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










Publication year range
1.
J Nutr Biochem ; 126: 109571, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38199310

ABSTRACT

Maternal nutrient intake influences the health of the offspring via microenvironmental systems in digestion and absorption. Maternal high fructose diet (HFD) impairs hippocampus-dependent memory in adult female rat offspring. However, the underlying mechanisms remain largely unclear. Maternal HFD causes microbiota dysbiosis. In this study, we find that the plasma level of butyrate, a major metabolite of microbiota, is significantly decreased in the adult female maternal HFD offspring. In these rats, GPR43, a butyrate receptor was downregulated in the hippocampus. Moreover, the expressions of mitochondrial transcription factor A (TFAM), and peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α) were downregulated in the hippocampus. The decreases of these functional proteins were reversed by fructooligosaccharides (FOS, a probiotic) treatment in adulthood. Astrocytes are critical for energy metabolism in the brain. Primary astrocyte culture from female maternal HFD offspring indicated that GPR43 and the mitochondrial biogenesis were significantly suppressed, which was reversed by supplemental butyrate incubation. The oxygen consumption rate (OCR) was reduced in the HFD group and rescued by butyrate. Intriguingly, the nuclear histone deacetylase 4 (HDAC4) was enhanced in the HFD group, suggesting an inhibitory role of butyrate on histone deacetylase activity. Inhibition of HDAC4 effectively restored the OCR, bioenergetics, and biogenesis of mitochondria. Together, these results suggested that the impaired butyrate signaling by maternal HFD could underlie the reduced mitochondrial functions in the hippocampus via HDAC4-mediated epigenetic changes.


Subject(s)
Astrocytes , Butyrates , Female , Animals , Rats , Butyrates/pharmacology , Energy Metabolism , Oxygen Consumption , Histone Deacetylases , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Diet, High-Fat
2.
Nutr Metab (Lond) ; 21(1): 4, 2024 Jan 02.
Article in English | MEDLINE | ID: mdl-38167066

ABSTRACT

Excessive fructose intake presents the major risk factor for metabolic cardiovascular disease. Perivascular adipose tissue (PVAT) is a metabolic tissue and possesses a paracrine function in regulating aortic reactivity. However, whether and how PVAT alters vascular function under fructose overconsumption remains largely unknown. In this study, male Sprague-Dawley rats (8 weeks old) were fed a 60% high fructose diet (HFD) for 12 weeks. Fasting blood sugar, insulin, and triglycerides were significantly increased by HFD intake. Plasma adiponectin was significantly enhanced in the HFD group. The expression of uncoupling protein 1 (UCP1) and mitochondrial mass were reduced in the aortic PVAT of the HFD group. Concurrently, the expression of peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) and mitochondrial transcription factor A (TFAM) were suppressed. Furthermore, decreased fusion proteins (OPA1, MFN1, and MFN2) were accompanied by increased fission proteins (FIS1 and phospho-DRP1). Notably, the upregulated α-smooth muscle actin (α-SMA) and osteocalcin in the PVAT were concurrent with the impaired reactivity of aortic contraction and relaxation. Coenzyme Q10 (Q, 10 mg/100 mL, 4 weeks) effectively reversed the aforementioned events induced by HFD. Together, these results suggested that the dysregulation of mitochondrial dynamics mediated HFD-triggered PVAT whitening to impair aortic reactivity. Fortunately, coenzyme Q10 treatment reversed HFD-induced PVAT whitening and aortic reactivity.

4.
Exp Neurobiol ; 31(5): 307-323, 2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36351841

ABSTRACT

Inflammation alters the neural stem cell (NSC) lineage from neuronal to astrogliogenesis. However, the underlying mechanism is elusive. Autophagy contributes to the decline in adult hippocampal neurogenesis under E. coli lipopolysaccharide (LPS) stimulation. SRY-box transcription Factor 2 (SOX2) is critical for NSC self-renewal and proliferation. In this study, we investigated the role of SOX2 in induced autophagy and hippocampal adult neurogenesis under LPS stimulation. LPS (5 ng•100 g-1•hour-1 for 7 days) was intraperitoneally infused into male Sprague-Dawley rats (8 weeks old) to induce mild systemic inflammation. Beclin 1 and autophagy protein 12 (Atg12) were significantly upregulated concurrent with decreased numbers of Ki67- and doublecortin (DCX)-positive cells in the dentate gyrus. Synchronically, the levels of phospho(p)-mTOR, the p-mTOR/mTOR ratio, p-P85s6k, and the p-P85s6k/P85s6k ratio were suppressed. In contrast, SOX2 expression was increased. The fluorescence micrographs indicated that the colocalization of Beclin 1 and SOX2 was increased in the subgranular zone (SGZ) of the dentate gyrus. Moreover, increased S100ß-positive astrocytes were colocalized with SOX2 in the SGZ. Intracerebroventricular infusion of 3-methyladenine (an autophagy inhibitor) effectively prevented the increases in Beclin 1, Atg12, and SOX2. The SOX2+-Beclin 1+ and SOX2+-S100ß+ cells were reduced. The levels of p-mTOR and p-P85s6k were enhanced. Most importantly, the number of DCX-positive cells was preserved. Altogether, these data suggest that LPS induced autophagy to inactivate the mTOR/P85s6k pathway, resulting in a decline in neural differentiation. SOX2 was upregulated to facilitate the NSC lineage, while the autophagy milieu could switch the SOX2-induced NSC lineage from neurogenesis to astrogliogenesis.

