Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 298(6): 101936, 2022 06.
Article in English | MEDLINE | ID: mdl-35430252

ABSTRACT

Valosin-containing protein (VCP) is a member of AAA-ATPase superfamily involved in various cellular functions. To investigate the pathophysiological role of VCP in metabolic disorders, we generated knock-in mice bearing an A232E mutation in VCP, a known human VCP pathogenic variant. When heterozygous mutant mice (A232E/+) were fed a high-fat diet, we observed that fatty liver was ameliorated and the proteolytic processing of the transcription factor sterol regulatory element-binding protein 1 (SREBP1) was impaired. Further co-immunoprecipitation analysis in wildtype mice revealed interactions of VCP with SREBP1 and a rhomboid protease, RHBDL4, in the liver, and these interactions were attenuated in A232E/+ mice. Consistent with these results, we show that knockdown or chemical inhibition of VCP or RHBDL4 in human hepatocytes impaired the proteolytic processing of SREBP1. Finally, we found that knockdown of E3 ligases such as glycoprotein 78 and HMG-CoA reductase degradation protein 1 disrupted the interaction of VCP with SREBP1 and impaired the proteolytic processing of SREBP1. These results suggest that VCP recognizes ubiquitinylated SREBP1 and recruits it to RHBDL4 to promote its proteolytic processing. The present study reveals a novel proteolytic processing pathway of SREBP1 and may lead to development of new therapeutic strategies to treat fatty liver diseases.


Subject(s)
Membrane Proteins , Sterol Regulatory Element Binding Protein 1 , Valosin Containing Protein , Adenosine Triphosphatases/metabolism , Animals , Membrane Proteins/metabolism , Mice , Non-alcoholic Fatty Liver Disease/physiopathology , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Valosin Containing Protein/genetics , Valosin Containing Protein/metabolism
2.
Sci Rep ; 12(1): 449, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35013417

ABSTRACT

Therapeutic approach for NAFLD is limited and there are no approved drugs. Pioglitazone (PGZ), a thiazolidinedione (TZD) that acts via peroxisome proliferator activated receptor gamma (PPARγ) is the only agent that has shown consistent benefit and efficacy in clinical trials. However, the mechanism of its therapeutic effect on NAFLD remains unclear. The poor understanding may be due to problems with mouse, a species most used for animal experiments. TZDs exacerbate fatty liver in mouse models while they improve it in rat models like in human patients. Therefore, we compared the effects of TZDs including PGZ and rosiglitazone (RGZ) in ob/ob mice and Lepmkyo/Lepmkyo rats, models of leptin-deficient obesity, and A-ZIP/F-1 mice and seipin knockout (SKO) rats, models of generalized lipodystrophy. Pparg mRNA expression was markedly upregulated in fatty livers of mouse models while it was unchanged in rat models. TZDs exacerbated fatty liver in ob/ob and A-ZIP/F-1 mice, improved it in Lepmkyo/Lepmkyo rats and showed no effect in SKO rats. Gene expression analyses of Pparg and its target gene, Fsp27 revealed that PPARγ in the adipose tissue is the exclusive therapeutic target of TZDs in rats but PPARγ in the liver in addition to the adipose tissue is also a major site of actions for TZDs in mice. Although the response to TZDs in mice is the complete opposite of that in human patients, no report has pointed out the problem with TZD studies using mouse models so far. The present study might provide useful suggestions in research on TZDs.


Subject(s)
Fatty Liver/drug therapy , Lipid Metabolism/drug effects , PPAR gamma/metabolism , Pioglitazone/therapeutic use , Thiazolidinediones/therapeutic use , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Fatty Liver/etiology , Fatty Liver/metabolism , Leptin/deficiency , Lipodystrophy/complications , Male , Mice, Inbred C57BL , Obesity/complications , PPAR gamma/agonists , Pioglitazone/pharmacology , Rats, Transgenic , Thiazolidinediones/pharmacology
3.
Sci Rep ; 11(1): 17691, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489483

