44 results on '"Sudha B. Biddinger"'
Search Results
2. Hepatocyte ABCA1 Deletion Impairs Liver Insulin Signaling and Lipogenesis
- Author
-
Chia-Chi C. Key, Mingxia Liu, C. Lisa Kurtz, Soonkyu Chung, Elena Boudyguina, Timothy A. Dinh, Alexander Bashore, Peter E. Phelan, Barry I. Freedman, Timothy F. Osborne, Xuewei Zhu, Lijun Ma, Praveen Sethupathy, Sudha B. Biddinger, and John S. Parks
- Subjects
hepatosteatosis ,cholesterol ,lipid raft ,vesicle trafficking ,selective insulin resistance ,mTORC ,plasma membrane ,hepatocyte ,Western-type diet ,Biology (General) ,QH301-705.5 - Abstract
Plasma membrane (PM) free cholesterol (FC) is emerging as an important modulator of signal transduction. Here, we show that hepatocyte-specific knockout (HSKO) of the cellular FC exporter, ATP-binding cassette transporter A1 (ABCA1), leads to decreased PM FC content and defective trafficking of lysosomal FC to the PM. Compared with controls, chow-fed HSKO mice had reduced hepatic (1) insulin-stimulated Akt phosphorylation, (2) activation of the lipogenic transcription factor Sterol Regulatory Element Binding Protein (SREBP)-1c, and (3) lipogenic gene expression. Consequently, Western-type diet-fed HSKO mice were protected from steatosis. Surprisingly, HSKO mice had intact glucose metabolism; they showed normal gluconeogenic gene suppression in response to re-feeding and normal glucose and insulin tolerance. We conclude that: (1) ABCA1 maintains optimal hepatocyte PM FC, through intracellular FC trafficking, for efficient insulin signaling; and (2) hepatocyte ABCA1 deletion produces a form of selective insulin resistance so that lipogenesis is suppressed but glucose metabolism remains normal.
- Published
- 2017
- Full Text
- View/download PDF
3. Hepatic insulin receptor deficiency impairs the SREBP-2 response to feeding and statins[S]
- Author
-
Ji Miao, Joel T. Haas, Praveen Manthena, Yanning Wang, Enpeng Zhao, Bhavapriya Vaitheesvaran, Irwin J. Kurland, and Sudha B. Biddinger
- Subjects
hepatic insulin signaling ,cholesterol biosynthesis ,sterol regulatory element binding protein ,liver insulin receptor knockout ,Biochemistry ,QD415-436 - Abstract
The liver plays a central role in metabolism and mediating insulin action. To dissect the effects of insulin on the liver in vivo, we have studied liver insulin receptor knockout (LIRKO) mice. Because LIRKO livers lack insulin receptors, they are unable to respond to insulin. Surprisingly, the most profound derangement observed in LIRKO livers by microarray analysis is a suppression of the cholesterologenic genes. Sterol regulatory element binding protein (SREBP)-2 promotes cholesterologenic gene transcription, and is inhibited by intracellular cholesterol. LIRKO livers show a slight increase in hepatic cholesterol, a 40% decrease in Srebp-2, and a 50–90% decrease in the cholesterologenic genes at the mRNA and protein levels. In control mice, SREBP-2 and cholesterologenic gene expression are suppressed by fasting and restored by refeeding; in LIRKO mice, this response is abolished. Similarly, the ability of statins to induce Srebp-2 and the cholesterologenic genes is lost in LIRKO livers. In contrast, ezetimibe treatment robustly induces Srepb-2 and its targets in LIRKO livers, raising the possibility that insulin may regulate SREBP-2 indirectly, by altering the accumulation or distribution of cholesterol within the hepatocyte. Taken together, these data indicate that cholesterol synthesis is a key target of insulin action in the liver.
- Published
- 2014
- Full Text
- View/download PDF
4. Evaluating human genetic support for hypothesized metabolic disease genes
- Author
-
Peter, Dornbos, Preeti, Singh, Dong-Keun, Jang, Anubha, Mahajan, Sudha B, Biddinger, Jerome I, Rotter, Mark I, McCarthy, and Jason, Flannick
- Subjects
Metabolic Diseases ,Physiology ,Humans ,Human Genetics ,Cell Biology ,Molecular Biology ,Article - Abstract
We investigate the extent to which human genetic data are incorporated into studies that hypothesize novel links between genes and metabolic disease. To lower the barriers to using genetic data, we present an approach to enable researchers to evaluate human genetic support for experimentally determined hypotheses.
