Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Biosci Biotechnol Biochem ; 88(1): 16-25, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-37777845

ABSTRACT

We previously demonstrated that dietary supplementation with Dunaliella tertiolecta (DT) increases uncoupling protein 1 (UCP1) expression in brown adipose tissue (BAT) and improves diet-induced obesity (DIO) in C57BL/6 J mice at thermoneutrality (30 °C). Here, we investigated whether DT improves DIO in a thermoneutral UCP1-deficient (KO) animal. KO mice were fed a high-fat diet supplemented with DT for 12 weeks. Compared to control group without DT, body weight was significantly reduced in DT group with no difference in food intake. Dunaliella tertiolecta-supplemented mice exhibited lower adiposity and well-maintained multilocular morphology in BAT, in which a significant increase in gene expression of PR domain containing 16 was detected in DT group compared to control group. Moreover, increase in UCP2 level and/or decrease in ribosomal protein S6 phosphorylation were detected in adipose tissues of DT group relative to control group. These results suggest that DT supplementation improves DIO by stimulating UCP1-independent energy dissipation at thermoneutrality.


Subject(s)
Energy Metabolism , Obesity , Animals , Mice , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mice, Knockout
2.
Int J Mol Sci ; 24(3)2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36769062

ABSTRACT

We previously demonstrated that the Alzheimer's disease (AD)-like model mice, Tg2576, housed at a high ambient temperature of 30 °C for 13 months, exhibited increased body temperature, which increased amyloid-ß (Aß) levels and tau stability, leading to tau phosphorylation and ultimately inducing memory impairment. Here, we aimed to exclude the possible effect of environmental factors associated with the difference in ambient temperature (23 °C vs. 30 °C) and to further clarify the effects of elevated body temperature on AD-like pathologies. We generated uncoupling protein 1 (UCP1) deletion in Tg2576 mice, Tg2576/UCP1-/-, because UCP1 deletion mice show a sustained rise in body temperature at normal room temperature. As expected, the body temperature in Tg2576/UCP1-/- mice was higher than that in Tg2576/ UCP1+/+ mice at 23 °C, which was accompanied by upregulated Aß levels due to increased ß-secretase (BACE1) and decreased neprilysin (NEP) protein levels in the brains of Tg2576/UCP1-/- mice compared with those in the Tg2576/ UCP1+/+ mice. Elevated body temperature also increased total tau levels, leading to enhanced phosphorylation, heat shock protein induction, and activated tau kinases. Furthermore, elevated body temperature enhanced glial activation and decreased synaptic protein levels in the brain. Taken together, these findings demonstrate that elevated body temperatures exacerbate AD-like pathologies.


Subject(s)
Alzheimer Disease , Mice , Animals , Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/metabolism , Body Temperature , Uncoupling Protein 1/metabolism , Mice, Transgenic , Aspartic Acid Endopeptidases/metabolism , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Brain/metabolism , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Disease Models, Animal
3.
Genes Cells ; 27(3): 202-213, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35007381

ABSTRACT

Thermogenic brown and beige adipocytes express uncoupling protein 1 (UCP1) and stimulate energy metabolism, protecting against obesity and metabolic diseases such as type 2 diabetes and hyperlipidemia. Cellular repressor of E1A-stimulated genes 1 (CREG1) can stimulate thermogenic fat formation, induce UCP1, and reduce diet-induced obesity (DIO) in mice at normal room temperature. In this study, we investigated the effect of CREG1 administration and the importance of UCP1 in DIO inhibition under thermoneutral conditions at 30°C, which attenuate thermogenic fat formation. Interestingly, subcutaneous administration of recombinant CREG1 protein via an osmotic pump in C57BL/6J mice for four weeks increased UCP1 expression in interscapular brown adipose tissue (IBAT), inhibited visceral white fat hypertrophy with partial browning, and reduced DIO compared to that in PBS-treated mice. The mRNA expression of energy metabolism-related genes was significantly increased in the IBAT of CREG1-treated mice compared to that in PBS-treated mice. In contrast, adipocyte-specific overexpression of CREG1 failed to improve DIO in UCP1-knockout mice at thermoneutrality. Our results indicate the therapeutic potential of CREG1 administration for obesity under thermogenic fat-attenuating conditions and highlight the indispensable role of UCP1 in the DIO-inhibitory effect of CREG1.