ABSTRACT

Leptin is an adipocyte-derived hormone that regulates appetite and energy expenditure via the hypothalamus. Since the majority of obese subjects are leptin resistant, leptin sensitizers, rather than leptin itself, are expected to be anti-obesity drugs. Endoplasmic reticulum (ER) stress in the hypothalamus plays a key role in the pathogenesis of leptin resistance. ATP-deficient cells are vulnerable to ER stress and ATP treatment protects cells against ER stress. Thus, we investigated the therapeutic effects of oral 1,3-butanediol (BD) administration, which increases plasma ß-hydroxybutyrate and hypothalamic ATP concentrations, in diet induced obese (DIO) mice with leptin resistance. BD treatment effectively decreased food intake and body weight in DIO mice. In contrast, BD treatment had no effect in leptin deficient ob/ob mice. Co-administration experiment demonstrated that BD treatment sensitizes leptin action in both DIO and ob/ob mice. We also demonstrated that BD treatment attenuates ER stress and leptin resistance at the hypothalamus level. This is the first report to confirm the leptin sensitizing effect of BD treatment in leptin resistant DIO mice. The present study provides collateral evidence suggesting that the effect of BD treatment is mediated by the elevation of hypothalamic ATP concentration. Ketone bodies and hypothalamic ATP are the potential target for the treatment of obesity and its complications.


Subject(s)
Body Weight/drug effects , Butylene Glycols/pharmacology , Endoplasmic Reticulum Stress/drug effects , Hypothalamus/drug effects , Leptin/pharmacology , Obesity/drug therapy , 3-Hydroxybutyric Acid/metabolism , Adenosine Triphosphate/metabolism , Animals , Butylene Glycols/therapeutic use , Energy Metabolism/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Obese , Obesity/metabolism
4.
Diabetes ; 69(11): 2352-2363, 2020 11.
Article in English | MEDLINE | ID: mdl-32796082

ABSTRACT

Inhibitors of 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), statins, which are used to prevent cardiovascular diseases, are associated with a modest increase in the risk of new-onset diabetes. To investigate the role of HMGCR in the development of ß-cells and glucose homeostasis, we deleted Hmgcr in a ß-cell-specific manner by using the Cre-loxP technique. Mice lacking Hmgcr in ß-cells (ß-KO) exhibited hypoinsulinemic hyperglycemia as early as postnatal day 9 (P9) due to decreases in both ß-cell mass and insulin secretion. Ki67-positive cells were reduced in ß-KO mice at P9; thus, ß-cell mass reduction was caused by proliferation disorder immediately after birth. The mRNA expression of neurogenin3 (Ngn3), which is transiently expressed in endocrine progenitors of the embryonic pancreas, was maintained despite a striking reduction in the expression of ß-cell-associated genes, such as insulin, pancreatic and duodenal homeobox 1 (Pdx1), and MAF BZIP transcription factor A (Mafa) in the islets from ß-KO mice. Histological analyses revealed dysmorphic islets with markedly reduced numbers of ß-cells, some of which were also positive for glucagon. In conclusion, HMGCR plays critical roles not only in insulin secretion but also in the development of ß-cells in mice.


Subject(s)
Gene Expression Regulation, Enzymologic/physiology , Hydroxymethylglutaryl CoA Reductases/metabolism , Insulin-Secreting Cells/enzymology , Insulin/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Glucose , Diabetes Mellitus , Feeding Behavior , Glucose Tolerance Test , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Hyperglycemia , Insulin/blood , Insulin-Secreting Cells/metabolism , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
5.
Diabetes ; 69(2): 158-164, 2020 02.
Article in English | MEDLINE | ID: mdl-31690648