- Published
- 2022
- Full Text
- View/download PDF
5. Insulin Prevents Hypercholesterolemia by Suppressing 12α-Hydroxylated Bile Acids
- Author
-
Ivana Semova, Amy E. Levenson, Joanna Krawczyk, Kevin Bullock, Mary E. Gearing, Alisha V. Ling, Kathryn A. Williams, Ji Miao, Stuart S. Adamson, Dong-Ju Shin, Satyapal Chahar, Mark J. Graham, Rosanne M. Crooke, Lee R. Hagey, David Vicent, Sarah D. de Ferranti, Srividya Kidambi, Clary B. Clish, and Sudha B. Biddinger
- Subjects
Simvastatin ,Cross-Over Studies ,Hypercholesterolemia ,Hyperlipidemias ,Cholesterol, LDL ,Ezetimibe ,Receptor, Insulin ,Bile Acids and Salts ,Mice ,Diabetes Mellitus, Type 1 ,Liver ,Physiology (medical) ,Animals ,Humans ,Insulin ,Steroid 12-alpha-Hydroxylase ,lipids (amino acids, peptides, and proteins) ,Cardiology and Cardiovascular Medicine - Abstract
Background: The risk of cardiovascular disease in type 1 diabetes remains extremely high, despite marked advances in blood glucose control and even the widespread use of cholesterol synthesis inhibitors. Thus, a deeper understanding of insulin regulation of cholesterol metabolism, and its disruption in type 1 diabetes, could reveal better treatment strategies. Methods: To define the mechanisms by which insulin controls plasma cholesterol levels, we knocked down the insulin receptor, FoxO1, and the key bile acid synthesis enzyme, CYP8B1. We measured bile acid composition, cholesterol absorption, and plasma cholesterol. In parallel, we measured markers of cholesterol absorption and synthesis in humans with type 1 diabetes treated with ezetimibe and simvastatin in a double-blind crossover study. Results: Mice with hepatic deletion of the insulin receptor showed marked increases in 12α-hydroxylated bile acids, cholesterol absorption, and plasma cholesterol. This phenotype was entirely reversed by hepatic deletion of FoxO1 . FoxO1 is inhibited by insulin and required for the production of 12α-hydroxylated bile acids, which promote intestinal cholesterol absorption and suppress hepatic cholesterol synthesis. Knockdown of Cyp8b1 normalized 12α-hydroxylated bile acid levels and completely prevented hypercholesterolemia in mice with hepatic deletion of the insulin receptor (n=5–30), as well as mouse models of type 1 diabetes (n=5–22). In parallel, the cholesterol absorption inhibitor, ezetimibe, normalized cholesterol absorption and low-density lipoprotein cholesterol in patients with type 1 diabetes as well as, or better than, the cholesterol synthesis inhibitor, simvastatin (n=20). Conclusions: Insulin, by inhibiting FoxO1 in the liver, reduces 12α-hydroxylated bile acids, cholesterol absorption, and plasma cholesterol levels. Thus, type 1 diabetes leads to a unique set of derangements in cholesterol metabolism, with increased absorption rather than synthesis. These derangements are reversed by ezetimibe, but not statins, which are currently the first line of lipid-lowering treatment in type 1 diabetes. Taken together, these data suggest that a personalized approach to lipid lowering in type 1 diabetes may be more effective and highlight the need for further studies specifically in this group of patients.
