Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Nat Metab ; 5(3): 445-465, 2023 03.
Article in English | MEDLINE | ID: mdl-36959514

ABSTRACT

Astrocytes provide key neuronal support, and their phenotypic transformation is implicated in neurodegenerative diseases. Metabolically, astrocytes possess low mitochondrial oxidative phosphorylation (OxPhos) activity, but its pathophysiological role in neurodegeneration remains unclear. Here, we show that the brain critically depends on astrocytic OxPhos to degrade fatty acids (FAs) and maintain lipid homeostasis. Aberrant astrocytic OxPhos induces lipid droplet (LD) accumulation followed by neurodegeneration that recapitulates key features of Alzheimer's disease (AD), including synaptic loss, neuroinflammation, demyelination and cognitive impairment. Mechanistically, when FA load overwhelms astrocytic OxPhos capacity, elevated acetyl-CoA levels induce astrocyte reactivity by enhancing STAT3 acetylation and activation. Intercellularly, lipid-laden reactive astrocytes stimulate neuronal FA oxidation and oxidative stress, activate microglia through IL-3 signalling, and inhibit the biosynthesis of FAs and phospholipids required for myelin replenishment. Along with LD accumulation and impaired FA degradation manifested in an AD mouse model, we reveal a lipid-centric, AD-resembling mechanism by which astrocytic mitochondrial dysfunction progressively induces neuroinflammation and neurodegeneration.


Subject(s)
Alzheimer Disease , Neuroinflammatory Diseases , Mice , Animals , Astrocytes/metabolism , Alzheimer Disease/metabolism , Fatty Acids/metabolism , Mitochondria/metabolism
3.
Neurosci Biobehav Rev ; 135: 104556, 2022 04.
Article in English | MEDLINE | ID: mdl-35122783

ABSTRACT

Alzheimer's disease (AD) is one of the fastest growing cognitive decline-related neurological diseases. To date, effective curative strategies have remained elusive. A growing body of evidence indicates that dietary patterns have significant effects on cognitive function and the risk of developing AD. Previous studies on the association between diet and AD risk have mainly focused on individual food components and specific nutrients, and the mechanisms responsible for the beneficial effects of dietary patterns on AD are not well understood. This article provides a comprehensive overview of the effects of dietary patterns, including the Mediterranean diet (MedDiet), dietary approaches to stop hypertension (DASH) diet, Mediterranean-DASH diet intervention for neurological delay (MIND), ketogenic diet, caloric restriction, intermittent fasting, methionine restriction, and low-protein and high-carbohydrate diet, on cognitive impairment and summarizes the underlying mechanisms by which dietary patterns attenuate cognitive impairment, especially highlighting the modulation of dietary patterns on cognitive impairment through gut microbiota. Furthermore, considering the variability in individual metabolic responses to dietary intake, we put forward a framework to develop personalized dietary patterns for people with cognitive disorders or AD based on individual gut microbiome compositions.


Subject(s)
Alzheimer Disease , Cognition Disorders , Cognitive Dysfunction , Diet, Mediterranean , Dietary Approaches To Stop Hypertension , Alzheimer Disease/psychology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Humans
4.
STAR Protoc ; 2(2): 100559, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34095867

ABSTRACT

Mitochondrial dysfunction and metabolic reprogramming are implicated in a variety of neurological disorders. Here, we present a protocol that enables complex profiling of brain metabolic function using acute mouse brain slices ex vivo. Utilizing differential metabolic conditions, substrates, and inhibitors, this protocol can be broadly applied to determine metabolic shift or reprogramming upon genetic manipulations, pathological insults, or therapeutic interventions and could thus further the understanding of the dynamic role of energy metabolism in brain physiological function and diseases. For complete details on the use and execution of this protocol, please refer to Qi et al. (2021).