ABSTRACT

Adipose tissue macrophages (ATMs) are involved in the development of insulin resistance in obesity. We have recently shown that myeloid cell-specific reduction of HMG-CoA reductase (Hmgcr m-/m- ), which is the rate-limiting enzyme in cholesterol biosynthesis, protects against atherosclerosis by inhibiting macrophage migration in mice. We hypothesized that ATMs are harder to accumulate in Hmgcr m-/m- mice than in control Hmgcr fl/fl mice in the setting of obesity. To test this hypothesis, we fed Hmgcr m-/m- and Hmgcr fl/fl mice a high-fat diet (HFD) for 24 weeks and compared plasma glucose metabolism as well as insulin signaling and histology between the two groups. Myeloid cell-specific reduction of Hmgcr improved glucose tolerance and insulin sensitivity without altering body weight in the HFD-induced obese mice. The improvement was due to a decrease in the number of ATMs. The ATMs were reduced by decreased recruitment of macrophages as a result of their impaired chemotactic activity. These changes were associated with decreased expression of proinflammatory cytokines in adipose tissues. Myeloid cell-specific reduction of Hmgcr also attenuated hepatic steatosis. In conclusion, reducing myeloid HMGCR may be a promising strategy to improve insulin resistance and hepatic steatosis in obesity.


Subject(s)
Adipose Tissue/drug effects , Hydroxymethylglutaryl CoA Reductases/metabolism , Inflammation/metabolism , Insulin Resistance , Myeloid Cells/metabolism , Obesity/chemically induced , Adipose Tissue/pathology , Animals , Blood Glucose , Diet, High-Fat/adverse effects , Fatty Liver/chemically induced , Gene Expression Regulation, Enzymologic , Hydroxymethylglutaryl CoA Reductases/genetics , Inflammation/chemically induced , Insulin/blood , Macrophages , Mice , Mice, Knockout
6.
Diabetes ; 65(10): 2954-65, 2016 10.
Article in English | MEDLINE | ID: mdl-27381370

ABSTRACT

Agonist-induced activation of peroxisome proliferator-activated receptor-γ (PPARγ) stimulates adipocyte differentiation and insulin sensitivity. Patients with heterozygous PPARγ dominant-negative mutation develop partial lipodystrophy and insulin resistance. Inconsistent with this evidence in humans, it was reported that heterozygous PPARγ knockout mice have increased insulin sensitivity and that mice with heterozygous PPARγ dominant-negative mutation have normal insulin sensitivity and improved glucose tolerance. In the context of the interspecies intranslatability of PPARγ-related findings, we generated a PPARγ mutant rat with a loss-of-function mutation (Pparg(mkyo)) without dominant-negative activity by using the ENU (N-ethyl-N-nitrosourea) mutagenesis method. Heterozygous Pparg(mkyo/+) rats showed reduced fat mass with adipocyte hypertrophy and insulin resistance, which were highly predictable from known actions of PPARγ agonists and phenotypes of patients with the PPARγ mutation. This report is the first in our knowledge to clearly demonstrate that both alleles of PPARγ are required for normal adipocyte development and insulin sensitivity in vivo. Furthermore, the study indicates that PPARγ regulates mainly adipocyte number rather than adipocyte size in vivo. The choice of appropriate species as experimental models is critical, especially for the study of PPARγ.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , PPAR gamma/metabolism , Adipose Tissue/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Animals, Genetically Modified , Blood Glucose/drug effects , Body Composition/drug effects , Body Composition/genetics , Cell Count , Cell Size/drug effects , Chromatin Immunoprecipitation , Heterozygote , Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , Lipodystrophy/genetics , Lipodystrophy/metabolism , Male , Mutation/genetics , PPAR gamma/genetics , Pioglitazone , Rats , Thiazolidinediones/pharmacology
7.
Hum Mol Genet ; 24(15): 4238-49, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25934999