- Published
- 2022
- Full Text
- View/download PDF
6. Inhibition of nonalcoholic fatty liver disease in mice by selective inhibition of mTORC1
- Author
-
Bridget S. Gosis, Shogo Wada, Chelsea Thorsheim, Kristina Li, Sunhee Jung, Joshua H. Rhoades, Yifan Yang, Jeffrey Brandimarto, Li Li, Kahealani Uehara, Cholsoon Jang, Matthew Lanza, Nathan B. Sanford, Marc R. Bornstein, Sunhye Jeong, Paul M. Titchenell, Sudha B. Biddinger, and Zoltan Arany
- Subjects
Multidisciplinary ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,General Science & Technology ,Liver Disease ,Chronic Liver Disease and Cirrhosis ,nutritional and metabolic diseases ,Mechanistic Target of Rapamycin Complex 1 ,Lipid Metabolism ,digestive system ,Article ,digestive system diseases ,Oral and gastrointestinal ,Hepatitis ,Mice ,Liver ,Non-alcoholic Fatty Liver Disease ,Humans ,Animals ,biological phenomena, cell phenomena, and immunity ,Sterol Regulatory Element Binding Protein 1 ,Digestive Diseases ,Gene Deletion - Abstract
INTRODUCTION: As many as 100 million people in the US have nonalcoholic fatty liver disease (NAFLD), characterized by increased liver lipid accumulation, which often leads to hepatocyte injury and fibrosis, characteristics of nonalcoholic steatohepatitis (NASH). NASH in turn can progress to cirrhosis and hepatocellular carcinoma. There are currently no US Food and Drug Administration–approved therapies for NAFLD or NASH. NAFLD occurs when there is disequilibrium between the processes of hepatic lipid synthesis and consumption. The nutrient sensor mechanistic target of rapamycin complex 1 (mTORC1) regulates several of these pathways. mTORC1 is thus an attractive target to modulate lipid homeostasis in the liver. However, mTORC1 also regulates numerous other cellular pathways, and blunting of mTORC1 modulation can lead to unexpected feedback loops and unwanted effects. RATIONALE: We hypothesized that selective modulation of hepatic mTORC1 signaling could benefit liver lipid metabolism and prevent NAFLD. In non-liver cell types, the protein folliculin (FLCN) has been shown to confer substrate specificity to mTORC1. Deletion of FLCN inhibits mTORC1-mediated phosphorylation of the transcription factor E3/B (TFE3/B) family of transcription factors, without affecting mTORC1-driven phosphorylation of its canonical substrates ribosomal protein S6 kinase beta-1 (S6K1) and eukaryotic translation initiation factor 4E–binding protein 1 (4E-BP1). Unphosphorylated TFE3 translocates to the nucleus and activates genes that promote lysosomal biogenesis, mitochondrial biogenesis, and oxidative metabolism. We reasoned that suppression of FLCN in the liver might promote fatty acid oxidation and lipid clearance without untoward effects of generalized mTORC1 inhibition. RESULTS: Hepatocyte-specific genetic deletion of Flcn in adult mice selectively inhibited mTORC1-mediated cytoplasmic sequestration of TFE3, with little effect on other mTORC1 targets, including S6K, 4E-BP1, and Lipin1. Hepatocyte loss of Flcn protected mice from both NAFLD and NASH and partially reversed these processes when already established. The protection against NAFLD and NASH required TFE3, which activated lipid clearance. Unleashed TFE3 additionally suppressed de novo lipogenesis. The latter was mediated in part by TFE3-mediated induction of insulin-induced gene 2 (Insig2) to inhibit proteolytic activation of sterol regulatory element–binding protein-1c (SREBP-1c), a critical lipogenic transcription factor. CONCLUSION: Our data establish FLCN as a critical regulator of lipid homeostasis in the liver. Flcn deletion affords selective inhibition of mTORC1, leading to nuclear translocation and activation of the transcription factor TFE3, which coordinates hepatic lipid metabolic pathways to protect against NAFLD and NASH in mice. Thus, our data reveal FLCN as a promising target for the treatment of NAFLD and NASH. The data also illuminate previously published and seemingly conflicting data, which likely reflected different effects on each arm of mTORC1 signaling. There have been numerous attempts by many to develop disease-specific treatments for NAFLD and NASH, thus far without success. A recurrent problem has been the many compensatory responses by the liver to targeting any one pathway; for example, inhibitors of acetyl–coenzyme A carboxylase led to compensatory activation of SREBP-1c and consequent hyperlipidemia. Targeting FLCN is thus particularly attractive, in that loss of FLCN simultaneously and favorably affects multiple aspects of hepatic lipid homeostasis, including promoting fatty acid oxidation and lysosomal biogenesis and inhibiting de novo lipogenesis.