Subject(s)
Brain/metabolism , Animals , Energy Metabolism , Glycolysis , Mice , Reproducibility of Results
5.
Cell Rep ; 34(1): 108572, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33406436

ABSTRACT

Alzheimer's disease (AD) risk gene ApoE4 perturbs brain lipid homeostasis and energy transduction. However, the cell-type-specific mechanism of ApoE4 in modulating brain lipid metabolism is unclear. Here, we describe a detrimental role of ApoE4 in regulating fatty acid (FA) metabolism across neuron and astrocyte in tandem with their distinctive mitochondrial phenotypes. ApoE4 disrupts neuronal function by decreasing FA sequestering in lipid droplets (LDs). FAs in neuronal LDs are exported and internalized by astrocytes, with ApoE4 diminishing the transport efficiency. Further, ApoE4 lowers FA oxidation and leads to lipid accumulation in both astrocyte and the hippocampus. Importantly, diminished capacity of ApoE4 astrocytes in eliminating neuronal lipids and degrading FAs accounts for their compromised metabolic and synaptic support to neurons. Collectively, our findings reveal a mechanism of ApoE4 disruption to brain FA and bioenergetic homeostasis that could underlie the accelerated lipid dysregulation and energy deficits and increased AD risk for ApoE4 carriers.


Subject(s)
Alzheimer Disease/metabolism , Apolipoprotein E4/physiology , Astrocytes/metabolism , Fatty Acids/metabolism , Lipid Metabolism , Neurons/metabolism , Animals , Brain/metabolism , Energy Metabolism , Gene Knock-In Techniques , Homeostasis , Humans , Lipid Droplets/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Mutation , Oxidation-Reduction
6.
Antioxid Redox Signal ; 34(8): 611-630, 2021 03 10.
Article in English | MEDLINE | ID: mdl-32143551

ABSTRACT

Significance: Alzheimer's disease (AD) is the leading cause of dementia. Thus far, 99.6% of clinical trials, including those targeting energy metabolism, have failed to exert disease-modifying efficacy. Altered mitochondrial function and disruption to the brain bioenergetic system have long-been documented as early events during the pathological progression of AD. Recent Advances: While therapeutic approaches that directly promote mitochondrial bioenergetic machinery or eliminate reactive oxygen species have exhibited limited translatability, emerging strategies targeting nonenergetic aspects of mitochondria provide novel therapeutic targets with the potential to modify AD risk and progression. Growing evidence also reveals a critical link between mitochondrial phenotype and neuroinflammation via metabolic reprogramming of glial cells. Critical Issues: Herein, we summarize major classes of mitochondrion-centered AD therapeutic strategies. In addition, the discrepancy in their efficacy when translated from preclinical models to clinical trials is addressed. Key factors that differentiate the responsiveness to bioenergetic interventions, including sex, apolipoprotein E genotype, and cellular diversity in the brain, are discussed. Future Directions: We propose that the future development of mitochondria-targeted AD therapeutics should consider the interactions between bioenergetics and other disease mechanisms, which may require cell-type-specific targeting to distinguish neurons and non-neuronal cells. Moreover, a successful strategy will likely include stratification by metabolic phenotype, which varies by sex and genetic risk profile and dynamically changes throughout the course of disease. As the network of mitochondrial integration expands across intracellular and systems level biology, assessment of intended, the good, versus unintended consequences, the bad, will be required to reach the potential of mitochondrial therapeutics.


Subject(s)
Alzheimer Disease/drug therapy , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Alzheimer Disease/metabolism , Animals , Humans , Mitochondria/metabolism
7.
J Agric Food Chem ; 67(18): 5122-5134, 2019 May 08.
Article in English | MEDLINE | ID: mdl-30995031

ABSTRACT

Neuroinflammation has been intensively demonstrated to be related to various neurodegenerative diseases including Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Alzheimer's disease (AD). A natural polymethoxylated flavone, nobiletin (NOB) has been reported to alleviate oxidative stress, insulin resistance, and obesity. In this study, we evaluated the protection effects of NOB on neuroinflammation and memory deficit. Three-month mice were administrated with NOB by oral gavage every day for 6 weeks (100 mg/kg/day); subsequently mice were injected intraperitoneally with lipopolysaccharide (LPS) for 7 days. Results of behavioral tests revealed that NOB dramatically ameliorated LPS-triggered memory deficit regarding synaptic dysfunctions and neuronal loss. Also, NOB suppressed the microglial activation and proinflammatory cytokine secretion, such as COX-2, IL-1ß, TNF-α, and iNOS. Similarly, upon LPS stimulation, pretreatment NOB diminished the secretion of the proinflammatory cytokines in BV-2 microglia cells by exposure to LPS via modulating MAPKs, PI3K/AKT, and NF-κB signaling pathways. In addition, NOB alleviated LPS-amplified redox imbalance, disturbance of mitochondrial membrane potential (MMP), and dampening of the expression of protein related to mitochondrial respiration. The present study provides compelling evidence that NOB decreased LPS-stimulated neuroinflammation and memory impairment through maintaining cellular oxidative balance and blocking the NF-κB transcriptional pathway, illustrating that the nutritional compound NOB may serve as a potential approach to alleviate neuroinflammation-related diseases.