ABSTRACT

Seipin, encoded by BSCL2 gene, is a protein whose physiological functions remain unclear. Mutations of BSCL2 cause the most-severe form of congenital generalized lipodystrophy (CGL). BSCL2 mRNA is highly expressed in the brain and testis in addition to the adipose tissue in human, suggesting physiological roles of seipin in non-adipose tissues. Since we found BSCL2 mRNA expression pattern among organs in rat is similar to human while it is not highly expressed in mouse brain, we generated a Bscl2/seipin knockout (SKO) rat using the method with ENU (N-ethyl-N-nitrosourea) mutagenesis. SKO rats showed total lack of white adipose tissues including mechanical fat such as bone marrow and retro-orbital fats, while physiologically functional brown adipose tissue was preserved. Besides the lipodystrophic phenotypes, SKO rats showed impairment of spatial working memory with brain weight reduction and infertility with azoospermia. We confirmed reduction of brain volume and number of sperm in human patients with BSCL2 mutation. This is the first report demonstrating that seipin is necessary for normal brain development and spermatogenesis in addition to white adipose tissue development.


Subject(s)
Adipogenesis/genetics , Brain/growth & development , GTP-Binding Protein gamma Subunits/genetics , Spermatogenesis/genetics , Animals , Brain/metabolism , GTP-Binding Protein gamma Subunits/biosynthesis , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Humans , Male , Mice , RNA, Messenger/biosynthesis , Rats , Spermatozoa/growth & development , Spermatozoa/metabolism , Testis/growth & development , Testis/metabolism
8.
Am J Physiol Endocrinol Metab ; 307(8): E712-9, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25159327

ABSTRACT

Leptin may reduce pancreatic lipid deposition, which increases with progression of obesity and can impair ß-cell function. The insulinotropic effect of glucagon-like peptide-1 (GLP-1) and the efficacy of GLP-1 receptor agonist are reduced associated with impaired ß-cell function. In this study, we examined whether leptin could restore the efficacy of exenatide, a GLP-1 receptor agonist, in type 2 diabetes with increased adiposity. We chronically administered leptin (500 µg·kg⁻¹·day⁻¹) and/or exenatide (20 µg·kg⁻¹·day⁻¹) for 2 wk in a mouse model of type 2 diabetes with increased adiposity induced by streptozotocin and high-fat diet (STZ/HFD mice). The STZ/HFD mice exhibited hyperglycemia, overweight, increased pancreatic triglyceride level, and reduced glucose-stimulated insulin secretion (GSIS); moreover, the insulinotropic effect of exenatide was reduced. However, leptin significantly reduced pancreatic triglyceride level, and adding leptin to exenatide (LEP/EX) remarkably enhanced GSIS. These results suggested that the leptin treatment restored the insulinotropic effect of exenatide in the mice. In addition, LEP/EX reduced food intake, body weight, and triglyceride levels in the skeletal muscle and liver, and corrected hyperglycemia to a greater extent than either monotherapy. The pair-feeding experiment indicated that the marked reduction of pancreatic triglyceride level and enhancement of GSIS by LEP/EX occurred via mechanisms other than calorie restriction. These results suggest that leptin treatment may restore the insulinotropic effect of exenatide associated with the reduction of pancreatic lipid deposition in type 2 diabetes with increased adiposity. Combination therapy with leptin and exenatide could be an effective treatment for patients with type 2 diabetes with increased adiposity.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Hypoglycemic Agents/therapeutic use , Leptin/therapeutic use , Overweight/complications , Pancreas/drug effects , Peptides/therapeutic use , Venoms/therapeutic use , Adiposity/drug effects , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat/adverse effects , Drug Implants , Drug Synergism , Drug Therapy, Combination , Exenatide , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/metabolism , Hyperglycemia/prevention & control , Hypoglycemic Agents/administration & dosage , Insulin/metabolism , Insulin Secretion , Leptin/administration & dosage , Leptin/genetics , Male , Mice, Inbred C57BL , Overweight/drug therapy , Overweight/etiology , Overweight/metabolism , Pancreas/metabolism , Peptides/administration & dosage , Recombinant Proteins/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/therapeutic use , Streptozocin , Triglycerides/metabolism , Venoms/administration & dosage
9.
Physiol Genomics ; 45(17): 786-93, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-23800849