5.
Environ Toxicol ; 37(11): 2728-2742, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36214339

ABSTRACT

Fructose overconsumption promotes tumor progression. Neuroblastoma is a common extracranial tumor with about 50% 5-year survival rate in high-risk children. The anti-tumor effect of Tribulus terrestris might bring new hope to neuroblastoma therapy. However, whether fructose disturbs the therapeutic effect of T. terrestris is currently unknown. In this study, the mouse neuroblastoma cell line, Neuro 2a (N2a) cells, was used to investigate the therapeutic effects of T. terrestris extract at various dosages (0.01, 1, 100 ng/ml) in regular EMEM medium or extra added fructose (20 mM) for 24 h. 100 ng/ml T. terrestris treatment significantly reduced the cell viability, whereas the cell viabilities were enhanced at the dosages of 0.01 or 1 ng/ml T. terrestris in the fructose milieu instead. The inhibition effect of T. terrestris on N2a migration was blunted in the fructose milieu. Moreover, T. terrestris effectively suppressed mitochondrial functions, including oxygen consumption rates, the activities of electron transport enzymes, the expressions of mitochondrial respiratory enzymes, and mitochondrial membrane potential. These suppressions were reversed in the fructose group. In addition, the T. terrestris-suppressed mitofusin and the T. terrestris-enhance mitochondrial fission 1 protein were maintained at basal levels in the fructose milieu. Together, these results demonstrated that T. terrestris extract effectively suppressed the survival and migration of neuroblastoma via inhibiting mitochondrial oxidative phosphorylation and disturbing mitochondrial dynamics. Whereas, the fructose milieu blunted the therapeutic effect of T. terrestris, particularly, when the dosage is reduced.


Subject(s)
Fructose , Neuroblastoma , Animals , Cell Line , Fructose/pharmacology , Mice , Mitochondria , Neuroblastoma/drug therapy , Plant Extracts/pharmacology , Tribulus
6.
Biomed J ; 45(3): 491-503, 2022 06.
Article in English | MEDLINE | ID: mdl-34229104

ABSTRACT

BACKGROUND: Aortic valve stenosis (AS) is a common, lethal cardiovascular disease. There is no cure except the valve replacement at last stage. Therefore, an understanding of the detail mechanism is imperative to prevent and intervene AS. Metabolic syndrome (MetS) is one of the major risk factors of AS whereas fructose overconsuming tops the list of MetS risk factors. However, whether the fructose under physiological level induces AS is currently unknown. METHODS: The human valve interstitial cells (hVICs), a crucial source to develop calcification, were co-incubated with fructose at 2 or 20 mM to mimic the serum fructose at fasting or post-fructose consumption, respectively, for 24 h. The cell proliferation was evaluated by WST-1 assays. The expressions of osteogenic and fibrotic proteins, PI3K/AKT signaling, insulin receptor substrate 1 and mitochondrial dynamic proteins were detected by Western blot analyses. The mitochondrial oxidative phosphorylation (OXPHOS) was examined by Seahorse analyzer. RESULTS: hVICs proliferation was significantly suppressed by 20 mM fructose. The expressions of alkaline phosphatase (ALP) and osteocalcin were enhanced concurrent with the upregulated PI3K p85, AKT, phospho(p)S473-AKT, and pS636-insulin receptor substrate 1 (p-IRS-1) by high fructose. Moreover, ATP production capacity and maximal respiratory capacity were enhanced in the high fructose groups. Synchronically, the expressions of mitochondrial fission 1 and optic atrophy type 1 were increased. CONCLUSIONS: These results suggested that high fructose stimulated the osteogenic differentiation of hVICs via the activation of PI3K/AKT/mitochondria signaling at the early stage. These results implied that high fructose at physiological level might have a direct, hazard effect on the progression of AS.


Subject(s)
Aortic Valve Stenosis , Osteogenesis , Cell Differentiation , Cells, Cultured , Fructose/pharmacology , Humans , Insulin Receptor Substrate Proteins/metabolism , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/pharmacology
7.
Am J Physiol Endocrinol Metab ; 320(6): E1173-E1182, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33969706