- Published
- 2022
7. microRNA-146a-5p association with the cardiometabolic disease risk factor TMAO
- Author
-
Jody Albright, Brian J. Bennett, Tangi L. Smallwood, Wendy A. Pitman, Praveen Sethupathy, Ryan E. Temel, Kunjie Hua, Alisha R. Coffey, Matt Kanke, Sudha B. Biddinger, Daniel Pomp, Erik R. Gertz, and Raad Z. Gharaibeh
- Subjects
Collaborative Cross Mice ,medicine.medical_specialty ,Physiology ,Hypercholesterolemia ,Population ,Context (language use) ,Biology ,Quantitative trait locus ,Diet, High-Fat ,Choline ,Cohort Studies ,Gene Knockout Techniques ,Methylamines ,Mice ,Risk Factors ,Internal medicine ,Chlorocebus aethiops ,microRNA ,Genetics ,medicine ,Animals ,RNA-Seq ,education ,Chromosome 12 ,Mice, Knockout ,education.field_of_study ,Genetic heterogeneity ,NF-kappa B ,Atherosclerosis ,Receptor, Insulin ,Disease Models, Animal ,MicroRNAs ,Insulin receptor ,Endocrinology ,Liver ,Knockout mouse ,biology.protein ,Diet, Atherogenic ,Female ,Research Article - Abstract
Trimethylamine-N-oxide (TMAO), a microbial choline metabolism byproduct that is processed in the liver and excreted into circulation, is associated with increased atherosclerotic lesion formation and cardiovascular disease risk. Genetic regulators of TMAO levels are largely unknown. In the present study, we used 288 mice from a genetically heterogeneous mouse population [Diversity Outbred (DO)] to determine hepatic microRNA associations with TMAO in the context of an atherogenic diet. We also validated findings in two additional animal models of atherosclerosis: liver-specific insulin receptor knockout mice fed a chow diet (LIRKO) and African green monkeys fed high-fat/high-cholesterol diet. Small RNA-sequencing analysis in DO mice, LIRKO mice, and African green monkeys identified only one hepatic microRNA (miR-146a-5p) that is aberrantly expressed across all three models. Moreover, miR-146a-5p levels are associated with circulating TMAO after atherogenic diet in each of these models. We also performed high-resolution genetic mapping and identified a novel quantitative trait locus on Chromosome 12 for TMAO levels. This interval includes two genes, Numb and Dlst, which are inversely correlated with both miR-146a and TMAO and are predicted targets of miR-146a. Both of these genes have been validated as direct targets of miR-146a, though in other cellular contexts. This is the first report to our knowledge of a link between miR-146 and TMAO. Our findings suggest that miR-146-5p, as well as one or more genes at the Chromosome 12 QTL (possibly Numb or Dlst), is strongly linked to TMAO levels and likely involved in the control of atherosclerosis.
- Published
- 2019
- Full Text
- View/download PDF
8. Markers of cholesterol synthesis are elevated in adolescents and young adults with type 2 diabetes
- Author
-
Joanna Krawczyk, David M. Maahs, Sudha B. Biddinger, Amy E. Levenson, Elaine M. Urbina, Philip R. Khoury, R. Paul Wadwa, Sarah D. de Ferranti, Thomas R. Kimball, Lawrence M. Dolan, Clary B. Clish, Ivana Semova, Kathryn A. Williams, Amy S. Shah, and Kevin Bullock
- Subjects
Adult ,medicine.medical_specialty ,endocrine system diseases ,Adolescent ,Endocrinology, Diabetes and Metabolism ,Campesterol ,030209 endocrinology & metabolism ,Lathosterol ,Type 2 diabetes ,Article ,Body Mass Index ,03 medical and health sciences ,chemistry.chemical_compound ,Young Adult ,0302 clinical medicine ,Internal medicine ,Internal Medicine ,medicine ,Humans ,030212 general & internal medicine ,Obesity ,Young adult ,business.industry ,Lanosterol ,Type 2 Diabetes Mellitus ,nutritional and metabolic diseases ,Phytosterols ,medicine.disease ,Sitosterols ,Endocrinology ,Cholesterol ,chemistry ,Diabetes Mellitus, Type 2 ,Case-Control Studies ,Pediatrics, Perinatology and Child Health ,lipids (amino acids, peptides, and proteins) ,business ,Body mass index ,Dyslipidemia ,Biomarkers - Abstract
Background Changes in cholesterol absorption and cholesterol synthesis may promote dyslipidemia and cardiovascular disease in individuals with type 2 diabetes mellitus (T2DM). Objective To assess cholesterol synthesis and absorption in lean individuals, obese individuals, and individuals with T2DM. Methods We measured lathosterol and lanosterol (markers of cholesterol synthesis) as well as campesterol and β-sitosterol (markers of cholesterol absorption) in the serum of 15-26 years old individuals with T2DM (n = 95), as well as their lean (n = 98) and obese (n = 92) controls. Results Individuals with T2DM showed a 51% increase in lathosterol and a 65% increase in lanosterol compared to lean controls. Similarly, obese individuals showed a 31% increase in lathosterol compared to lean controls. Lathosterol and lanosterol were positively correlated with body mass index, fasting insulin and glucose, serum triglycerides, and C-reactive protein, and negatively correlated with HDL-cholesterol. In contrast, campesterol and β-sitosterol were not altered in individuals with T2DM. Moreover, campesterol and β-sitosterol were negatively correlated with body mass index, fasting insulin, and C-reactive protein and were positively correlated with HDL-cholesterol. Conclusions Adolescents and young adults with T2DM show evidence of increased cholesterol synthesis compared to non-diabetic lean controls. These findings suggest that T2DM may promote cardiovascular disease by increasing cholesterol synthesis, and provide additional rationale for the use of cholesterol synthesis inhibitors in this group. This article is protected by copyright. All rights reserved.