Subject(s)
Flavones/administration & dosage , Inflammation/complications , Memory Disorders/prevention & control , Mitogen-Activated Protein Kinase Kinases/immunology , NF-kappa B/immunology , Animals , Brain/drug effects , Brain/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Humans , Inflammation/genetics , Inflammation/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Memory Disorders/etiology , Memory Disorders/genetics , Memory Disorders/immunology , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Mitogen-Activated Protein Kinase Kinases/genetics , NF-kappa B/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
8.
Front Physiol ; 10: 1531, 2019.
Article in English | MEDLINE | ID: mdl-31969828

ABSTRACT

As an organ with a highly heterogenous cellular composition, the brain has a bioenergetic system that is more complex than peripheral tissues. Such complexities are not only due to the diverse bioenergetic phenotypes of a variety of cell types that differentially contribute to the metabolic profile of the brain, but also originate from the bidirectional metabolic communications and coupling across cell types. While brain energy metabolism and mitochondrial function have been extensively investigated in aging and age-associated neurodegenerative disorders, the role of various cell types and their inter-cellular communications in regulating brain metabolic and synaptic functions remains elusive. In this review, we summarize recent advances in differentiating bioenergetic phenotypes of neurons, astrocytes, and microglia in the context of their functional specificity, and their metabolic shifts upon aging and pathological conditions. Moreover, the metabolic coordination between the two most abundant cell populations in brain, neurons and astrocytes, is discussed regarding how they jointly establish a dynamic and responsive system to maintain brain bioenergetic homeostasis and to combat against threats such as oxidative stress, lipid toxicity, and neuroinflammation. Elucidating the mechanisms by which brain cells with distinctive bioenergetic phenotypes individually and collectively shape the bioenergetic system of the brain will provide rationale for spatiotemporally precise interventions to sustain a metabolic equilibrium that is resilient against synaptic dysfunction in aging and neurodegeneration.

9.
Food Chem Toxicol ; 122: 181-193, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30316844

ABSTRACT

Circadian rhythms are intimately linked to cellular redox status homeostasis via the regulation of mitochondrial function. Tea polyphenols (TP) are nutraceuticals that possess powerful antioxidant properties, especially ameliorating oxidative stress. The objective of this study was to investigate whether circadian clock is involved in the protection effect of TP on oxidative stress cell models. TP ameliorate H2O2-triggered relatively shallow daily oscillations and phase shift of circadian clock genes transcription and protein expression. Meanwhile, TP attenuate H2O2-stimulated excessive secretions of reactive oxygen species (ROS) and restore the depletions of mitochondrial function in a Bmal1-dependent manner. Furthermore, TP treatment accelerates nuclear translocation of Nrf2 and modulates the downstream expressions of antioxidant enzymes. Intriguingly, knockdown of Bmal1 notably blocked Nrf2/ARE/HO-1 redox-sensitive transcription pathway. Our study revealed that TP, as a Bmal1-enhancing natural compound, alleviated redox imbalance via strengthening Keap1/Nrf2 antioxidant defense pathway and ameliorating mitochondrial dysfunction in a Bmal1-dependent manner.