Subject(s)
Diabetes Mellitus, Type 2 , Adipose Tissue, White/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Diet , Diet, High-Fat/adverse effects , Mice , Mice, Inbred C57BL , Obesity/etiology , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
4.
J Mater Sci Mater Med ; 33(2): 15, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-35072800

ABSTRACT

It was reported by Jung and Day in 2011 that a cotton-like glass fiber pad made of borate glass 13-93B3 demonstrated a remarkable wound healing effect. It was approved for sale as a novel wound dressing in the management of acute and chronic wounds in 2016. However, the detailed mechanism of its wound healing effect has not been reported. In the present study, glass fibers of different composition in the system CaO-B2O3-SiO2 were prepared and their in vitro properties investigated to determine the role of the constituent components in wound healing. Fine glass fibers that were 0.6-2.0 µm in diameter were obtained by a melt blown method. However, these fibers were accompanied by small glass beads because of the low viscosity of the glass melts. 13-93B3 glass released an appreciable amount of borate and calcium ions into simulated body fluid (SBF). The amounts of these released ions decreased with partial replacement of the B2O3 in 13-93B3 with SiO2. The addition of large amounts of the borate and calcium ions into the culture medium decreased the viability of the L929 fibroblasts. Partial replacement of the B2O3 in 13-93B3 with SiO2 induced the formation of an apatite-like phase amenable to the adsorption of biological components on its surface in SBF. The wound healing effect of these glass fibers of different composition is worth examining in future animal experiments.


Subject(s)
Boron Compounds/pharmacology , Calcium Compounds/pharmacology , Fibroblasts/physiology , Glass/chemistry , Oxides/pharmacology , Silicon Dioxide/pharmacology , Animals , Bandages , Boron Compounds/chemistry , Calcium Compounds/chemistry , Cell Line , Cell Survival , Materials Testing , Mice , Microscopy, Electron, Scanning , Oxides/chemistry , Silicon Dioxide/chemistry
5.
J Biochem ; 171(1): 63-73, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34647124

ABSTRACT

Brown and beige adipocytes, which express thermogenic uncoupling protein-1 (UCP1), stimulate glucose and lipid metabolism, improving obesity and metabolic diseases such as type 2 diabetes and hyperlipidemia. Overexpression of cellular repressor of E1A-stimulated genes 1 (CREG1) promotes adipose tissue browning and inhibits diet-induced obesity (DIO) in mice. In this study, we investigated the effects of CREG1 administration on DIO inhibition and adipose browning. Subcutaneous administration of recombinant CREG1 protein to C57BL/6 mice stimulated UCP1 expression in interscapular brown adipose tissue (IBAT) and improved DIO, glucose tolerance and fatty liver compared with those in phosphate-buffered saline-treated mice. Injection of Creg1-expressing adenovirus into inguinal white adipose tissue (IWAT) significantly increased browning and mRNA expression of beige adipocyte marker genes compared with that in mice injected with control virus. The effect of Creg1 induction on beige adipocyte differentiation was supported in primary culture using preadipocytes isolated from IWAT of Creg1-transgenic mice compared with that of wild-type mice. Our results indicate a therapeutic effect of CREG1 on obesity and its associated pathology and a potential of CREG1 to stimulate brown/beige adipocyte formation.


Subject(s)
Diabetes Mellitus, Type 2 , Animals , Diet , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/etiology , Thermogenesis
6.
Biol Proced Online ; 23(1): 11, 2021 Jun 04.
Article in English | MEDLINE | ID: mdl-34088267

ABSTRACT

BACKGROUND: Although retinitis pigmentosa (RP) is most frequently studied in mouse models, rats, rabbits, and pigs are also used as animal models of RP. However, no studies have reported postnatal photoreceptor cell loss before complete development in these models. Here, we generated a transgenic rat strain, named the P347L rat, in which proline at position 347 in the rhodopsin protein was replaced with leucine. RESULTS: A pathological analysis of photoreceptor cells in the P347L rat model was performed, and drugs with potential use as therapeutic agents against RP were investigated. The data clearly showed rapid degeneration and elimination of the outer nuclear layer even before the photoreceptor cells were fully established in P347L rats. To test the usefulness of the P347L rat in the search for new therapeutic agents against RP, the effects of rapamycin on RP were investigated in this rat strain. The findings suggest that rapamycin promotes autophagy and autophagosomal uptake of the rhodopsin that has accumulated abnormally in the cytoplasm, thereby alleviating stress and delaying photoreceptor cell death. CONCLUSIONS: In this RP model, the time to onset of retinal degeneration was less than that of previously reported RP models with other rhodopsin mutations, enabling quicker in vivo evaluation of drug efficacy. Administration of rapamycin delayed the photoreceptor cell degeneration by approximately 1 day.