- Published
- 2020
9. YAP suppresses gluconeogenic gene expression through PGC1α
- Author
-
Dong-Ju Shin, Jonathan M. Dreyfuss, Hui Pan, Zhiqiang Lin, Sudha B. Biddinger, Fernando D. Camargo, Ji Miao, and Yue Hu
- Subjects
Male ,0301 basic medicine ,Cell signaling ,Carcinoma, Hepatocellular ,Primary Cell Culture ,Regulator ,Cell Cycle Proteins ,Biology ,Article ,Mice ,Random Allocation ,03 medical and health sciences ,Coactivator ,Transcriptional regulation ,Animals ,Humans ,Transcription factor ,Adaptor Proteins, Signal Transducing ,Regulation of gene expression ,Hippo signaling pathway ,Gene knockdown ,Hepatology ,Liver Neoplasms ,Gluconeogenesis ,YAP-Signaling Proteins ,Hep G2 Cells ,Phosphoproteins ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Cell biology ,030104 developmental biology ,Gene Expression Regulation ,Biochemistry ,Glucose-6-Phosphatase ,Phosphoenolpyruvate Carboxykinase (GTP) ,Transcription Factors - Abstract
Cell growth and proliferation are tightly coupled to metabolism, and dissecting the signaling molecules which link these processes is an important step towards understanding development, regeneration and cancer. The transcriptional regulator Yes-associated protein 1 (YAP) is a key regulator of liver size, development and function. We now show that YAP can also suppress gluconeogenic gene expression. Yap deletion in primary hepatocytes potentiates the gluconeogenic gene response to glucagon and dexamethasone, whereas constitutively active YAP suppresses it. The effects of YAP are mediated by the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1 (PGC1α). YAP inhibits the ability of PGC1α to bind to and activate transcription from the promoters of its gluconeogenic targets and the effects of YAP are blunted upon knockdown of PGC1α. In vivo, constitutively active YAP lowers plasma glucose levels and increases liver size. YAP therefore appears to reprogram cellular metabolism, diverting substrates away from the energy consuming process of gluconeogenesis, and towards the anabolic process of growth.
