Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Front Immunol ; 15: 1322842, 2024.
Article En | MEDLINE | ID: mdl-38455054

Autophagy is a conserved cellular mechanism that enables the degradation and recycling of cellular organelles and proteins via the lysosomal pathway. In neurodevelopment and maintenance of neuronal homeostasis, autophagy is required to regulate presynaptic functions, synapse remodeling, and synaptic plasticity. Deficiency of autophagy has been shown to underlie the synaptic and behavioral deficits of many neurological diseases such as autism, psychiatric diseases, and neurodegenerative disorders. Recent evidence reveals that dysregulated autophagy plays an important role in the initiation and progression of neuroinflammation, a common pathological feature in many neurological disorders leading to defective synaptic morphology and plasticity. In this review, we will discuss the regulation of autophagy and its effects on synapses and neuroinflammation, with emphasis on how autophagy is regulated by epigenetic mechanisms under healthy and diseased conditions.


Epigenesis, Genetic , Neuroinflammatory Diseases , Humans , Neurons/metabolism , Autophagy/genetics , Neuronal Plasticity/genetics
2.
Sci Rep ; 13(1): 12968, 2023 08 10.
Article En | MEDLINE | ID: mdl-37563287

Diabetic retinopathy is a common complication of long-term diabetes and that could lead to vision loss. Unfortunately, early diabetic retinopathy remains poorly understood. There is no effective way to prevent or treat early diabetic retinopathy until patients develop later stages of diabetic retinopathy. Elevated acellular capillary density is considered a reliable quantitative trait present in the early development of retinopathy. Hence, in this study, we interrogated whole retinal vascular transcriptomic changes via a Nile rat model to better understand the early pathogenesis of diabetic retinopathy. We uncovered the complexity of associations between acellular capillary density and the joint factors of blood glucose, diet, and sex, which was modeled through a Bayesian network. Using segmented regressions, we have identified different gene expression patterns and enriched Gene Ontology (GO) terms associated with acellular capillary density increasing. We developed a random forest regression model based on expression patterns of 14 genes to predict the acellular capillary density. Since acellular capillary density is a reliable quantitative trait in early diabetic retinopathy, and thus our model can be used as a transcriptomic clock to measure the severity of the progression of early retinopathy. We also identified NVP-TAE684, geldanamycin, and NVP-AUY922 as the top three potential drugs which can potentially attenuate the early DR. Although we need more in vivo studies in the future to support our re-purposed drugs, we have provided a data-driven approach to drug discovery.


Diabetes Mellitus , Diabetic Retinopathy , Animals , Diabetic Retinopathy/pathology , Retinal Vessels/pathology , Transcriptome , Bayes Theorem , Murinae , Diabetes Mellitus/pathology
3.
PLoS Biol ; 21(3): e3002033, 2023 03.
Article En | MEDLINE | ID: mdl-36928253

Aging is a systemic process, which is a risk factor for impaired physiological functions, and finally death. The molecular mechanisms driving aging process and the associated cognitive decline are not fully understood. The hypothalamus acts as the arbiter that orchestrates systemic aging through neuroinflammatory signaling. Our recent findings revealed that Menin plays important roles in neuroinflammation and brain development. Here, we found that the hypothalamic Menin signaling diminished in aged mice, which correlates with systemic aging and cognitive deficits. Restoring Menin expression in ventromedial nucleus of hypothalamus (VMH) of aged mice extended lifespan, improved learning and memory, and ameliorated aging biomarkers, while inhibiting Menin in VMH of middle-aged mice induced premature aging and accelerated cognitive decline. We further found that Menin epigenetically regulates neuroinflammatory and metabolic pathways, including D-serine metabolism. Aging-associated Menin reduction led to impaired D-serine release by VMH-hippocampus neural circuit, while D-serine supplement rescued cognitive decline in aged mice. Collectively, VMH Menin serves as a key regulator of systemic aging and aging-related cognitive decline.


