizagem/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Apoptose/efeitos dos fármacos
2.
Cereb Cortex ; 34(2)2024 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-38220573

RESUMO

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.


Assuntos
Disfunção Cognitiva , Diabetes Mellitus , Fatores de Crescimento de Fibroblastos , Camundongos , Animais , Glutamina/metabolismo , Glicogênio Sintase Quinase 3 beta , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases , Ácido Glutâmico/metabolismo , Glucose/metabolismo , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/etiologia , Neurotransmissores
3.
Biomed Pharmacother ; 168: 115737, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37862975

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 1 , Camundongos , Masculino , Animais , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Camundongos Endogâmicos C57BL , Fígado , Fatores de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/metabolismo , Modelos Animais de Doenças , Glucose/metabolismo , Homeostase , Apoptose , Camundongos Knockout
4.
FASEB J ; 37(9): e23134, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37561545

RESUMO

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.


Assuntos
Insuficiência Cardíaca , Proteínas Quinases , Animais , Camundongos , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Insuficiência Cardíaca/genética , Insuficiência Cardíaca/patologia , Homeostase , Camundongos Knockout , Proteômica , Taurina , Piruvato Desidrogenase Quinase de Transferência de Acetil/genética
5.
CNS Neurosci Ther ; 29(12): 3815-3828, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37334756

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Doença de Parkinson , Masculino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Homeostase , Modelos Animais de Doenças , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia
6.
J Adv Res ; 43: 233-245, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36585111

RESUMO

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.


Assuntos
Encefalopatias Metabólicas , Disfunção Cognitiva , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Masculino , Camundongos , Animais , Lipidômica , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/psicologia , Doenças Neuroinflamatórias , Encéfalo/metabolismo , Metabolômica/métodos , Estresse Oxidativo , Disfunção Cognitiva/metabolismo , Encefalopatias Metabólicas/metabolismo
7.
J Interv Med ; 5(2): 57-63, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35936663

RESUMO

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.

8.
Comput Struct Biotechnol J ; 20: 3935-3945, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35950184

RESUMO

This study aimed to explore whether chronic l-lactate exposure could affect the peripheral tissues of mice and to determine the underlying pathogenesis. Herein, male C57BL/6 mice were divided into control and l-lactate groups. After l-lactate treatment for eight weeks (1 g/kg), metabolic changes in liver, kidney, muscle, and serum samples were determined by 1H nuclear magnetic resonance (1H NMR)-based metabolomics. Additionally, organ function was evaluated by serum biochemical and histopathological examinations. Reactive oxygen species (ROS) levels were measured using dihydroethidium staining; levels of signals involved in lactate metabolism and ROS-related pathways were detected using western blotting or polymerase chain reaction. Apoptosis was detected by TUNEL-fluorescence staining. Metabolomic analysis revealed that l-lactate mice showed decreased levels of glutathione (GSH), taurine, ATP, and increased glucose content, compared to control mice. Furthermore, l-lactate mice presented significantly higher serum levels of alanine aminotransferase and aspartate aminotransferase and increased glycogen content in hepatic tissues, compared to control mice. l-lactate mice also had a greater number of apoptotic nuclei in the livers than controls. Moreover, l-lactate exposure reduced mRNA and protein levels of superoxide dismutase-2 and c-glutamylcysteine ligase, elevated levels of cytochrome P450 2E1 and NADPH oxidase-2, and increased the protein expressions of LDHB, Bax/Bcl-2, cleaved caspase-3, and sirtuin-1 in hepatic tissues. Together, these results indicate that chronic l-lactate exposure increases oxidative stress and apoptosis in hepatocytes via upregulation of Bax/Bcl-2 expression and the consequent mitochondrial cytochrome-C release and caspase-3 activation, which contributes to the pathogenesis of hepatic dysfunction.