ABSTRACT

Retinopathy is a leading cause of blindness, and there is currently no cure. Earlier identification of the progression of retinopathy could provide a better chance for intervention. Diet has profound effects on retinal function. A maternal high-fructose diet (HFD) triggers diseases in multiple organs. However, whether maternal HFD impairs retinal function in adult offspring is currently unknown. By using the rodent model of maternal HFD during pregnancy and lactation, our data indicated a reduced b-wave of electroretinography (ERG) in HFD female offspring at 3 mo of age compared with age-matched offspring of dams fed regular chow (ND). Immunofluorescence and Western blot analyses indicated that the distributions and expressions of synaptophysin, postsynaptic density protein 95 (PSD95), and phospho(p)-Ca2+/calmodulin-stimulated protein kinase IIα (CaMKIIα) were significantly suppressed in the HFD group. Furthermore, the ATP content and the mitochondrial respiratory protein, Mt CPX 4-2, were decreased. Moreover, the expressions of peroxisome proliferator-activated receptor γ coactivator 1-α (PGC-1α) and mitochondrial transcription factor A (TFAM) in the retina of the HFD group were downregulated. Treatment with coenzyme Q10 (Q10), a key mediator of the electron transport chain, effectively reversed these abovementioned dysfunctions. Together, these results suggested that maternal HFD impaired retinal function in adult female offspring. The mechanism underlying early-onset retinopathy may involve the reduction in the capacity of mitochondrial energy production and the suppression of synaptic plasticity. Most importantly, mitochondria could be a feasible target to reprogram maternal HFD-damaged retinal function.NEW & NOTEWORTHY In this study, we provide novel evidence that maternal high-fructose diet during gestation and lactation could induce early-onset retinopathy in adult female offspring. Of note, the insufficient energy content, downregulated mitochondrial respiratory complex 4-2, and impaired mitochondrial biogenesis might contribute to the decrease of synaptic plasticity resulting in retinal function suppression. Oral application with coenzyme Q10 for 4 wk could at least partially reverse the aforementioned molecular events and retinal function.


Subject(s)
Fructose/adverse effects , Mitochondria/drug effects , Neuronal Plasticity/drug effects , Prenatal Exposure Delayed Effects , Retinal Diseases/chemically induced , Age Factors , Animals , Diet, High-Fat/adverse effects , Dietary Carbohydrates/adverse effects , Dietary Carbohydrates/pharmacology , Down-Regulation/drug effects , Female , Fructose/pharmacology , Male , Maternal Nutritional Physiological Phenomena , Mitochondria/physiology , Organelle Biogenesis , Pregnancy , Prenatal Exposure Delayed Effects/etiology , Prenatal Exposure Delayed Effects/physiopathology , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Sprague-Dawley , Retinal Diseases/physiopathology
8.
Article in English | MEDLINE | ID: mdl-32492926

ABSTRACT

Maternal high-fructose diets (HFD) impair the learning and memory capacity of adult female offspring via histone deacetylase 4 (HDAC4). Hippocampal adult neurogenesis is important for supporting the function of existing neural circuits. In this study, we investigated the effects of maternal HFD on hippocampal neural stem cell (NSC) proliferation and neuronal differentiation in adult offspring. Increased nuclear HDAC4 enzyme activity was detected in the hippocampus of HFD female offspring. The Western blot analyses indicated that the expressions of sex-determining region Y box2 (SOX2) and the transcription factor Paired Box 6 (PAX6), which are critical for the progression of NSC proliferation and differentiation, were downregulated. Concurrently, the expression of Ki67 (a cellular marker for proliferation) and doublecortin (DCX), which are related to NSC division and neuronal differentiation, was suppressed. Intracerebroventricular infusion with class II HDAC inhibitor (Mc1568, 4 weeks) led to the upregulation of these proteins. Environmental stimulation reversed the expression of Ki67 and DCX and the counts of Ki67- and DCX-positive cells in the hippocampi of HFD offspring as a result of providing the enriched housing for 4 weeks. Together, these results demonstrate that the suppressive effects of maternal HFD on hippocampal NSC proliferation and neuronal differentiation are reversibly mediated through HDAC4 and can be effectively reversed by environmental stimulation. The advantageous effects of environmental enrichment were possibly mediated by HDAC4 suppression.


Subject(s)
Dentate Gyrus , Diet , Fructose , Histone Deacetylases , Adult , Cell Differentiation , Cell Proliferation , Female , Hippocampus , Humans , Neurogenesis
9.
J Nutr Biochem ; 81: 108378, 2020 07.
Article in English | MEDLINE | ID: mdl-32330843

ABSTRACT

The mechanisms beneath the initiation of neuroinflammation are still inconclusive. Growing evidence proposes the maternal effect on the development of neuroinflammation. In this study, we evaluated the upstream regulators and the indices of neuroinflammation in the hippocampi of female offspring at 3 months old. The accumulation of nuclear factor-κB (NF-κB, 65 kDa), a cytokine-encoding transcription factor, was increased in microglia. The enhanced microglial activation was detected in CA1, CA3 and dentate gyrus (DG) HFD group with upregulation of CD11b and ionized calcium binding adaptor molecule 1 (Iba-1). Moreover, proinflammatory cytokines (including TNFα, IL-1ß and IL-6) were significantly increased in HFD group. Peroxisome proliferator-activated receptors γ (PPARγ) is a transcription factor involved in the suppression of NF-κB expression and in encoding endogenous antioxidants (such as catalase and glutathione peroxidases). On the contrary, the expression of nuclear PPARγ was suppressed in hippocampal neurons of the HFD group. In addition, the expressions of glutathione peroxidase 1 (GPx1) was suppressed in HFD group. Oral application with pioglitazone, a PPARγ agonist, effectively ceased the neuroinflammation and reversed the expression of antioxidants in HFD group. Together, these results for the first time demonstrated that maternal HFD triggered the waxing and waning of NF-κB and PPARγ may initiate neuroinflammation in the hippocampus of adult female offspring. Our findings further suggest that PPARγ could be the feasible targets to reprogram the hippocampal impairment induced by maternal HFD.