Aging , Cognitive Dysfunction , Hypothalamus , Animals , Mice , Aging/genetics , Aging/metabolism , Cognitive Dysfunction/genetics , Cognitive Dysfunction/metabolism , Hypothalamus/metabolism , Serine/metabolism , Transcription Factors/metabolism
4.
Cell Rep ; 39(10): 110853, 2022 06 07.
Article En | MEDLINE | ID: mdl-35675768

Fragile X syndrome (FXS) is a leading cause of inherited intellectual disability and autism. Whereas dysregulated RNA translation in Fmr1 knockout (KO) mice, a model of FXS, is well studied, little is known about aberrant transcription. Using single-molecule mRNA detection, we show that mRNA encoding the AMPAR subunit GluA2 (but not GluA1) is elevated in dendrites and at transcription sites of hippocampal neurons of Fmr1 KO mice, indicating elevated GluA2 transcription. We identify CPEB3, a protein implicated in memory consolidation, as an upstream effector critical to GluA2 mRNA expression in FXS. Increased GluA2 mRNA is translated into an increase in GluA2 subunits, a switch in synaptic AMPAR phenotype from GluA2-lacking, Ca2+-permeable to GluA2-containing, Ca2+-impermeable, reduced inhibitory synaptic transmission, and loss of NMDAR-independent LTP at glutamatergic synapses onto CA1 inhibitory interneurons. These factors could contribute to an excitatory/inhibitory imbalance-a common theme in FXS and other autism spectrum disorders.


Fragile X Syndrome , RNA-Binding Proteins , Receptors, AMPA , Animals , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Interneurons/metabolism , Mice , Mice, Knockout , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism
6.
Stem Cell Res ; 59: 102642, 2022 Mar.
Article En | MEDLINE | ID: mdl-34971934

Neural precursor cells (NPCs) transplanted into the adult neocortex generate neurons that synaptically integrate with host neurons, supporting the possibility of achieving functional tissue repair. However, poor survival and functional neuronal recovery of transplanted NPCs greatly limits engraftment. Here, we test the hypothesis that combining blood vessel-forming vascular cells with neuronal precursors improves engraftment. By transplanting mixed embryonic neocortical cells into adult mice with neocortical strokes, we show that transplant-derived neurons synapse with appropriate targets while donor vascular cells form vessels that fuse with the host vasculature to perfuse blood within the graft. Although all grafts became vascularized, larger grafts had greater contributions of donor-derived vessels that increased as a function of their distance from the host-graft border. Moreover, excluding vascular cells from the donor cell population strictly limited graft size. Thus, inclusion of vessel-forming vascular cells with NPCs is required for more efficient engraftment and ultimately for tissue repair.

7.
Autophagy ; 16(8): 1506-1523, 2020 08.
Article En | MEDLINE | ID: mdl-31690173

Epidemiological and clinical studies have long shown that exposure to high levels of heavy metals are associated with increased risks of neurodegenerative diseases. It is widely accepted that autophagic dysfunction is involved in pathogenesis of various neurodegenerative disorders; however, the role of heavy metals in regulation of macroautophagy/autophagy is unclear. Here, we show that manganese (Mn) induces a decline in nuclear localization of TFEB (transcription factor EB), a master regulator of the autophagy-lysosome pathway, leading to autophagic dysfunction in astrocytes of mouse striatum. We further show that Mn exposure suppresses autophagic-lysosomal degradation of mitochondria and induces accumulation of unhealthy mitochondria. Activation of autophagy by rapamycin or TFEB overexpression ameliorates Mn-induced mitochondrial respiratory dysfunction and reactive oxygen species (ROS) generation in astrocytes, suggesting a causal relation between autophagic failure and mitochondrial dysfunction in Mn toxicity. Taken together, our data demonstrate that Mn inhibits TFEB activity, leading to impaired autophagy that is causally related to mitochondrial dysfunction in astrocytes. These findings reveal a previously unappreciated role for Mn in dysregulation of autophagy and identify TFEB as a potential therapeutic target to mitigate Mn toxicity. ABBREVIATIONS: BECN1: beclin 1; CTSD: cathepsin D; DMEM: Dulbecco's Modified Eagle Medium; GFAP: glial fibrillary acid protein; GFP: green fluorescent protein; HBSS: hanks balanced salt solution; LAMP: lysosomal-associated membrane protein; LDH: lactate dehydrogenase; Lys Inh: lysosomal inhibitors; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; Mn: manganese; MTOR: mechanistic target of rapamycin kinase; OCR: oxygen consumption rate; PBS: phosphate-buffered saline; PFA: paraformaldehyde; PI: propidium iodide; ROS: reactive oxygen species; s.c.: subcutaneous; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TFEB: transcription factor EB.