7.
Nagoya J Med Sci ; 82(3): 487-498, 2020 Aug.
Article in English | MEDLINE | ID: mdl-33132433

ABSTRACT

Impaired wound healing is one of the most common complications of diabetes, and is known to be caused by multiple complicated factors. For instance, impaired angiogenesis, neuropathy, and hyperglycemia all function to delay subsequent wound closure. Alternatively, moist wound healing, which provides an appropriate environment for wounds, was reported to permit rapid healing by managing wound exudate. Accordingly, wound dressing materials that facilitate moist healing have been developed. The present study sought to clarify the effects of wound dressing material for moist healing of diabetic wounds, in terms of the dynamics of angiogenic factors and macrophages, using a mouse model of naturally occurring diabetes. Wounds with full-thickness skin defects were inflicted on the backs of mice and covered with dressing materials of hydrogel or gauze (control), which were retained for 3, 5, 7, 10, or 14 days following wound generation. During this time, the localization of neutrophils, fibroblasts and macrophages as well as the expression of vascular endothelial growth factor (VEGF) in the wounds and surrounding areas was observed each day. Healing clearly occurred in the hydrogel group with an increase in neutrophils and the angiogenic factor, VEGF. Moreover, the use of hydrogel resulted in a rapid rise in M1 macrophages, which appeared in the early stage of the injury, as well as rapid subsequent appearance of M2 macrophages. Thus, herein, we demonstrate that the formation of a moist environment via wound dressing material effectively improves diabetic wound healing.


Subject(s)
Colloids/therapeutic use , Macrophages/drug effects , Macrophages/metabolism , Animals , Diabetes Mellitus/drug therapy , Diabetes Mellitus/metabolism , Immunohistochemistry , Male , Mice , Skin/drug effects , Skin/metabolism , Wound Healing/drug effects
8.
Nutrients ; 12(6)2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32516922

ABSTRACT

We investigated the effect of evodiamine-containing microalga Dunaliella tertiolecta (DT) on the prevention of diet-induced obesity in a thermoneutral C57BL/6J male (30 °C). It attenuates the activity of brown adipose tissue (BAT), which accelerates diet-induced obesity. Nine-week-old mice were fed a high-fat diet supplemented with 10 g (Low group) or 25 g (High group) DT powder per kg food for 12 weeks. Compared to control mice without DT supplementation, body weight gain was significantly reduced in the High group with no difference in food intake. Tissue analyses indicated maintenance of multilocular morphology in BAT and reduced fat deposition in liver in DT-supplemented mice. Molecular analysis showed a significant decrease in mammalian target of rapamycin-ribosomal S6 protein kinase signaling pathway in white adipose tissue and upregulation in mRNA expression of brown fat-associated genes including fibroblast growth factor-21 (Fgf21) and uncoupling protein 1 (Ucp1) in BAT in the High group compared to the control. In the experiments using C3H10T1/2 adipocytes, DT extract upregulated mRNA expression of brown fat-associated genes in dose-dependent and time-dependent manners, accompanied by a significant increase in secreted FGF21 levels. Our data show the ability of DT as a nutraceutical to prevent brown fat attenuation and diet-induced obesity in vivo.


Subject(s)
Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Dietary Supplements , Microalgae/chemistry , Obesity/metabolism , Obesity/prevention & control , Quinazolines/administration & dosage , Quinazolines/pharmacology , Thermogenesis/drug effects , Weight Gain/drug effects , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Fibroblast Growth Factors/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Obesity/etiology , Quinazolines/isolation & purification , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Uncoupling Protein 1/metabolism
9.
Biochim Biophys Acta Mol Basis Dis ; 1866(7): 165762, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32179129