Subject(s)
Fructose/pharmacology , Hippocampus/metabolism , Inflammation/metabolism , NF-kappa B/metabolism , PPAR gamma/metabolism , Animals , Cytokines/metabolism , Diet/methods , Female , Fructose/adverse effects , Hippocampus/drug effects , Inflammation/etiology , Inflammation Mediators/metabolism , Male , Microglia/drug effects , Microglia/metabolism , Neurogenic Inflammation , Neurons/metabolism , Pioglitazone/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
10.
J Neuroinflammation ; 16(1): 224, 2019 Nov 15.
Article in English | MEDLINE | ID: mdl-31729994

ABSTRACT

BACKGROUND: Decreased heart rate variability (HRV) leads to cardiovascular diseases and increased mortality in clinical studies. However, the underlying mechanisms are still inconclusive. Systemic inflammation-induced neuroinflammation is known to impair the autonomic center of cardiovascular regulation. The dynamic stability of blood pressure and heart rate (HR) is regulated by modulation of the reciprocal responses of sympathetic and parasympathetic tone by the baroreflex, which is controlled by the nucleus of the solitary tract (NTS). METHODS: Systemic inflammation was induced by E. coli lipopolysaccharide (LPS, 1.2 mg/kg/day, 7 days) peritoneal infusion via an osmotic minipump in normotensive Sprague-Dawley rats. Systolic blood pressure (SBP) and HR were measured by femoral artery cannulation and recorded on a polygraph under anesthesia. The low-frequency (LF; 0.25-0.8 Hz) and high-frequency (HF; 0.8-2.4 Hz) components of SBP were adopted as the indices for sympathetic vasomotor tone and parasympathetic vasomotor tone, while the baroreflex effectiveness index (BEI) was adopted from the analysis of SBP and pulse interval (PI). The plasma levels of proinflammatory cytokines and mitochondrial DNA (mtDNA) oxidative damage were analyzed by ELISA. Protein expression was evaluated by Western blot. The distribution of oxidative mtDNA was probed by immunofluorescence. Pharmacological agents were delivered via infusion into the cisterna magna with an osmotic minipump. RESULTS: The suppression of baroreflex sensitivity was concurrent with increased SBP and decreased HR. Neuroinflammatory factors, including TNF-α, CD11b, and Iba-1, were detected in the NTS of the LPS group. Moreover, indices of mtDNA damage, including 8-OHdG and γ-H2AX, were significantly increased in neuronal mitochondria. Pentoxifylline or minocycline intracisternal (IC) infusion effectively prevented mtDNA damage, suggesting that cytokine and microglial activation contributed to mtDNA damage. Synchronically, baroreflex sensitivity was effectively protected, and the elevated blood pressure was significantly relieved. In addition, the mtDNA repair mechanism was significantly enhanced by pentoxifylline or minocycline. CONCLUSION: These results suggest that neuronal mtDNA damage in the NTS induced by neuroinflammation could be the core factor in deteriorating baroreflex desensitization and subsequent cardiovascular dysfunction. Therefore, the enhancement of base excision repair (BER) signaling in mitochondria could be a potential therapeutic strategy for cardiovascular reflex dysregulation.


Subject(s)
Baroreflex/physiology , DNA, Mitochondrial , Inflammation/physiopathology , Solitary Nucleus/physiopathology , Animals , Baroreflex/drug effects , Blood Pressure/physiology , DNA, Mitochondrial/drug effects , Heart Rate/physiology , Inflammation/chemically induced , Lipopolysaccharides/toxicity , Male , Rats , Rats, Sprague-Dawley
11.
J Nutr Biochem ; 69: 87-97, 2019 07.
Article in English | MEDLINE | ID: mdl-31063919

ABSTRACT

High fructose ingestion enhances mortality which has been linked to autonomic dysregulation. However, the underlying mechanisms are still largely unknown. In the present study, we demonstrated that 3 months of high fructose diet (HFD) ingestion induced mortality in 18-week-old Wistar Kyoto rats (WKY) during anesthesia. Concurrently, the low frequency (LF) and the high frequency (HF) elements of the power spectral analyses of SBP were increased. Of note, the decreased ratio of LF and HF (LF/HF), an index of sympathetic and parasympathetic balance, suggested an autonomic imbalance. In the rostral ventrolateral medulla (RVLM), a center of sympathetic outflow, the levels of presynaptic (synaptophysin) and postsynaptic (postsynaptic density protein 95 and phospho-Ca2+/calmodulin-dependent protein kinase II) proteins were increased. The down-regulation of insulin receptor ß and insulin receptor substrate 1 suggested the status of insulin desensitization. Moreover, the up-regulation of AMP-activated protein kinase and sirtuin 1 suggested the enhancement of energy sensing to activate autophagy. Simultaneously, the accumulations of Beclin-1, ATG12 and LC3B were increased in RVLM. Pioglitazone (PIO), an insulin sensitizer, effectively relieved the accumulation of Beclin-1 and ATG12 as well as the synaptic proteins synchronized with the reverses of insulin and energy sensing signals. Moreover, the autonomic dysregulation and anesthesia-associated mortality were intervened. Together, these results suggested that the HFD-induced, anesthesia-associated mortality rate was related to the autonomic abnormality derived from the RVLM synaptic alteration, which is strongly related to insulin desensitization-associated autophagy. PIO intervened the HFD-induced mortality via reversal of the above-mentioned molecules.