- Published
- 2017
- Full Text
- View/download PDF
10. Obesity and type 2 diabetes are associated with elevated PCSK9 levels in young women
- Author
-
R. Paul Wadwa, Elaine M. Urbina, Amy S. Shah, Thomas R. Kimball, Amy E. Levenson, Philip R. Khoury, Sarah D. de Ferranti, David M. Maahs, Lawrence M. Dolan, and Sudha B. Biddinger
- Subjects
medicine.medical_specialty ,business.industry ,Cross-sectional study ,Endocrinology, Diabetes and Metabolism ,PCSK9 ,030209 endocrinology & metabolism ,Type 2 diabetes ,030204 cardiovascular system & hematology ,medicine.disease ,Obesity ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Diabetes mellitus ,Pediatrics, Perinatology and Child Health ,Cohort ,Internal Medicine ,Medicine ,Young adult ,business ,Body mass index - Abstract
Background Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key regulator of low-density lipoprotein cholesterol and cardiovascular disease risk, and is an emerging therapeutic target. Objective We compared serum PCSK9 levels in young adults, with and without type 2 diabetes. Subjects and methods Cross-sectional analysis was conducted in a cohort, aged 15 to 26 years, in Cincinnati, OH, from 2005 to 2010. Serum PCSK9 levels were measured in 94 youth with type 2 diabetes, 93 obese control subjects, and 99 lean control subjects. Correlative analyses were conducted to determine significant covariates of PCSK9 by group and sex, and multivariate linear regression models were used to study the independent determinants of PCSK9. Results In females, PCSK9 levels were significantly increased in the obese and type 2 diabetes subjects relative to the lean controls (P
- Published
- 2017
- Full Text
- View/download PDF
11. Acute suppression of insulin resistance-associated hepatic miR-29 in vivo improves glycemic control in adult mice
- Author
-
Rodica P. Bunaciu, Rebecca L. Cubitt, Sudha B. Biddinger, James L. Graham, C. Lisa Kurtz, Peter J. Havel, Praveen Sethupathy, M. Mahmood Hussain, Ji Miao, Matt Kanke, Yu-Han Hung, Phillip J. White, and Liye Zhou
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,Physiology ,Medical Physiology ,Oligonucleotides ,Mice, Obese ,Type 2 diabetes ,Inbred C57BL ,Energy homeostasis ,Obese ,DNA Methyltransferase 3A ,Transcriptome ,Pathogenesis ,Mice ,0302 clinical medicine ,insulin resistance ,2.1 Biological and endogenous factors ,Aetiology ,Liver Disease ,Diabetes ,Liver ,Zucker ,030220 oncology & carcinogenesis ,Type 2 ,Research Article ,Biotechnology ,medicine.medical_specialty ,Biochemistry & Molecular Biology ,UCD-T2DM ,Biology ,liver ,03 medical and health sciences ,Insulin resistance ,In vivo ,Diabetes mellitus ,Internal medicine ,microRNA ,medicine ,Diabetes Mellitus ,Genetics ,Animals ,Humans ,Obesity ,Metabolic and endocrine ,Nutrition ,Base Sequence ,Enho ,medicine.disease ,Macaca mulatta ,Rats, Zucker ,Rats ,Mice, Inbred C57BL ,MicroRNAs ,030104 developmental biology ,Endocrinology ,HEK293 Cells ,Diabetes Mellitus, Type 2 ,microRNA-29 ,Insulin Resistance ,Digestive Diseases - Abstract
MicroRNAs (miRNAs) are important posttranscriptional regulators of metabolism and energy homeostasis. Dysregulation of certain miRNAs in the liver has been shown to contribute to the pathogenesis of Type 2 diabetes (T2D), in part by impairing hepatic insulin sensitivity. By small RNA-sequencing analysis, we identified seven hepatic miRNAs (including miR-29b) that are consistently aberrantly expressed across five different rodent models of metabolic dysfunction that share the feature of insulin resistance (IR). We also showed that hepatic miR-29b exhibits persistent dysregulation during disease progression in a rat model of diabetes, UCD-T2DM. Furthermore, we observed that hepatic levels of miR-29 family members are attenuated by interventions known to improve IR in rodent and rhesus macaque models. To examine the function of the miR-29 family in modulating insulin sensitivity, we used locked nucleic acid (LNA) technology and demonstrated that acute in vivo suppression of the miR-29 family in adult mice leads to significant reduction of fasting blood glucose (in both chow-fed lean and high-fat diet-fed obese mice) and improvement in insulin sensitivity (in chow-fed lean mice). We carried out whole transcriptome studies and uncovered candidate mechanisms, including regulation of DNA methyltransferase 3a ( Dnmt3a) and the hormone-encoding gene Energy homeostasis associated ( Enho). In sum, we showed that IR/T2D is linked to dysregulation of hepatic miR-29b across numerous models and that acute suppression of the miR-29 family in adult mice leads to improved glycemic control. Future studies should investigate the therapeutic utility of miR-29 suppression in different metabolic disease states. Enho; insulin resistance; liver; microRNA-29 (miR-29); UCD-T2DM