ABSTRACT

Brown adipose tissue (BAT) plays a critical role in lipid metabolism and may protect from hyperlipidemia; however, its beneficial effect appears to depend on the ambient temperature of the environment. In this study, we investigated the effects of uncoupling protein 1 (UCP1) deficiency on lipid metabolism, including the pathophysiology of hyperlipidemia, in apolipoprotein E knockout (APOE-KO) mice at a normal (23 °C) and thermoneutral (30 °C) temperature. Unexpectedly, UCP1 deficiency caused improvements in hyperlipidemia, atherosclerosis, and glucose metabolism, regardless of an increase in hepatic lipid deposition, in Ucp1/Apoe double-knockout (DKO) mice fed a high-fat diet at 23 °C, with BAT hyperplasia and robust browning of inguinal white adipose tissue (IWAT) observed. Proteomics and gene expression analyses revealed significant increases in many proteins involved in energy metabolism and strong upregulation of brown/beige adipocyte-related genes and fatty acid metabolism-related genes in browned IWAT, suggesting an induction of beige fat formation and stimulation of lipid metabolism in DKO mice at 23 °C. Conversely, mRNA levels of fatty acid oxidation-related genes decreased in the liver of DKO mice. The favorable phenotypic changes were lost at 30 °C, with BAT whitening and disappearance of IWAT browning, while fatty liver further deteriorated in DKO mice compared with that in APOE-KO mice. Finally, longevity analysis revealed a significant lifespan extension of DKO mice compared with that of APOE-KO mice at 23 °C. Irrespective of the fundamental role of UCP1 thermogenesis, our results highlight the importance of beige fat for the improvement of hyperlipidemia and longevity under the atherogenic status at normal room temperature.


Subject(s)
Apolipoproteins E/genetics , Fatty Liver/genetics , Hyperlipidemias/genetics , Uncoupling Protein 1/genetics , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White , Animals , Energy Metabolism/genetics , Fatty Acids/genetics , Fatty Acids/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation , Humans , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Lipid Metabolism/genetics , Mice , Mice, Knockout, ApoE , Proteomics , Thermogenesis/genetics
10.
FASEB J ; 33(7): 8069-8082, 2019 07.
Article in English | MEDLINE | ID: mdl-30917000

ABSTRACT

Increased formation of brown and beige adipocytes is critical for adaptive thermogenesis to maintain homeothermy in cold or to circumvent diet-induced obesity (DIO). Cellular repressor of adenovirus early region 1A-stimulated genes 1 (CREG1) exhibits the ability to stimulate brown adipogenesis, including the induction of uncoupling protein 1 (UCP1), in vitro. Thus, we aimed to clarify whether CREG1 promotes brown adipocyte formation and inhibits DIO at the whole-animal level. In mouse brown adipose tissue (BAT), CREG1 expression was markedly increased in cold but was decreased under thermoneutrality, suggesting CREG1 involvement in BAT thermogenesis. Moreover, in BAT and white adipose tissue, expression of UCP1 and fibroblast growth factor-21 and browning were both significantly higher in adipocyte P2-Creg1-transgenic (Tg) mice than in wild-type (WT) littermates. Following stimulation with a ß3-adrenergic agonist, energy consumption was elevated in the Tg mice, which showed increased resistance to DIO and improvement of obesity-associated complications including fatty liver relative to WT mice. The CREG1 stimulatory effect on brown adipogenesis was confirmed in Tg-BAT primary cultures. It was also found that CREG1 binds to retinoid X receptor α, which interacts with thyroid hormone receptor for brown adipogenesis. Our findings demonstrate that CREG1 stimulates brown adipocyte formation and browning, ameliorating obesity and its related pathology in vivo.-Hashimoto, M., Kusudo, T., Takeuchi, T., Kataoka, N., Mukai, T., Yamashita, H. CREG1 stimulates brown adipocyte formation and ameliorates diet-induced obesity in mice.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Diet, High-Fat/adverse effects , Obesity/metabolism , Repressor Proteins/metabolism , Adipocytes, Brown/pathology , Adipose Tissue, Brown/pathology , Animals , Mice , Mice, Knockout , Mice, Transgenic , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Repressor Proteins/genetics , Thermogenesis , Uncoupling Protein 1/biosynthesis
11.
FEBS Open Bio ; 7(7): 1009-1016, 2017 07.
Article in English | MEDLINE | ID: mdl-28680813

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is increasing in prevalence worldwide and has been identified as a risk factor for cirrhosis and hepatocellular carcinoma. However, there is no effective pharmacologic treatment for NAFLD. FABP1 is a liver-specific fatty acid-binding protein (FABP) that plays important roles in intracellular lipid metabolism in the liver. We investigated the effect of repression of FABP1 expression on NAFLD, using adenovirus-mediated silencing of FABP1. FABP1 knockdown in the liver decreased the liver weight and hepatic triglyceride (TG) accumulation. The expression of inflammatory and oxidative stress markers in the liver was also reduced. The level of thiobarbituric acid-reactive substances, a marker of lipid peroxidation, in the liver of FABP1 knockdown mice was significantly decreased. These results suggest that FABP1 reduction in the liver is an effective approach against NAFLD.