Subject(s)
Autophagy/drug effects , Fructose/adverse effects , Medulla Oblongata/drug effects , Pioglitazone/pharmacology , Animals , Autophagy/physiology , Autophagy-Related Protein 12/metabolism , Beclin-1/metabolism , Diet/adverse effects , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Male , Medulla Oblongata/metabolism , Medulla Oblongata/pathology , Metabolic Syndrome/drug therapy , Metabolic Syndrome/etiology , Microtubule-Associated Proteins/metabolism , Mortality , Proteins/metabolism , Rats, Wistar
12.
Am J Physiol Endocrinol Metab ; 316(4): E622-E634, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30668149

ABSTRACT

Excessive maternal high-fructose diet (HFD) during pregnancy and lactation has been reported to cause metabolic disorders in the offspring. Whether the infant's brain metabolism is disturbed by maternal HFD is largely unknown. Brain energy metabolism is elevated dramatically during fetal and postnatal development, whereby maternal nutrition is a key factor that determines cellular metabolism. Astrocytes, a nonneuronal cell type in the brain, are considered to support the high-energy demands of neurons by supplying lactate. In this study, the effects of maternal HFD on astrocytic glucose metabolism were investigated using hippocampal primary cultures of female infants. We found that glycolytic capacity and mitochondrial respiration and electron transport chain were suppressed by maternal HFD. Mitochondrial DNA copy number and mitochondrial transcription factor A expression were suppressed by maternal HFD. Western blots and immunofluorescent images further indicated that the glucose transporter 1 was downregulated whereas the insulin receptor-α, phospho-insulin receptor substrate-1 (Y612) and the p85 subunit of phosphatidylinositide 3-kinase were upregulated in the HFD group. Pioglitazone, which is known to increase astrocytic glucose metabolism, effectively reversed the suppressed glycolysis, and lactate release was restored. Moreover, pioglitazone also normalized oxidative phosphorylation with an increase of cytosolic ATP. Together, these results suggest that maternal HFD impairs astrocytic energy metabolic pathways that were reversed by pioglitazone.


Subject(s)
Astrocytes/drug effects , Dietary Sugars/pharmacology , Fructose/pharmacology , Glycolysis/drug effects , Hypoglycemic Agents/pharmacology , Oxidative Phosphorylation/drug effects , Pioglitazone/pharmacology , Animals , Astrocytes/metabolism , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Female , Fetal Development , Glucose Transporter Type 1/drug effects , Glucose Transporter Type 1/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Primary Cell Culture , Rats , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism
13.
Biomed J ; 41(3): 169-183, 2018 06.
Article in English | MEDLINE | ID: mdl-30080657

ABSTRACT

BACKGROUND: α-synuclein (SNCA) accumulation in the substantia nigra is one of the characteristic pathologies of Parkinson's disease (PD). A53T missense mutations in the SNCA gene has been proved to enhance the expression of SNCA and accelerate the onset of PD. Mitochondrial dysfunction in SNCA aggregation has been under debate for decades but the causal relationship remains uncertain. At a later stage of PD, the cellular dysfunctions are complicated and multiple factors are tangled. Our aim here is to investigate the mitochondrial functional changes and clarify the main causal mechanism at earlier-stage of PD. METHODS: We used the mutant A53T SNCA-expressed neuro 2a (N2a) cells without detectable cell death to investigate: 1) whether SNCA overexpression impairs the mitochondrial respiration and biogenesis. 2) The role of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) signal in SNCA-induced mitochondria dysfunction. RESULTS: Accompanying with the increment of SNCA, reactive oxygen species (ROS) accumulation was increased. The maximal respiratory capacity was suppressed. Meanwhile, mitochondrial complex 1 activity and the activity of nicotinamide adenine dinucleotide (NADH) cytochrome C reductase (NCCR) were decreased. Moreover, the mitochondrial DNA (mtDNA) copy number was decreased. On the other hand, the nuclear peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC-1α), Nrf2, and the cytosolic mitochondrial transcription factor A (TFAM) were increased at an early stage and declined thereafter. Above factors triggered by SNCA were reversed by tBHQ, a Nrf2 activator. CONCLUSION: These results suggested that at an early stage, SNCA-overexpressed increase mtROS accumulation, mitochondrial dysfunction and mtDNA decrement. Nrf2, PGC-1α and TFAM were upregulated to compromise mitochondrial dysfunction. tBHQ effectively reversed the SNCA-induced mitochondrial dysfunction.