Subject(s)
ARNTL Transcription Factors/drug effects , Circadian Clocks , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Polyphenols/pharmacology , Tea/chemistry , ARNTL Transcription Factors/metabolism , Animals , Apoptosis/drug effects , CLOCK Proteins/genetics , Circadian Rhythm , Heme Oxygenase-1/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Hydrogen Peroxide/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , Mice , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Oxidative Stress/drug effects , Signal Transduction/drug effects
10.
Food Funct ; 9(6): 3374-3386, 2018 Jun 20.
Article in English | MEDLINE | ID: mdl-29868672

ABSTRACT

The global rise in obesity and type 2 diabetes has precipitated the need for therapeutic intervention in the arsenal against adiposity. (-)-Epigallocatechin-3-gallate (EGCG), a major nutraceutical component of green tea, has been regarded as a nutraceutical that has powerful antioxidant and anti-obesity bioactivities. In the present study, we showed that EGCG alleviates intracellular lipid accumulation markedly, and the inhibitory effect was largely limited to the early stage of adipocyte differentiation. Consistently, EGCG notably evoked the phosphorylation of AMPK and ACC and blunted the key enzymes of de novo lipogenesis. Interestingly, EGCG elicited iWAT-preadipocyte-derived mature white adipocyte beiging via activating thermogenic gene Ucp1 expression and mitochondrial biogenesis. Furthermore, our results also revealed that EGCG attenuated insulin signaling pathway blockage induced by TNF-α through the abrogation of redox imbalance and mitochondrial dysfunction. These findings indicate that EGCG is capable of suppressing adipogenesis and evoking white adipocyte beiging and therefore it may potentially serve as a novel approach to combat obesity.


Subject(s)
Adipocytes, Beige/drug effects , Adipogenesis/drug effects , Catechin/analogs & derivatives , Insulin Resistance , Obesity/physiopathology , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes, Beige/cytology , Adipocytes, Beige/metabolism , Animals , Catechin/pharmacology , Mice , Obesity/drug therapy , Obesity/genetics , Obesity/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Tumor Necrosis Factor-alpha/genetics , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(6): 549-562, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29501626

ABSTRACT

SCOPE: Circadian clock plays a principal role in orchestrating our daily physiology and metabolism, and their perturbation can evoke metabolic diseases such as fatty liver and insulin resistance. Nobiletin (NOB) has been demonstrated to possess antitumor and neuroprotective activities. The objective of the current study is to determine potential effects of NOB on modulating the core clock gene Bmal1 regarding ameliorating glucolipid metabolic disorders. RESULTS: Our results revealed that NOB partially reverse the relatively shallow daily oscillations of circadian clock genes and reset phase-shifting circadian rhythms in primary hepatocytes under metabolic disorders conditions. Importantly, NOB was found to be effective at amplifying glucose uptake via stimulating IRS-1/AKT signaling pathway, as well as blunting palmitate-induced lipogenesis in HepG2 cells via modulating AMPK-Sirt1 signaling pathway and key enzymes of de novo lipogenesis in a Bmal1-dependent manner. NOB attenuated palmitate-stimulated excessive secretions of ROS, restored the depletions of mitochondrial membrane potential, which is similar to the recovery in expressions of mitochondrial respiration complex I-IV. CONCLUSION: This study is the first to provide compelling evidences that NOB prevent cellular glucolipid metabolic imbalance and mitochondrial function in a Bmal1-dependent manner. Overall, NOB may serve as a nutritional preventive strategy in recovering metabolic disorders relevant to circadian clock.


Subject(s)
Circadian Clocks/drug effects , Flavones/pharmacology , Hepatocytes/metabolism , Insulin Resistance , Lipid Metabolism/drug effects , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Hep G2 Cells , Hepatocytes/cytology , Humans , Insulin Receptor Substrate Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism
12.
Food Funct ; 9(3): 1510-1523, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29423494