12.
PLoS One ; 8(12): e83264, 2013.
Article in English | MEDLINE | ID: mdl-24391749

ABSTRACT

Evodiamine, an alkaloid extracted from the dried unripe fruit of the tree Evodia rutaecarpa Bentham (Rutaceae), reduces obesity and insulin resistance in obese/diabetic mice; however, the mechanism underlying the effect of evodiamine on insulin resistance is unknown. This study investigated the effect of evodiamine on signal transduction relating to insulin resistance using obese/diabetic KK-Ay mice and an in vitro adipocyte culture. There is a significant decrease in the mammalian target of rapamycin (mTOR) and ribosomal S6 protein kinase (S6K) signaling in white adipose tissue (WAT) in KK-Ay mice treated with evodiamine, in which glucose tolerance is improved. In addition, reduction of insulin receptor substrate 1 (IRS1) serine phosphorylation, an indicator of insulin resistance, was detected in their WAT, suggesting suppression of the negative feedback loop from S6K to IRS1. As well as the stimulation of IRS1 and Akt serine phosphorylation, insulin-stimulated phosphorylation of mTOR and S6K is time-dependent in 3T3-L1 adipocytes, whereas evodiamine does not affect their phosphorylation except for an inhibitory effect on mTOR phosphorylation. Moreover, evodiamine inhibits the insulin-stimulated phosphorylation of mTOR and S6K, leading to down-regulation of IRS1 serine phosphorylation in the adipocytes. Evodiamine also stimulates phosphorylation of AMP-activated protein kinase (AMPK), an important regulator of energy metabolism, which may cause down-regulation of mTOR signaling in adipocytes. A similar effect on AMPK, mTOR and IRS1 phosphorylation was found in adipocytes treated with rosiglitazone. These results suggest evodiamine improves glucose tolerance and prevents the progress of insulin resistance associated with obese/diabetic states, at least in part, through inhibition of mTOR-S6K signaling and IRS1 serine phosphorylation in adipocytes.


Subject(s)
Insulin Receptor Substrate Proteins/antagonists & inhibitors , Insulin Resistance , Quinazolines/pharmacology , Ribosomal Protein S6 Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Evodia/chemistry , Female , Glucose/metabolism , Glucose Tolerance Test , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/chemistry , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Obese , Phosphorylation/drug effects , Serine/chemistry , Signal Transduction/drug effects
13.
Neurosci Res ; 65(4): 384-92, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19737583

ABSTRACT

We previously reported that implantation of dendritic cells (DCs) into the injured site activates neural stem/progenitor cells (NSPCs) and promotes functional recovery after spinal cord injury (SCI) in mice. Working toward clinical application of DC therapy for SCI, we analyzed whether DCs promote functional recovery after SCI in a non-human primate, the common marmoset (CM). CMs are usually born as dizygotic twins. They are thus natural bone marrow and peripheral blood chimeras due to sharing of the placental circulation between dizygotic twins, leading to functional immune tolerance. In this study, to identify adequate CM donor-and-host pairs, mixed leukocyte reaction (MLR) assays were performed. Then, CM-DCs were generated from the bone marrow of the twin selected to be donor and transplanted into the injured site of the spinal cord of the other twin selected to be host, 7 days after injury. Histological analyses revealed fewer areas of demyelination around the injured site in DC-treated CMs than in controls. Immunohistochemical analysis showed that more motor neurons and corticospinal tracts were preserved after SCI in DC-treated CMs. Motor functions were evaluated using three different behavior tests and earlier functional recovery was observed in DC-treated CMs. These results suggest DC therapy to possibly be beneficial in primates with SCI and that this treatment has potential for clinical application.