9.
Mol Neurobiol ; 59(9): 5656-5672, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35761156

RESUMO

Fibroblast growth factor 21 (FGF21) is an endocrine hormone that exerts beneficial effects on glucose and lipid metabolic homeostasis. However, the impact of FGF21 on type 1 diabetes-associated cognitive decline (DACD) and its mechanisms of action remain unclear. In this study, we aimed to evaluate the effects of FGF21 on lactate uptake and usage in a mouse model of streptozotocin-induced DACD. Six-week-old male C57BL/6 mice were divided into the control, diabetic, and FGF21 (which received 2 mg/kg recombinant human FGF21) groups. At the end of the treatment period, learning and memory performance, nuclear magnetic resonance-based metabonomics, and expressions of various hippocampal protein were analyzed to determine the efficacy of FGF21. The results showed that compared to the control mice, the diabetic mice had reduced long-term memory performance after the hyperglycemic insult; decreased hippocampal levels of lactate dehydrogenase-B (LDH-B) activity, bioenergy metabolites, and monocarboxylate transporter 2 (MCT2); and increased lactate levels. Impaired phosphoinositide 3-kinase (PI3K) signaling was also observed in the diabetic mice. However, FGF21 treatment improved LDH-B activity, ß-nicotinamide adenine dinucleotide, and ATP levels, and increased MCT2 expression and PI3K signaling pathway, which in turn improved the learning and memory defects. These findings demonstrated that the effects of FGF21 on DACD were associated with its ability to improve LDH-B-mediated lactate usage and MCT2-dependent lactate uptake. Further, these beneficial effects of FGF21 in the hippocampus were mediated by the PI3K signaling pathways.


Assuntos
Disfunção Cognitiva , Diabetes Mellitus Experimental , Animais , Disfunção Cognitiva/complicações , Disfunção Cognitiva/tratamento farmacológico , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/uso terapêutico , Humanos , Ácido Láctico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo
10.
J Ethnopharmacol ; 295: 115379, 2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-35595221

RESUMO

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.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Animais , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Infarto Cerebral/tratamento farmacológico , Medicamentos de Ervas Chinesas , Isquemia/tratamento farmacológico , Metabolômica , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Ratos , Reperfusão , Traumatismo por Reperfusão/metabolismo
11.
Mol Nutr Food Res ; 66(10): e2100772, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35225418

RESUMO

SCOPE: Obesity is becoming a major public health problem due to excess dietary fat intake. Dendrobium officinale (D. officinale) is a medicine food homology plant and exerts multiple health-promoting effects. However, its antiobesity effects and the potential mechanisms remain unclear. METHODS AND RESULTS: High-fat diet (HFD)-fed mice are administered D. officinale dietary fiber (DODF) daily by gavage for 11 weeks. The results show that treatment with DODF alleviates obesity, liver steatosis, inflammation, and oxidant stress in HFD-induced obese mice. Improved glucose homeostasis in obese mice after DODF treatment is achieved by enhancing insulin pathway and hepatic glycogen synthesis. DODF restructures the gut microbiota in obese mice by decreasing the relative abundance of Bilophila and increasing the relative abundances of Akkermansia, Bifidobacterium, and Muribaculum. Also, DODF reshapes the metabolic phenotype of obese mice as indicated by up-regulating energy metabolism, increasing acetate and taurine, and reducing serum low density/very low density lipoproteins (LDL/VLDL). These beneficial effects are partly transferred by FMT, implying the gut microbiota as a target for the protective effect of DODF on obesity-related symptoms. CONCLUSION: The results suggest that DODF can be used as a novel prebiotics to maintain the gut microbial homeostasis and improve metabolic health, preventing obesity and related metabolic syndrome.


Assuntos
Dendrobium , Microbioma Gastrointestinal , Animais , Dieta Hiperlipídica/efeitos adversos , Fibras na Dieta/farmacologia , Glucose/farmacologia , Homeostase , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/prevenção & controle
12.
Int J Cancer ; 150(9): 1525-1536, 2022 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-34985768

RESUMO

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.