ABSTRACT

As a major nutraceutical component of green tea (-)-epigallocatechin-3-gallate (EGCG) has attracted interest from scientists due to its well-documented antioxidant and antiobesity bioactivities. In the current study, we aimed to investigate the protective effect of EGCG on metabolic misalignment and in balancing the redox status in mice liver and HepG2 cells under insulin resistance condition. Our results indicated that EGCG accelerates the glucose uptake and evokes IRS-1/Akt/GLUT2 signaling pathway via dampening the expression of protein tyrosine phosphatase 1B (PTP1B). Consistently, ectopic expression of PTP1B by Ad-PTP1B substantially impaired EGCG-elicited IRS-1/Akt/GLUT2 signaling pathway. Moreover, EGCG co-treatment stimulated nuclear translocation of Nrf2 by provoking P13K/AKT signaling pathway and thus modulated the downstream expressions of antioxidant enzymes such as HO-1 and NQO-1 in HepG2 cells. Furthermore, knockdown Nrf2 by small interfering RNA (siRNA) notably enhanced the expression of PTP1B and blunt EGCG-stimulated glucose uptake. Consistent with these results, in vivo study revealed that EGCG supplement significantly ameliorated high-fat and high-fructose diet (HFFD)-triggered insulin resistance and oxidative stress by up-regulating the IRS-1/AKT and Keap1/Nrf2 transcriptional pathways. Administration of an appropriate chemopreventive agent, such as EGCG, could potentially serve as an additional therapeutic intervention in the arsenal against obesity.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Catechin/analogs & derivatives , Insulin Resistance , NF-E2-Related Factor 2/metabolism , Obesity/drug therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Animals , Catechin/pharmacology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , Insulin/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Obesity/genetics , Obesity/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction/drug effects
13.
Toxicol Lett ; 284: 103-112, 2018 Mar 01.
Article in English | MEDLINE | ID: mdl-29248572

ABSTRACT

1,3-dichloro-2-propanol (1,3-DCP) is a widely concerned food processing contaminant which has been investigated for decades. While the neurotoxicity of 1,3-DCP and related mechanisms are still elusive. Herein, the effect of 1,3-DCP on neurotoxicity was investigated using BV-2 microglia cells. 1,3-DCP significantly decreased cell viability from 78.6% to 59.2% at doses between 2 and 20 mM. AO/EB and JC-1 staining indicated that 1,3-DCP induced apoptosis by means of the decrease of mitochondrial membrane potential. Meanwhile, western blot showed that 1,3-DCP stimulated inflammation of BV-2 cells through phosphorylation of MAPKs and activation of NF-κB pathways mediated by reactive oxygen species (ROS). Furthermore, the degree of inflammation and apoptosis has eased through MAPKs and NF-κB pathways with cells pretreated by N-acetylcysteine (NAC). Overall, these results presented here suggested that 1,3-DCP has neurotoxic effect on BV-2 microglia mainly via MAPKs and NF-κB pathways mediated by ROS.


Subject(s)
Apoptosis/drug effects , Microglia/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , alpha-Chlorohydrin/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Food Contamination , Membrane Potential, Mitochondrial/drug effects , Mice , Microglia/immunology , Microglia/pathology , Phosphorylation , Signal Transduction , alpha-Chlorohydrin/toxicity
14.
Food Funct ; 8(12): 4421-4432, 2017 Dec 13.
Article in English | MEDLINE | ID: mdl-29090295

ABSTRACT

Many studies have shown that oxidative stress is a major cause of cellular injuries in a variety of human diseases including cognitive impairment. Tea polyphenols (TPs), natural plant flavonoids found in tea plant leaves, possess the bioactivity to affect the pathogenesis of several chronic diseases via antioxidant associated mechanisms. However, the possible antioxidant and neuroprotective properties of TPs in the brain of mice housed in constant darkness and in H2O2-stimulated SH-SY5Y cells are yet to be elucidated. In this study, pretreatment with TPs markedly attenuated H2O2-elicited cell viability loss and mitochondrial dysfunction, suppressed the induced apoptosis and reduced the elevated levels of intracellular ROS and H2O2. Additionally, TPs modulate the nuclear translocation of Nrf2 and the TrkB/CREB/BDNF signaling pathway by provoking the PI3K/AKT pathway and thus, they transcriptionally regulate the downstream expression of antioxidant enzymes including HO-1, NQO-1, and BDNF in SH-SY5Y cells. Furthermore, an in vivo study revealed that housing mice in constant darkness, simulating shift work disruption in humans, notably affects the AKT/CREB/BDNF signal pathway and the nuclear translocation of Nrf2 and its downstream phase II detoxification enzymes in brain tissue. Remarkably, TP supplementation through drinking water eliminated these changes. These results suggest that TPs possess protective effects against oxidative stress-triggered cognitive impairment, which might be a potential nutritional preventive strategy for neurodegenerative diseases implicated with oxidative stress in shift workers.