Astrocytes/metabolism , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Manganese/pharmacology , Mitochondria/pathology , Animals , Astrocytes/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Down-Regulation/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Neostriatum/drug effects , Neostriatum/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , Sirolimus/pharmacology
8.
Curr Opin Neurobiol ; 59: 207-212, 2019 12.
Article En | MEDLINE | ID: mdl-31634675

In neurons, autophagy is crucial to proper axon guidance, vesicular release, dendritic spine architecture, spine pruning and synaptic plasticity and, when dysregulated, is associated with brain disorders, including autism spectrum disorders, and neurodegenerative diseases such as Parkinson's and Alzheimer's disease. Once thought to play a housekeeping function of removing misfolded proteins or compromised organelles, neuronal autophagy is now regarded as a finely tuned, real time surveillance and clearance system crucial to synaptic integrity and function. Here we review the role of autophagy in synaptic plasticity and its regulation by epigenetic mechanisms.


Autophagy , Epigenesis, Genetic , Dendritic Spines , Neuronal Plasticity , Neurons
9.
Am Ann Deaf ; 163(5): 554-573, 2019.
Article En | MEDLINE | ID: mdl-30713198

Deaf and hard of hearing (DHH) students tend to experience delayed development of grammatical skills in written language. However, much remains unknown about the mechanism behind this phenomenon. In the present study, the researchers used a self-paced moving-window reading task to investigate DHH students' understanding of causal and adversative connectives in Chinese. The students were similar to a hearing control group in their comprehension of the relationship between the clauses in a causal sentence. However, the DHH students were more likely than their hearing peers to find it harder to understand adversative connectives than causal connectives. More studies are needed to reveal how DHH students deal with other syntactic structures that are particularly difficult for them to learn. Educators should pay close attention to creating learning environments in which DHH students can acquire those syntactic structures in the process of using language.


Comprehension , Deafness/psychology , Education of Hearing Disabled , Reading , Adolescent , Female , Humans , Linguistics , Male , Reaction Time , Young Adult
10.
Proc Natl Acad Sci U S A ; 115(41): E9707-E9716, 2018 10 09.
Article En | MEDLINE | ID: mdl-30242133

Fragile X syndrome (FXS) is the most frequent form of heritable intellectual disability and autism. Fragile X (Fmr1-KO) mice exhibit aberrant dendritic spine structure, synaptic plasticity, and cognition. Autophagy is a catabolic process of programmed degradation and recycling of proteins and cellular components via the lysosomal pathway. However, a role for autophagy in the pathophysiology of FXS is, as yet, unclear. Here we show that autophagic flux, a functional readout of autophagy, and biochemical markers of autophagy are down-regulated in hippocampal neurons of fragile X mice. We further show that enhanced activity of mammalian target of rapamycin complex 1 (mTORC1) and translocation of Raptor, a defining component of mTORC1, to the lysosome are causally related to reduced autophagy. Activation of autophagy by delivery of shRNA to Raptor directly into the CA1 of living mice via the lentivirus expression system largely corrects aberrant spine structure, synaptic plasticity, and cognition in fragile X mice. Postsynaptic density protein (PSD-95) and activity-regulated cytoskeletal-associated protein (Arc/Arg3.1), proteins implicated in spine structure and synaptic plasticity, respectively, are elevated in neurons lacking fragile X mental retardation protein. Activation of autophagy corrects PSD-95 and Arc abundance, identifying a potential mechanism by which impaired autophagy is causally related to the fragile X phenotype and revealing a previously unappreciated role for autophagy in the synaptic and cognitive deficits associated with fragile X syndrome.


Autophagy , CA1 Region, Hippocampal/metabolism , Fragile X Syndrome/metabolism , Synapses/metabolism , Animals , CA1 Region, Hippocampal/pathology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Synapses/genetics , Synapses/pathology
11.
Nature ; 560(7719): E33, 2018 08.
Article En | MEDLINE | ID: mdl-29950723

The microarray data generated and analysed in this Article have been uploaded to the Gene Expression Omnibus (GEO) under accession number GSE113383 . Accordingly, the 'Data availability' section of the Methods of the original Article has been rephrased online.