Subject(s)
Mitochondria/metabolism , NF-E2-Related Factor 2/physiology , Oxygen Consumption , alpha-Synuclein/physiology , Animals , Cell Line, Tumor , Hydroquinones/pharmacology , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/physiology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/physiology
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 274-285, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29054390

ABSTRACT

Diet-associated insulin resistance (IR) is intimately correlated with the progression of metabolic syndrome and hippocampal dysfunction. Pioglitazone (PIO), a selective peroxisome proliferator-activated receptor gamma (PPARγ) agonist, has been applied to enhance insulin sensitivity. With limited permeability to blood-brain-barrier, it is unclear that whether oral PIO available to cure both the peripheral IR and the impairment in the hippocampus. We evaluated the levels of peripheral and hippocampal IR via the homeostatic model assessment of insulin resistance and hippocampal IRS-1/Akt phosphorylation, respectively, of Wistar Kyoto rats fed with a regular chew or high fructose diet (HFD) for 12weeks. Gavage with PIO (30mg/kg/day, 2weeks) significantly reduced the peripheral IR and reversed the level of hippocampal PPARγ. Moreover, HFD-activated microglia and astrocyte were effectively relieved by PIO. The suppressed brain-derived neurotrophic factor, CaMKIIα, and postsynaptic density protein 95 in the hippocampus were effectively reversed by PIO. However, the hippocampal IR and inhibition of adult neurogenesis in dentate gyrus were not restored by PIO. Together, PIO oral application may reverse the HFD-induced peripheral IR and maintain the existed neuronal circuit by ameliorating glial activation and enhancing synaptic density through BDNF but failed to restore adult neurogenesis in the hippocampus.


Subject(s)
Adult Stem Cells/drug effects , Fructose/adverse effects , Gliosis/prevention & control , Hippocampus/drug effects , Insulin Resistance , Neural Stem Cells/drug effects , Neurogenesis/drug effects , Thiazolidinediones/pharmacology , Administration, Oral , Adult Stem Cells/physiology , Animals , Gliosis/metabolism , Gliosis/pathology , Hippocampus/metabolism , Hippocampus/pathology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Male , Neural Stem Cells/physiology , Neuroprotective Agents/pharmacology , Pioglitazone , Rats , Rats, Inbred WKY , Thiazolidinediones/administration & dosage
15.
J Nutr Biochem ; 41: 73-83, 2017 03.
Article in English | MEDLINE | ID: mdl-28063367

ABSTRACT

Both genetic and dietary factors determine the development of hypertension. Whether dietary factor impacts the development of hereditary hypertension is unknown. Here, we evaluated the effect of daily high-fructose diet (HFD) on the development of hypertension in adolescent spontaneously hypertensive rats (SHR). Six-week-old SHR were randomly divided into two groups to receive HFD or normal diet (ND) for 3 weeks. The temporal profile of systolic blood pressure, alongside the sympathetic vasomotor activity, in the SHR-HFD showed significantly greater increases at 9-12 weeks of age compared with the age-matched SHR-ND group. Immunofluorescence was used to identify the distribution of reactive oxygen species (ROS), oxidants and antioxidants in rostral ventrolateral medulla (RVLM) where sympathetic premotor neurons reside. In RVLM of SHR-HFD, the levels of ROS accumulation and lipid peroxidation were elevated. The changes in protein expression were measured by Western blot. NADPH oxidase subunit gp91phox and angiotensin II type I receptor were up-regulated in RVLM neuron. On the other hand, the expression of extracellular superoxide dismutase was suppressed. Both molecular and hemodynamic changes in the SHR-HFD were rescued by oral pioglitazone treatment from weeks 7 to 9. Furthermore, central infusion with tempol, a ROS scavenger, effectively ameliorated ROS accumulation in RVLM and diminished the heightened pressor response and enhanced sympathetic activity in the SHR-HFD. Together, these results suggest that HFD intake at adolescent SHR may impact the development of hypertension via increasing oxidative stress in RVLM which could be effectively attenuated by pioglitazone treatment.


Subject(s)
Diet, Carbohydrate Loading/adverse effects , Fructose/adverse effects , Hypertension/etiology , Medulla Oblongata/metabolism , NADPH Oxidase 2/metabolism , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction , Animals , Antihypertensive Agents/therapeutic use , Hypertension/metabolism , Hypertension/pathology , Hypertension/prevention & control , Hypoglycemic Agents/therapeutic use , Lipid Peroxidation/drug effects , Male , Medulla Oblongata/drug effects , Medulla Oblongata/pathology , NADPH Oxidase 2/chemistry , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Oxidative Stress/drug effects , Pioglitazone , Random Allocation , Rats, Inbred SHR , Rats, Inbred WKY , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/agonists , Signal Transduction/drug effects , Thiazolidinediones/therapeutic use
16.
Free Radic Biol Med ; 97: 58-74, 2016 08.
Article in English | MEDLINE | ID: mdl-27223823