Subject(s)
Brain/drug effects , CREB-Binding Protein/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/administration & dosage , Plant Extracts/administration & dosage , Polyphenols/administration & dosage , Receptor, trkB/metabolism , Animals , Apoptosis/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , CREB-Binding Protein/genetics , Camellia sinensis/chemistry , Cell Survival/drug effects , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Receptor, trkB/genetics , Signal Transduction/drug effects
15.
Food Chem Toxicol ; 110: 189-199, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29061316

ABSTRACT

Circadian rhythms are autonomous anticipatory oscillators that control a large array of physiological and metabolic processes. Compelling evidence points toward an interplay between circadian rhythms and cellular redox metabolism. Dysregulation of circadian rhythms is associated with neurodegenerative diseases and accelerated aging. Tea polyphenols (TP) is one of the most used antioxidants and exerts beneficial effect on neurodegenerative diseases. The aim of this study is to investigate whether circadian clock mechanisms are involved in the protection effect of TP against neural redox imbalance and mitochondrial dysfunction in SH-SY5Y cells. In the current study, our results revealed that TP, as a Bmal1-enhancing natural compound, can reverse the relatively shallow daily oscillations of circadian clock genes transcription and protein expression in SH-SY5Y neuronal cells under oxidative stress conditions. Furthermore, TP pretreatment significantly ameliorated H2O2-elicited mitochondria impairment via manipulating mitochondrial dynamics and mitochondrial membrane potential, which is consistent with the recovery in expression of mitochondrial respiration complex I-IV in Bmal1-dependent efficiency. Furthermore, Bmal1 is involved in TP-stimulated Nrf2/ARE/HO-1 and AKT/CREB/BDNF signaling pathway. Hence, TP may serve as a nutritional preventive strategy in the recovery of oxidative stress-related neurodegenerative disease via modulating circadian clock.


Subject(s)
ARNTL Transcription Factors/metabolism , Camellia sinensis/chemistry , Circadian Rhythm/drug effects , Neurons/drug effects , Plant Extracts/pharmacology , Polyphenols/pharmacology , ARNTL Transcription Factors/genetics , Cell Line , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Signal Transduction/drug effects
16.
Mol Nutr Food Res ; 61(12)2017 12.
Article in English | MEDLINE | ID: mdl-28869341

ABSTRACT

SCOPE: Normal physiological processes require a robust biological timer called the circadian clock. Dysregulation of circadian rhythms contributes to a variety of metabolic syndrome, including obesity and insulin resistance. (-)-Epigallocatechin-3-gallate (EGCG) has been demonstrated to possess antioxidant, anti-inflammatory, and cardioprotective bioactivities. The objective of this study was to explore whether the circadian clock is involved in the protective effect of EGCG against insulin resistance. METHODS AND RESULTS: The results demonstrated that EGCG reverses the relatively shallow daily oscillations of circadian clock genes transcription and protein expression induced by glucosamine in HepG2 cells. EGCG also alleviates insulin resistance by enhancing tyrosine phosphorylated levels of IRS-1, stimulating the translocation of GLUT2, and activating PI3K/AKT as well as AMPK signaling pathways in a Bmal1-dependent manner both in HepG2 cells and primary hepatocytes. Glucosamine-stimulated excessive secretions of ROS and depletions of mitochondrial membrane potential were notably attenuated in EGCG co-treated HepG2 cells, which consistent with the recovery in expression of mitochondrial respiration complexes. CONCLUSION: The results demonstrated that EGCG possesses a Bmal1-dependent efficacy against insulin resistance conditions by strengthening the insulin signaling and eliminating oxidative stress, suggesting that EGCG may serve as a promising natural nutraceutical for the regulation of metabolic disorders relevant to circadian clocks.


Subject(s)
ARNTL Transcription Factors/metabolism , Catechin/analogs & derivatives , Hepatocytes/drug effects , Insulin Resistance , Mitochondria, Liver/drug effects , AMP-Activated Protein Kinases/metabolism , ARNTL Transcription Factors/genetics , Animals , Catechin/pharmacology , Cells, Cultured , Circadian Clocks/drug effects , Glucosamine/pharmacology , Glucose/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Insulin Resistance/physiology , Insulin-Like Growth Factor I/metabolism , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Oxidative Stress/drug effects
17.
FASEB J ; 31(11): 4998-5011, 2017 11.
Article in English | MEDLINE | ID: mdl-28739640