12.
Nature ; 548(7665): 52-57, 2017 08 03.
Article En | MEDLINE | ID: mdl-28746310

It has been proposed that the hypothalamus helps to control ageing, but the mechanisms responsible remain unclear. Here we develop several mouse models in which hypothalamic stem/progenitor cells that co-express Sox2 and Bmi1 are ablated, as we observed that ageing in mice started with a substantial loss of these hypothalamic cells. Each mouse model consistently displayed acceleration of ageing-like physiological changes or a shortened lifespan. Conversely, ageing retardation and lifespan extension were achieved in mid-aged mice that were locally implanted with healthy hypothalamic stem/progenitor cells that had been genetically engineered to survive in the ageing-related hypothalamic inflammatory microenvironment. Mechanistically, hypothalamic stem/progenitor cells contributed greatly to exosomal microRNAs (miRNAs) in the cerebrospinal fluid, and these exosomal miRNAs declined during ageing, whereas central treatment with healthy hypothalamic stem/progenitor cell-secreted exosomes led to the slowing of ageing. In conclusion, ageing speed is substantially controlled by hypothalamic stem cells, partially through the release of exosomal miRNAs.


Aging/genetics , Aging/physiology , Exosomes/genetics , Hypothalamus/cytology , Hypothalamus/physiology , Longevity/physiology , MicroRNAs/genetics , Neural Stem Cells/physiology , Aging/cerebrospinal fluid , Aging/pathology , Animals , Cellular Microenvironment , Exosomes/metabolism , Hypothalamus/pathology , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Inflammation , Longevity/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/cerebrospinal fluid , MicroRNAs/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Polycomb Repressive Complex 1/deficiency , Proto-Oncogene Proteins/deficiency , SOXB1 Transcription Factors/deficiency , Time Factors
14.
Neurobiol Dis ; 95: 82-92, 2016 Nov.
Article En | MEDLINE | ID: mdl-27425889

Diabetes is a major stroke risk factor and is associated with poor functional recovery after stroke. Accumulating evidence indicates that the worsened outcomes may be due to hyperglycemia-induced cerebral vascular complications, especially disruption of the blood-brain barrier (BBB). The present study tested a hypothesis that the activation of hypoxia inducible factor-1 (HIF-1) was involved in hyperglycemia-aggravated BBB disruption in an ischemic stroke model. Non-diabetic control and Streptozotocin-induced type I diabetic mice were subjected to 90min transient middle cerebral artery occlusion (MCAO) followed by reperfusion. Our results demonstrated that hyperglycemia induced higher expression of HIF-1α and vascular endothelial growth factor (VEGF) in brain microvessels after MCAO/reperfusion. Diabetic mice showed exacerbated BBB damage and tight junction disruption, increased infarct volume as well as worsened neurological deficits. Furthermore, suppressing HIF-1 activity by specific knock-out endothelial HIF-1α ameliorated BBB leakage and brain infarction in diabetic animals. Moreover, glycemic control by insulin abolished HIF-1α up-regulation in diabetic animals and reduced BBB permeability and brain infarction. These findings strongly indicate that HIF-1 plays an important role in hyperglycemia-induced exacerbation of BBB disruption in ischemic stroke. Endothelial HIF-1 inhibition warrants further investigation as a therapeutic target for the treatment of stroke patients with diabetes.


Blood-Brain Barrier/metabolism , Brain Ischemia/metabolism , Brain/metabolism , Hyperglycemia/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Stroke/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Mice, Transgenic , Signal Transduction/physiology , Stroke/physiopathology , Up-Regulation
15.
Nat Commun ; 6: 6704, 2015 Apr 07.
Article En | MEDLINE | ID: mdl-25848677

Metabolic homeostasis is regulated by the brain, but whether this regulation involves learning and memory of metabolic information remains unexplored. Here we use a calorie-based, taste-independent learning/memory paradigm to show that Drosophila form metabolic memories that help in balancing food choice with caloric intake; however, this metabolic learning or memory is lost under chronic high-calorie feeding. We show that loss of individual learning/memory-regulating genes causes a metabolic learning defect, leading to elevated trehalose and lipid levels. Importantly, this function of metabolic learning requires not only the mushroom body but also the hypothalamus-like pars intercerebralis, while NF-κB activation in the pars intercerebralis mimics chronic overnutrition in that it causes metabolic learning impairment and disorders. Finally, we evaluate this concept of metabolic learning/memory in mice, suggesting that the hypothalamus is involved in a form of nutritional learning and memory, which is critical for determining resistance or susceptibility to obesity. In conclusion, our data indicate that the brain, and potentially the hypothalamus, direct metabolic learning and the formation of memories, which contribute to the control of systemic metabolic homeostasis.