ABSTRACT

Leptin is one of the key molecules in maintaining energy homeostasis. Although genetically leptin-deficient Lep(ob)/Lep(ob) mice have greatly contributed to elucidating leptin physiology, the use of more than one species can improve the accuracy of analysis results. Using the N-ethyl-N-nitrosourea mutagenesis method, we generated a leptin-deficient Lep(mkyo)/Lep(mkyo) rat that had a nonsense mutation (Q92X) in leptin gene. Lep(mkyo)/Lep(mkyo) rats showed obese phenotypes including severe fatty liver, which were comparable to Lep(ob)/Lep(ob) mice. To identify genes that respond to leptin in the liver, we performed microarray analysis with Lep(mkyo)/Lep(mkyo) rats and Lep(ob)/Lep(ob) mice. We sorted out genes whose expression levels in the liver of Lep(mkyo)/Lep(mkyo) rats were changed from wild-type (WT) rats and were reversed toward WT rats by leptin administration. In this analysis, livers were sampled for 6 h, a relatively short time after leptin administration to avoid the secondary effect of metabolic changes such as improvement of fatty liver. We did the same procedure in Lep(ob)/Lep(ob) mice and selected genes whose expression patterns were common in rat and mouse. We verified their gene expressions by real-time quantitative PCR. Finally, we identified eight genes that primarily respond to leptin in the liver commonly in rat and mouse. These genes might be important for the effect of leptin in the liver.


Subject(s)
Gene Expression , Leptin/genetics , Liver/physiology , Obesity/genetics , Rats, Mutant Strains/genetics , Animals , Codon, Nonsense , Disease Models, Animal , Ethylnitrosourea/toxicity , Fatty Liver/genetics , Fatty Liver/pathology , Leptin/blood , Leptin/deficiency , Leptin/pharmacology , Lipid Metabolism/genetics , Liver/drug effects , Male , Mice, Mutant Strains , Mutagenesis , Real-Time Polymerase Chain Reaction
10.
J Clin Endocrinol Metab ; 97(10): 3663-71, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22872692

ABSTRACT

CONTEXT: Lipodystrophy is a disease characterized by a paucity of adipose tissue and low circulating concentrations of adipocyte-derived leptin. Leptin-replacement therapy improves eating and metabolic disorders in patients with lipodystrophy. OBJECTIVE: The aim of the study was to clarify the pathogenic mechanism of eating disorders in lipodystrophic patients and the action mechanism of leptin on appetite regulation. SUBJECTS AND INTERVENTIONS: We investigated food-related neural activity using functional magnetic resonance imaging in lipodystrophic patients with or without leptin replacement therapy and in healthy controls. We also measured the subjective feelings of appetite. RESULTS: Although there was little difference in the enhancement of neural activity by food stimuli between patients and controls under fasting, postprandial suppression of neural activity was insufficient in many regions of interest including amygdala, insula, nucleus accumbens, caudate, putamen, and globus pallidus in patients when compared with controls. Leptin treatment effectively suppressed postprandial neural activity in many of these regions of interest, whereas it showed little effect under fasting in patients. Consistent with these results, postprandial formation of satiety feeling was insufficient in patients when compared with controls, which was effectively reinforced by leptin treatment. CONCLUSIONS: This study demonstrated the insufficiency of postprandial suppression of food-related neural activity and formation of satiety feeling in lipodystrophic patients, which was effectively restored by leptin. The findings in this study emphasize the important pathological role of leptin in eating disorders in lipodystrophy and provide a clue to understanding the action mechanism of leptin in human, which may lead to development of novel strategies for prevention and treatment of obesity.


Subject(s)
Leptin/administration & dosage , Leptin/physiology , Lipodystrophy/drug therapy , Lipodystrophy/physiopathology , Satiety Response/drug effects , Satiety Response/physiology , Adult , Amygdala/physiology , Appetite/drug effects , Appetite/physiology , Cerebral Cortex/physiology , Corpus Striatum/physiology , Eating/drug effects , Eating/physiology , Fasting/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Female , Humans , Leptin/deficiency , Magnetic Resonance Imaging , Male , Nucleus Accumbens/physiology , Postprandial Period/physiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...