ABSTRACT

Obesity, which is caused by an energy imbalance between calorie intake and consumption, has become a major international health burden. Obesity increases the risk of insulin resistance and age-related cognitive decline, accompanied by peripheral inflammation. (-)-Epigallocatechin-3-gallate (EGCG), the major polyphenol in green tea, possesses antioxidant, anti-inflammatory, and cardioprotective activities; however, few reports have focused on its potential effect on cognitive disorders. In this study, our goal was to investigate the protective effects of EGCG treatment on insulin resistance and memory impairment induced by a high-fat and high-fructose diet (HFFD). We randomly assigned 3-mo-old C57BL/6J mice to 3 groups with different diets: control group, HFFD group, and HFFD plus EGCG group. Memory loss was assessed by using the Morris water maze test, during which EGCG was observed to prevent HFFD-elicited memory impairment and neuronal loss. Consistent with these results, EGCG attenuated HFFD-induced neuronal damage. Of note, EGCG significantly ameliorated insulin resistance and cognitive disorder by up-regulating the insulin receptor substrate-1 (IRS-1)/AKT and ERK/cAMP response element binding protein (CREB)/brain-derived neurotrophic factor (BDNF) signaling pathways. Long-term HFFD-triggered neuroinflammation was restored by EGCG supplementation by inhibiting the MAPK and NF-κB pathways, as well as the expression of inflammatory mediators, such as TNF-α. EGCG also reversed high glucose and glucosamine-induced insulin resistance in SH-SY5Y neuronal cells by improving the oxidized cellular status and mitochondrial function. To our knowledge, this study is the first to provide compelling evidence that the nutritional compound EGCG has the potential to ameliorate HFFD-triggered learning and memory loss.-Mi, Y., Qi, G., Fan, R., Qiao, Q., Sun, Y., Gao, Y., Liu, X. EGCG ameliorates high-fat- and high-fructose-induced cognitive defects by regulating the IRS/AKT and ERK/CREB/BDNF signaling pathways in the CNS.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Catechin/analogs & derivatives , Cyclic AMP Response Element-Binding Protein/metabolism , Dietary Carbohydrates/adverse effects , Dietary Fats/adverse effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fructose/adverse effects , Insulin Receptor Substrate Proteins/metabolism , Learning Disabilities/metabolism , MAP Kinase Signaling System/drug effects , Memory Disorders/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Catechin/pharmacology , Cell Line , Dietary Carbohydrates/pharmacology , Dietary Fats/pharmacology , Fructose/pharmacology , Learning Disabilities/chemically induced , Learning Disabilities/pathology , Memory Disorders/chemically induced , Memory Disorders/pathology , Mice
18.
Sci Rep ; 7(1): 2275, 2017 05 23.
Article in English | MEDLINE | ID: mdl-28536473

ABSTRACT

Athyrium multidentatum (Doll.) Ching (AMC), a unique and nutritious potherb widely distributed in china, has been extensively used in traditional Chinese medicine. Previous studies indicated that AMC extract exhibited antioxidant and antitumor properties. However, the chemical composition of AMC and molecular mechanism of AMC toxicity to HepG2 cells have not yet been elucidated. Hence, this study aimed to investigate the chemical compositions and the underlying mechanisms of the antiproliferative and apoptotic effects of AMC on HepG2. HPLC-MS analysis showed that AMC contain five compounds with chlorogenic acid accounting for 43 percent. Also, AMC strongly inhibited the cell growth and induced apoptosis and cell cycle arrest in HepG2 cells by significantly upregulating the protein expressions of Fas, Fas-L, Bax/Bcl-2, cyto-c, cleaved caspase-3, and PARP in a dose-dependent manner, which indicates AMC induces apoptosis in HepG2 cells through both intrinsic and extrinsic pathways. Moreover, AMC provoked the production of ROS, H2O2, and NO, modulating the PI3K/Akt, MAPK, NFκB and Nrf2 pathways and their downstream transcriptional cascades, ultimately evoked oxidative stress and apoptosis in HpeG2 cells. Further in vivo experiments demonstrated that AMC significantly suppressed the tumor growth, suggesting that AMC may be a novel promising agent for hepatocellular carcinoma treatment.