Brain/metabolism , Energy Intake , Lipid Metabolism , Memory/physiology , Mushroom Bodies/metabolism , NF-kappa B/metabolism , Trehalose/metabolism , Animals , Behavior, Animal , Drosophila , Homeostasis , Hypothalamus/metabolism , Learning/physiology , Mice , Overnutrition/metabolism , Overnutrition/psychology
16.
Nat Immunol ; 16(5): 525-33, 2015 May.
Article En | MEDLINE | ID: mdl-25848866

Innate immunological signals induced by pathogen- and/or damage-associated molecular patterns are essential for adaptive immune responses, but it is unclear if the brain has a role in this process. Here we found that while the abundance of tumor-necrosis factor (TNF) quickly increased in the brain of mice following bacterial infection, intra-brain delivery of TNF mimicked bacterial infection to rapidly increase the number of peripheral lymphocytes, especially in the spleen and fat. Studies of various mouse models revealed that hypothalamic responses to TNF were accountable for this increase in peripheral lymphocytes in response to bacterial infection. Finally, we found that hypothalamic induction of lipolysis mediated the brain's action in promoting this increase in the peripheral adaptive immune response. Thus, the brain-fat axis is important for rapid linkage of innate immunity to adaptive immunity.


Adipose Tissue/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Hypothalamus/immunology , Listeriosis/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/administration & dosage , Adaptive Immunity , Animals , CD4-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/microbiology , Cell Count , Cells, Cultured , Fatty Acids/blood , Hypothalamus/microbiology , Immunity, Innate , Lipolysis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/genetics , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/cerebrospinal fluid
17.
Nat Med ; 20(9): 1001-8, 2014 Sep.
Article En | MEDLINE | ID: mdl-25086906

The brain, in particular the hypothalamus, plays a role in regulating glucose homeostasis; however, it remains unclear whether this organ is causally and etiologically involved in the development of diabetes. Here, we found that hypothalamic transforming growth factor-ß (TGF-ß) production is excessive under conditions of not only obesity but also aging, which are two general etiological factors of type 2 diabetes. Pharmacological and genetic approaches revealed that central TGF-ß excess caused hyperglycemia and glucose intolerance independent of a change in body weight. Further, using cell-specific genetic analyses in vivo, we found that astrocytes and proopiomelanocortin neurons are responsible for the production and prodiabetic effect of central TGF-ß, respectively. Mechanistically, TGF-ß excess induced a hypothalamic RNA stress response, resulting in accelerated mRNA decay of IκBα, an inhibitor of proinflammatory nuclear factor-κB. These results reveal an atypical, mRNA metabolism-driven hypothalamic nuclear factor-κB activation, a mechanism that links obesity as well as aging to hypothalamic inflammation and ultimately to type 2 diabetes.


Aging/physiology , Diabetes Mellitus/physiopathology , Obesity/physiopathology , RNA/metabolism , Stress, Physiological , Transforming Growth Factor beta/metabolism , Astrocytes/metabolism , Diabetes Mellitus/etiology , Diabetes Mellitus/metabolism , Glucose Tolerance Test , Humans , Hypothalamus/physiopathology , NF-kappa B/metabolism , Pro-Opiomelanocortin/metabolism
18.
Mol Metab ; 3(3): 313-24, 2014 Jun.
Article En | MEDLINE | ID: mdl-24749061

Neural stem cells (NSCs) were recently revealed to exist in the hypothalamus of adult mice. Here, following our observation showing that a partial loss of hypothalamic NSCs caused weight gain and glucose intolerance, we studied if NSCs-based cell therapy could be developed to control these disorders. While hypothalamus-implanted NSCs failed to survive in mice with obesity, NF-κB inhibition induced survival and neurogenesis of these cells, leading to effects in counteracting obesity and glucose intolerance. To generate an alternative cell source, we revealed that iPS-derived NSCs were converted into htNSCs by neuropeptide treatment. Of note, obesity condition potentiated the transfer of carotid artery-injected NSCs into the hypothalamus. These iPS-derived cells when engineered with NF-κB inhibition were also effective in reducing obesity and glucose intolerance, and neurogenesis towards POMCergic and GABAergic lineages was accountable. In conclusion, building NSCs in the hypothalamus represents a strategy for controlling obesity and glucose disorders.