ABSTRACT

Oxidative stress in rostral ventrolateral medulla (RVLM), where sympathetic premotor neurons reside, is involved in the development of hypertension under systemic inflammation. Mitochondrial dysfunction contributes to tissue oxidative stress. In this study, we sought to investigate whether hypertension developed under systemic inflammation is attributable to impaired mitochondrial biogenesis in RVLM. In normotensive Sprague-Dawley rats, intraperitoneal infusion of a low dose Escherichia coli lipopolysaccharide (LPS) for 7 days promoted a pressor response, alongside a decrease in mitochondrial DNA (mtDNA) copy number, reductions in protein expression of nuclear DNA-encoded transcription factors for mitochondrial biogenesis, including mitochondrial transcription factor A (TFAM) and nuclear factor erythroid-derived 2-like 2 (Nrf2), and suppression of nuclear translocation of the phosphorylated Nrf2 (p-Nrf2) in RVLM neurons; all of which were abrogated by treatment with intracisternal infusion of an interleukin-1ß (IL-1ß) blocker, IL-1Ra, or a mobile mitochondrial electron carrier, coenzyme Q10 (CoQ10). Microinjection into RVLM of IL-1ß suppressed the expressions of p-Nrf2 and TFAM, and evoked a pressor response; conversely, the Nrf2 inducer, tert-butylhydroquinone, lessened the LPS-induced suppression of TFAM expression and pressor response. At cellular level, exposure of neuronal N2a cells to IL-1ß decreased mtDNA copy number, increased protein interaction of Nrf2 to its negative regulator, kelch-like ECH-associated protein 1 (Keap1), and reduced DNA binding activity of p-Nrf2 to Tfam gene. Together these results indicate that defect mitochondrial biogenesis in RVLM neurons entailing redox-sensitive and IL-1ß-dependent suppression of TFAM because of the increase in the formation of Keap1/Nrf2 complex, reductions in nuclear translocation of the activated Nrf2 and its binding to the Tfam gene promoter may underlie hypertension developed under the LPS-induced systemic inflammation.


Subject(s)
Hypertension/genetics , Inflammation/genetics , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2 Transcription Factor/genetics , Transcription Factors/genetics , Animals , DNA Copy Number Variations/drug effects , DNA, Mitochondrial/drug effects , Humans , Hypertension/etiology , Hypertension/metabolism , Hypertension/pathology , Inflammation/chemically induced , Inflammation/complications , Inflammation/pathology , Interleukin-1beta/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Kidney Medulla/drug effects , Kidney Medulla/pathology , Lipopolysaccharides/toxicity , Mitochondria/genetics , Mitochondria/metabolism , NF-E2 Transcription Factor/metabolism , Neurons/metabolism , Neurons/pathology , Organelle Biogenesis , Oxidation-Reduction , Oxidative Stress/drug effects , Rats
17.
Neurobiol Learn Mem ; 130: 105-17, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26872592

ABSTRACT

Impairment of learning and memory has been documented in the later life of offspring to maternal consumption with high energy diet. Environmental stimulation enhances the ability of learning and memory. However, potential effects of environmental stimulation on the programming-associated deficit of learning and memory have not been addressed. Here, we examined the effects of enriched-housing on hippocampal learning and memory in adult female offspring rats from mother fed with 60% high fructose diet (HFD) during pregnancy and lactation. Impairment of spatial learning and memory performance in HFD group was observed in offspring at 3-month-old. Hippocampal brain-derived neurotrophic factor (BDNF) was decreased in the offspring. Moreover, the HFD group showed an up-regulation of histone deacetylase 4 (HDAC4) in the nuclear fractions of hippocampal neurons. Stimulation to the offspring for 4weeks after winning with an enriched-housing environment effectively rescued the decrease in cognitive function and hippocampal BDNF level; alongside a reversal of the increased distribution of nuclear HDAC4. Together these results suggest that later life environmental stimulation effectively rescues the impairment of hippocampal learning and memory in female offspring to maternal HFD intake through redistributing nuclear HDAC4 to increase BDNF expression.


Subject(s)
Environment , Fructose/administration & dosage , Histone Deacetylases/metabolism , Learning/drug effects , Memory/drug effects , Prenatal Exposure Delayed Effects/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cognition/drug effects , Female , Hippocampus/drug effects , Hippocampus/metabolism , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Spatial Learning/drug effects
18.
J Biomed Sci ; 21: 8, 2014 Jan 27.
Article in English | MEDLINE | ID: mdl-24467657