Subject(s)
Apoptosis/drug effects , Ferns/chemistry , Mitochondria/drug effects , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Animals , Cell Cycle Checkpoints/drug effects , Cell Line , Hep G2 Cells , Hepatoblastoma/drug therapy , Hepatoblastoma/metabolism , Hepatoblastoma/pathology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Mitochondria/metabolism , Plant Extracts/chemistry , Xenograft Model Antitumor Assays
19.
Sci Rep ; 7: 45728, 2017 04 04.
Article in English | MEDLINE | ID: mdl-28374807

ABSTRACT

Sesamol, a nutritional phenolic antioxidant compound enriched in sesame seeds, has been shown to have potential anticancer activities. This study aims at characterizing the antitumor efficacy of sesamol and unveiling the importance of mitochondria in sesamol-induced effects using a human hepatocellular carcinoma cell line, HepG2 cells. Results of this study showed that sesamol treatment suppressed colony formation, elicited S phase arrest during cell cycle progression, and induced both intrinsic and extrinsic apoptotic pathway in vitro with a dose-dependent manner. Furthermore, sesamol treatment elicited mitochondrial dysfunction by inducing a loss of mitochondrial membrane potential. Impaired mitochondria and accumulated H2O2 production resulted in disturbance of redox-sensitive signaling including Akt and MAPKs pathways. Mitochondrial biogenesis was inhibited as suggested by the decline in expression of mitochondrial complex I subunit ND1, and the upstream AMPK/PGC1α signals. Importantly, sesamol inhibited mitophagy and autophagy through impeding the PI3K Class III/Belin-1 pathway. Autophagy stimulator rapamycin reversed sesamol-induced apoptosis and mitochondrial respiration disorders. Moreover, it was also shown that sesamol has potent anti-hepatoma activity in a xenograft nude mice model. These data suggest that mitochondria play an essential role in sesamol-induced HepG2 cells death, and further research targeting mitochondria will provide more chemotherapeutic opportunities.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Benzodioxoles/pharmacology , Phenols/pharmacology , Animals , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Sirolimus/pharmacology , Xenograft Model Antitumor Assays
20.
Biochim Biophys Acta Mol Basis Dis ; 1863(6): 1575-1589, 2017 06.
Article in English | MEDLINE | ID: mdl-28412321

ABSTRACT

In response to the daily light-dark (LD) cycle, organisms on Earth have evolved with the approximately 24-h endogenous oscillations to coordinate behavioral and physiological processes, including feeding, sleep, and metabolism homeostasis. Circadian desynchrony triggered by an energy-dense diet rich in fats and fructose is intimately connected with a series of metabolic disorders. Previous studies revealed that (-)-Epigallocatechin-3-gallate (EGCG) could mitigate metabolic misalignment; however, only a few reports have focused on its potential effect on directly manipulating circadian rhythms to ameliorate metabolic syndrome. Our goal was to investigate the regulating effect of EGCG treatment on metabolic misalignment triggered by a high-fat and high-fructose diet (HFFD) associating with the circadian clock. Our results indicated that HFFD treatment partially exhibited poor circadian oscillations of the core clock gene and the clock-controlled gene in the liver and fat relative to the control group. EGCG administration may ameliorate the diet-dependent decline in circadian function by controlling the Sirt1-PGC1αloop, implying the existence of an EGCG-entrainable oscillator. Subsequently, reducing fatty acid synthesis and elevating ß-oxidation in the liver coupled with the increasing brown adipose tissue (BAT) energy expenditure observed in the EGCG group of mice prevented the adipocyte hypertrophy and fat accumulations common to BAT and white adipose tissue (WAT) derived from the HFFD mice. This study is the first to provide compelling evidences that EGCG may ameliorate diet-induced metabolic misalignment by regulating the rhythmic expression of the circadian clock genes in the liver and fat.


Subject(s)
Catechin/analogs & derivatives , Circadian Clocks/drug effects , Dietary Carbohydrates/adverse effects , Fructose/adverse effects , Metabolic Syndrome/drug therapy , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Catechin/pharmacology , Dietary Carbohydrates/pharmacology , Fructose/pharmacology , Liver/metabolism , Liver/pathology , Metabolic Syndrome/chemically induced , Metabolic Syndrome/metabolism , Metabolic Syndrome/pathology , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...