Assuntos
Colina , Neoplasias da Próstata , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Linhagem Celular Tumoral , Proliferação de Células , Colina/metabolismo , Humanos , Masculino , Próstata/patologia , Neoplasias da Próstata/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
13.
J Proteome Res ; 20(11): 5024-5035, 2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34699241

RESUMO

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.


Assuntos
Fator 2 de Crescimento de Fibroblastos , Qualidade de Vida , Animais , Fator 2 de Crescimento de Fibroblastos/genética , Intestinos , Metabolômica , Camundongos , Espectroscopia de Prótons por Ressonância Magnética
14.
Biochim Biophys Acta Mol Basis Dis ; 1867(12): 166266, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34481869

RESUMO

Sexual dimorphism exists in the onset and development of type 1 diabetes (T1D), but its potential pathological mechanism is poorly understood. In the present study, we examined sex-specific changes in the gut microbiome and host metabolome of T1D mice via 16S rRNA gene sequencing and nuclear magnetic resonance (NMR)-based metabolomics approach, and aimed to investigate potential mechanism of the gut microbiota-host metabolic interaction in the sexual dimorphism of T1D. Our results demonstrate that female mice had a greater shift in the gut microbiota than male mice during the development of T1D; however, host metabolome was more susceptible to T1D in male mice. The correlation network analysis indicates that T1D-induced host metabolic changes may be regulated by the gut microbiota in a sex-specific manner, mainly involving short-chain fatty acids (SCFAs) metabolism, energy metabolism, amino acid metabolism, and choline metabolism. Therefore, our study suggests that sex-dependent "gut microbiota-host metabolism axis" may be implicated in the sexual dimorphism of T1D, and the link between microbes and metabolites might contribute to the prevention and treatment of T1D.


Assuntos
Diabetes Mellitus Tipo 1/genética , Ácidos Graxos Voláteis/genética , Microbioma Gastrointestinal/genética , Metaboloma/genética , Animais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Ácidos Graxos Voláteis/metabolismo , Feminino , Humanos , Espectroscopia de Ressonância Magnética , Masculino , Metabolômica , Camundongos , Camundongos Endogâmicos NOD/genética , Camundongos Endogâmicos NOD/metabolismo , Caracteres Sexuais
15.
J Proteome Res ; 20(8): 3900-3912, 2021 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-34237942

RESUMO

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.


Assuntos
Dieta Hiperlipídica , Fatores de Crescimento de Fibroblastos , Animais , Dieta Hiperlipídica/efeitos adversos , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fígado , Masculino , Metabolômica , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Transdução de Sinais
16.
Neurochem Int ; 143: 104941, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33333211

RESUMO

Type 1 diabetes (T1D) has been reported to cause cognitive decline, but brain metabolic changes during this process are still far from being fully understood. Here, we found that streptozotocin (STZ)-induced T1D mice exhibited impaired learning and memory at 11 weeks after STZ treatment but not at 3 weeks. Therefore, we studied metabolic alterations in six different brain regions of T1D mice with and without cognitive decline, and attempted to identify key metabolic pathways related to diabetic cognitive dysfunction. The results demonstrate that lactate had already increased in all brain regions of T1D mice prior to cognitive decline, but a decreased TCA cycle was only observed in hippocampus, cortex and striatum of T1D mice with cognitive impairment. Reduced N-acetylaspartate and choline were found in all brain regions of T1D mice, irrespective of cognitive decline. In addition, disrupted neurotransmitter metabolism was noted to occur in T1D mice before cognitive deficit. Of note, we found that the level of uridine was significantly reduced in cerebellum, cortex, hypothalamus and midbrain of T1D mice when cognitive decline was presented. Therefore, brain region-specific metabolic alterations may comprise possible biomarkers for the early-diagnosis and monitoring of diabetic cognitive decline. Moreover, down-regulated TCA cycle and pyrimidine metabolism could be closely related to T1D-associated cognitive impairment.