ABSTRACT

BACKGROUND: The increase in fructose ingestion has been linked to overdrive of sympathetic activity and hypertension associated with the metabolic syndrome. The premotor neurons for generation of sympathetic vasomotor activity reside in the rostral ventrolateral medulla (RVLM). Activation of RVLM results in sympathoexcitation and hypertension. Neurons in the central nervous system are able to utilize fructose as a carbon source of ATP production. We examined in this study whether fructose affects ATP content in RVLM and its significance in the increase in central sympathetic outflow and hypertension induced by the high fructose diet (HFD). RESULTS: In normotensive rats fed with high fructose diet (HFD) for 12 weeks, there was a significant increase in tissue ATP content in RVLM, accompanied by the increases in the sympathetic vasomotor activity and blood pressure. These changes were blunted by intracisternal infusion of an ATP synthase inhibitor, oligomycin, to the HFD-fed animals. In the catecholaminergic-containing N2a cells, fructose dose-dependently upregulated the expressions of glucose transporter 2 and 5 (GluT2, 5) and the rate-limiting enzyme of fructolysis, ketohexokinase (KHK), leading to the increases in pyruvate and ATP production, as well as the release of the neurotransmitter, dopamine. These cellular events were significantly prevented after the gene knocking down by lentiviral transfection of small hairpin RNA against KHK. CONCLUSION: These results suggest that increases in ATP content in RVLM may be engaged in the augmented sympathetic vasomotor activity and hypertension associated with the metabolic syndrome induced by the HFD. At cellular level, the increase in pyruvate levels via fructolysis is involved in the fructose-induced ATP production and the release of neurotransmitter.


Subject(s)
Adenosine Triphosphate/metabolism , Fructose/administration & dosage , Hypertension/metabolism , Oxidative Stress/genetics , Animals , Blood Pressure/genetics , Diet , Heart Rate , Hypertension/chemically induced , Hypertension/genetics , Medulla Oblongata/metabolism , Rats , Sympathetic Nervous System/metabolism , Vasomotor System/metabolism
19.
Mol Nutr Food Res ; 54 Suppl 2: S196-204, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20461740

ABSTRACT

Resveratrol exhibits potential anti-carcinogenic activities. Heme oxygenase-1 (HO-1) is involved in angiogenesis and tumor metastasis. Matrix metalloproteinases (MMPs) are key enzymes in the degradation of extracellular matrix, and their expression may be dysregulated in lung cancer metastasis. In this study, we investigated the anti-invasive mechanism of resveratrol in lung cancer cells. HO-1 was shown to be elevated (approximately 4.7-fold) in lung cancer tumor samples as compared with matched normal tissues. After treatment of lung adenocarcinoma cell line A549 cells with resveratrol (50 microM) for 24 h, the migratory and invasive abilities (38 and 30% inhibition, respectively) of A549 cells were significantly reduced. Resveratrol significantly inhibited HO-1-mediated MMP-9 (35% inhibition) and MMP-2 (28% inhibition) expression in lung cancer cells. Nuclear factor (NF)-kappaB inhibitor induced a marked reduction in MMP-9 and MMP-2 expression, suggesting NF-kappaB pathway could play an important role. Furthermore, HO-1 inhibition and silencing significantly suppressed MMPs and invasion of lung cancer cells. Our results suggest that resveratrol inhibited HO-1 and subsequently MMP-9 and MMP-2 expression in lung cancer cells. The inhibitory effects of resveratrol on MMP expression and invasion of lung cancer cells are, in part, associated with the HO-1-mediated NF-kappaB pathway.


Subject(s)
Adenocarcinoma/drug therapy , Down-Regulation/drug effects , Heme Oxygenase-1/metabolism , Lung Neoplasms/drug therapy , Matrix Metalloproteinases/metabolism , NF-kappa B/antagonists & inhibitors , Stilbenes/pharmacology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/secondary , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Gene Silencing , Heme Oxygenase-1/genetics , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Osmolar Concentration , RNA, Small Interfering , Resveratrol , Signal Transduction/drug effects
20.
Am J Respir Cell Mol Biol ; 43(5): 530-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-19933377

ABSTRACT

High-mobility group box 1 (HMGB1) is a versatile protein with intranuclear and extracellular functions. It is involved in invasion and metastasis in various human malignancies. However, the role of HMGB1 in non-small cell lung cancer (NSCLC) is unclear. We hypothesized that HMGB1 expression is a determinant of cellular invasiveness and metastasis in lung cancer. We examined HMGB1 expression in 48 NSCLC specimens by quantitative real-time PCR. High HMGB1 expression was significantly associated with clinically advanced stages (stage III-IV) (P < 0.05) and was correlated to expression of matrix metalloproteinase-9 (MMP-9) (P < 0.05). Patients with high levels of HMGB1 expression had poorer clinical prognosis. The expression level of MMP-9 and metastatic ability in vitro were significantly higher in an HMGB1-overexpressing human NSCLC cell lines (A549 and H23). The treatment with HMGB1 small interfering RNA reduced MMP-9 expression and the cellular metastatic ability in NSCLC cells. We also demonstrated that phosphoinositide 3-kinase/Akt and NF-κB-related pathways contributed to the HMGB1-induced MMP-9 expression and cellular metastatic ability.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , HMGB1 Protein/metabolism , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Matrix Metalloproteinase 9/metabolism , Adult , Aged , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cell Movement , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , HMGB1 Protein/genetics , Humans , Lung Neoplasms/genetics , Male , Matrix Metalloproteinase 2/metabolism , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...