19.
Free Radic Biol Med ; 68: 8-21, 2014 Mar.
Article En | MEDLINE | ID: mdl-24296245

Stroke is a leading cause of adult morbidity and mortality with very limited treatment options. Evidence from preclinical models of ischemic stroke has demonstrated that the antioxidant N-acetylcysteine (NAC) effectively protects the brain from ischemic injury. Here, we evaluated a new pathway through which NAC exerted its neuroprotection in a transient cerebral ischemia animal model. Our results demonstrated that pretreatment with NAC increased protein levels of hypoxia-inducible factor-1α (HIF-1α), the regulatable subunit of HIF-1, and its target proteins erythropoietin (EPO) and glucose transporter (GLUT)-3, in the ipsilateral hemispheres of rodents subjected to 90min middle cerebral artery occlusion (MCAO) and 24h reperfusion. Interestingly, after NAC pretreatment and stroke, the contralateral hemisphere also demonstrated increased levels of HIF-1α, EPO, and GLUT-3, but to a lesser extent. Suppressing HIF-1 activity with two widely used pharmacological inhibitors, YC-1 and 2ME2, and specific knockout of neuronal HIF-1α abolished NAC's neuroprotective effects. The results also showed that YC-1 and 2ME2 massively enlarged infarcts, indicating that their toxic effect was larger than just abolishing NAC's neuroprotective effects. Furthermore, we determined the mechanism of NAC-mediated HIF-1α induction. We observed that NAC pretreatment upregulated heat-shock protein 90 (Hsp90) expression and increased the interaction of Hsp90 with HIF-1α in ischemic brains. The enhanced association of Hsp90 with HIF-1α increased HIF-1α stability. Moreover, Hsp90 inhibition attenuated NAC-induced HIF-1α protein accumulation and diminished NAC-induced neuroprotection in the MCAO model. These results strongly indicate that HIF-1 plays an important role in NAC-mediated neuroprotection and provide a new molecular mechanism involved in the antioxidant's neuroprotection in ischemic stroke.


Acetylcysteine/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Infarction, Middle Cerebral Artery/pathology , Stroke/metabolism , Acetylcysteine/metabolism , Animals , Brain Ischemia/metabolism , Brain Ischemia/pathology , Disease Models, Animal , Humans , Infarction, Middle Cerebral Artery/metabolism , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/metabolism , Rats , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Signal Transduction , Stroke/pathology
20.
Sci China Life Sci ; 56(9): 804-10, 2013 Sep.
Article En | MEDLINE | ID: mdl-23864528

Epidemiological evidence and experimental studies suggest that drinking green tea is associated with a lower risk of obesity and related diseases. However, the mechanisms of these effects are not clear. In the present study, we investigated the anti-obesity mechanisms of green tea catechins (GTCs) through modulation of peroxisome proliferator activated-receptor (PPAR) pathways in high-fat diet-induced obesity in rats. GTC supplementation significantly attenuated the increased body and liver weights and the elevated serum and liver triglyceride levels. Meanwhile, GTCs increased the PPARγ levels in subcutaneous white adipose tissue (SWAT) and decreased the PPARγ levels in visceral white adipose tissue (VWAT). In addition, GTC treatment up-regulated the levels of PPARδ in SWAT, VWAT, and brown adipose tissue and increased the expression of genes involved in fatty acid oxidation in brown adipose tissue. Our results suggest that GTCs exert their anti-obesity mechanism in part by modulating PPAR signaling pathways.


Catechin/pharmacology , Obesity/prevention & control , Peroxisome Proliferator-Activated Receptors/drug effects , Tea/chemistry , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Base Sequence , Blotting, Western , DNA Primers , Male , Peroxisome Proliferator-Activated Receptors/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
...