Assuntos
Encéfalo/metabolismo , Disfunção Cognitiva/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/metabolismo , Animais , Disfunção Cognitiva/induzido quimicamente , Disfunção Cognitiva/psicologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/psicologia , Diabetes Mellitus Tipo 1/induzido quimicamente , Diabetes Mellitus Tipo 1/psicologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Estreptozocina
17.
Front Aging Neurosci ; 13: 778527, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35002679

RESUMO

Microglial polarization and the subsequent neuroinflammatory response were identified as key contributors to the progress of Parkinson's disease (PD). Researchers have shown that fibroblast growth factor 21 (FGF21) plays multiple biological functions, including anti-inflammation and neuroprotection. However, the knowledge of FGF21 on microglial polarization in PD in vivo is far from completion. In this study, both in vivo and in vitro models were used to investigate whether FGF21 enhances the brain function by modulating microglial polarization in PD. The protective effects of FGF21 in vivo were conducted using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced PD mice model alongside intraperitoneally received FGF21. A behavioral test battery and tyrosine hydroxylase (TH) immunohistochemistry were conducted to evaluate the neuronal function and nigrostriatal tract integrity. Immunofluorescence assay and Western blot were used to examine M1/M2 microglial polarization. Then, a microglia-neuron co-culture system was adopted in vitro to identify the underlying molecular mechanisms of FGF21. The results showed that FGF21 significantly alleviated motor and cognitive impairment in mice with PD. FGF21 also protected TH-positive neuron cells in the striatum and midbrain. Mechanistically, FGF21 suppressed M1 microglial polarization and the subsequent mRNA expression of pro-inflammatory factors while promoting M2 microglial polarization with increasing anti-inflammatory factors in mice with PD. Furthermore, sirtuin 1 (SIRT1) and the nuclear factor-kappa B (NF-κB) pathway were involved in the FGF21-induced M2 microglial polarization. Conversely, SIRT1 inhibitor EX527 significantly prevented both the FGF21-induced SIRT1 expression and M2 microglial polarization. Moreover, FGF21 pretreatment of microglia significantly prevented neuronal cell apoptosis in a microglia-neuron co-culture system. In conclusion, our data demonstrate that FGF21 exerted its protective effects in the pathology of PD through SIRT1/NF-κB pathway-mediated microglial polarization. Given the safety record of human clinical trials, FGF21 could be a promising therapy for clinical trials to ameliorate motor and nonmotor deficits in patients with PD.

18.
Comput Struct Biotechnol J ; 18: 2063-2074, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32802278

RESUMO

Type 1 diabetes (T1D) can cause brain region-specific metabolic disorders, but whether gender influences T1D-related brain metabolic changes is rarely reported. Therefore, here we examined metabolic changes in six different brain regions of male and female mice under normal and T1D conditions using an integrated method of NMR-based metabolomics and linear mixed-model, and aimed to explore sex-specific metabolic changes from normal to T1D. The results demonstrate that metabolic differences occurred in all brain regions between two genders, while the hippocampal metabolism is more likely to be affected by T1D. At the 4th week after streptozotocin treatment, brain metabolic disorders mainly occurred in the cortex and hippocampus in female T1D mice, but the striatum and hippocampus in male T1D mice. In addition, anaerobic glycolysis was significantly altered in male mice, mainly in the striatum, midbrain, hypothalamus and hippocampus, but not in female mice. We also found that female mice exhibited a hypometabolism status relative to male mice from normal to T1D. Collectively, this study suggests that T1D affected brain region-specific metabolic alterations in a sex-specific manner, and may provide a metabolic view on diabetic brain diseases between genders.

19.
FASEB J ; 34(10): 13333-13344, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32816365

RESUMO

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.


Assuntos
Proliferação de Células , Fatores Quimiotáticos/metabolismo , Neoplasias Colorretais/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Glicólise , Proteínas S100/metabolismo , Animais , Transportador de Glucose Tipo 1/genética , Células HCT116 , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
20.
J Proteome Res ; 19(8): 3011-3021, 2020 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-32450697

RESUMO

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.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Animais , Feminino , Fígado , Espectroscopia de Ressonância Magnética , Masculino , Metabolômica , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...