Subject(s)
Dendritic Cells/transplantation , Recovery of Function , Spinal Cord Injuries/surgery , Animals , Callithrix , Flow Cytometry , Magnetic Resonance Imaging , Microscopy, Electron, Transmission , Motor Activity/physiology , Nerve Regeneration/physiology , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology
14.
J Neurosci ; 27(22): 5903-14, 2007 May 30.
Article in English | MEDLINE | ID: mdl-17537961

ABSTRACT

Loss-of-function mutations in human SCN1A gene encoding Nav1.1 are associated with a severe epileptic disorder known as severe myoclonic epilepsy in infancy. Here, we generated and characterized a knock-in mouse line with a loss-of-function nonsense mutation in the Scn1a gene. Both homozygous and heterozygous knock-in mice developed epileptic seizures within the first postnatal month. Immunohistochemical analyses revealed that, in the developing neocortex, Nav1.1 was clustered predominantly at the axon initial segments of parvalbumin-positive (PV) interneurons. In heterozygous knock-in mice, trains of evoked action potentials in these fast-spiking, inhibitory cells exhibited pronounced spike amplitude decrement late in the burst. Our data indicate that Nav1.1 plays critical roles in the spike output from PV interneurons and, furthermore, that the specifically altered function of these inhibitory circuits may contribute to epileptic seizures in the mice.


Subject(s)
Axons/chemistry , Epilepsy/genetics , Interneurons/chemistry , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Inhibition , Parvalbumins/biosynthesis , Sodium Channels/genetics , Sodium Channels/metabolism , Action Potentials/genetics , Animals , Axons/metabolism , Cell Line , Epilepsy/metabolism , Humans , Interneurons/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Mutant Strains , NAV1.1 Voltage-Gated Sodium Channel , Nerve Net/chemistry , Nerve Net/metabolism , Nerve Tissue Proteins/physiology , Neural Inhibition/genetics , Sodium Channels/physiology
15.
Hum Mol Genet ; 15(18): 2752-62, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16891409

ABSTRACT

Trisomy 21 or Down syndrome (DS) is the most common genetic birth defect associated with mental retardation. The over-expression of genes on chromosome 21, including SOD1 (Cu/Zn superoxide dismutase) and APP (amyloid-beta precursor protein) is believed to underlie the increased oxidative stress and neurodegeneration commonly described in DS. However, a segmental trisomy 16 mouse model for DS, Ts1Cje, has a subset of triplicated human chromosome 21 gene orthologs that exclude APP and SOD1. Here, we report that Ts1Cje brain shows decreases of mitochondrial membrane potential and ATP production, increases of reactive oxygen species, hyperphosphorylation of tau without NFT formation, increase of GSK3beta and JNK/SAPK activities and unaltered AbetaPP metabolism. Our findings suggest that genes on the trisomic Ts1Cje segment other than APP and SOD1 can cause oxidative stress, mitochondrial dysfunction and hyperphosphorylation of tau, all of which may play critical roles in the pathogenesis of mental retardation in DS.


Subject(s)
Down Syndrome/genetics , Down Syndrome/metabolism , Mitochondria/metabolism , tau Proteins/metabolism , Adenosine Triphosphate/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Down Syndrome/complications , Down Syndrome/pathology , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/metabolism , Humans , Intellectual Disability/etiology , Intellectual Disability/genetics , Intellectual Disability/metabolism , MAP Kinase Signaling System , Male , Membrane Potentials , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Neurofibrillary Tangles/pathology , Oxidative Stress , Phosphorylation , Trisomy , tau Proteins/chemistry
16.
Nat Genet ; 36(8): 842-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258581

ABSTRACT

Juvenile myoclonic epilepsy (JME) is the most frequent cause of hereditary grand mal seizures. We previously mapped and narrowed a region associated with JME on chromosome 6p12-p11 (EJM1). Here, we describe a new gene in this region, EFHC1, which encodes a protein with an EF-hand motif. Mutation analyses identified five missense mutations in EFHC1 that cosegregated with epilepsy or EEG polyspike wave in affected members of six unrelated families with JME and did not occur in 382 control individuals. Overexpression of EFHC1 in mouse hippocampal primary culture neurons induced apoptosis that was significantly lowered by the mutations. Apoptosis was specifically suppressed by SNX-482, an antagonist of R-type voltage-dependent Ca(2+) channel (Ca(v)2.3). EFHC1 and Ca(v)2.3 immunomaterials overlapped in mouse brain, and EFHC1 coimmunoprecipitated with the Ca(v)2.3 C terminus. In patch-clamp analysis, EFHC1 specifically increased R-type Ca(2+) currents that were reversed by the mutations associated with JME.


Subject(s)
Myoclonic Epilepsy, Juvenile/genetics , Animals , Apoptosis/genetics , Calcium-Binding Proteins/genetics , Cells, Cultured , Humans , Mice , Molecular Sequence Data , Mutation, Missense , Pedigree
SELECTION OF CITATIONS
SEARCH DETAIL