- Published
- 2019
12. Type 1 Diabetes is Associated with an Increase in Cholesterol Absorption Markers but a Decrease in Cholesterol Synthesis Markers in a Young Adult Population
- Author
-
Sarah D. de Ferranti, Kathryn A. Williams, Thomas R. Kimball, Amy S. Shah, Ivana Semova, Kevin Bullock, Clary B. Clish, David M. Maahs, Sudha B. Biddinger, Franziska K. Bishop, Philip R. Khoury, Amy E. Levenson, Elaine M. Urbina, R. Paul Wadwa, Lawrence M. Dolan, and Joanna Krawczyk
- Subjects
Adult ,Male ,medicine.medical_specialty ,Adolescent ,Endocrinology, Diabetes and Metabolism ,Campesterol ,Population ,Lathosterol ,030204 cardiovascular system & hematology ,Article ,Mass Spectrometry ,03 medical and health sciences ,chemistry.chemical_compound ,Young Adult ,0302 clinical medicine ,Risk Factors ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Humans ,030212 general & internal medicine ,education ,Child ,Dyslipidemias ,Glycated Hemoglobin ,Type 1 diabetes ,education.field_of_study ,Nutrition and Dietetics ,Anthropometry ,business.industry ,medicine.disease ,Blood pressure ,Endocrinology ,Cholesterol ,Diabetes Mellitus, Type 1 ,chemistry ,Cardiovascular Diseases ,Female ,Cardiology and Cardiovascular Medicine ,business ,Body mass index ,Dyslipidemia ,Biomarkers - Abstract
BACKGROUND: To optimize treatment and prevent cardiovascular disease in subjects with type 1 diabetes, it is important to determine how cholesterol metabolism changes with type 1 diabetes. OBJECTIVE: To compare plasma levels of campesterol and β-sitosterol, markers of cholesterol absorption, as well as lathosterol, a marker of cholesterol synthesis, in youth with and without type 1 diabetes. METHODS: Serum samples were obtained from adolescent subjects with type 1 diabetes [n= 175, mean age 15.2 years, mean duration of diabetes 8.2 years] and without diabetes [n=74, mean age 15.4 years]. Campesterol, β-sitosterol, and lathosterol, were measured using targeted liquid chromatography tandem mass spectrometry, normalized to the control serum mean, and expressed in arbitrary units. The markers were then compared between groups and correlated with the available cardiometabolic variables. RESULTS: Campesterol and β-sitosterol levels were 30% higher in subjects with type 1 diabetes and positively correlated with hemoglobin A1c levels. In contrast, lathosterol levels were 20% lower in subjects with type 1 diabetes and positively correlated with triglycerides, body mass index, and systolic blood pressure. CONCLUSION: Plasma markers suggest that cholesterol absorption is increased whereas cholesterol synthesis is decreased in type 1 diabetes. Further studies to address the impact of these changes on the relative efficacy of cholesterol absorption and synthesis inhibitors in subjects with type 1 diabetes are urgently needed.
- Published
- 2019
13. Effect of Leptin Administration on Circulating Apolipoprotein CIII levels in Patients With Lipodystrophy
- Author
-
Ranganath Muniyappa, Monica C. Skarulis, Mary Walter, Brent S. Abel, Amy E. Levenson, Sudha B. Biddinger, Michael Ring, Andrea Kassai, Rebecca J. Brown, Simeon I. Taylor, and Phillip Gorden
- Subjects
Adult ,Leptin ,Male ,medicine.medical_specialty ,Lipodystrophy ,Post hoc ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Context (language use) ,030204 cardiovascular system & hematology ,Biochemistry ,Mice ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,Animals ,Humans ,Medicine ,In patient ,030212 general & internal medicine ,skin and connective tissue diseases ,Apolipoprotein C-III ,Lipoprotein lipase ,business.industry ,Biochemistry (medical) ,Hypertriglyceridemia ,Original Articles ,Middle Aged ,medicine.disease ,Cross-Sectional Studies ,Case-Control Studies ,lipids (amino acids, peptides, and proteins) ,Female ,sense organs ,Apolipoprotein CIII ,business - Abstract
Apolipoprotein CIII (apoCIII), an inhibitor of lipoprotein lipase, plays an important role in triglyceride metabolism. However, the role of apoCIII in hypertriglyceridemia in lipodystrophy and the effects of leptin replacement on apoCIII levels are unknown.The objective of the study was to test the hypotheses that apoCIII is elevated in hypertriglyceridemic patients with lipodystrophy and that leptin replacement in these patients lowers circulating apoCIII.Using a post hoc cross-sectional case-control design, we compared serum apoCIII levels from patients with lipodystrophy not associated with HIV (n = 60) and age-, gender-, race-, and ethnicity-matched controls (n = 54) participating in ongoing studies at the National Institutes of Health. In a prospective, open-label, ongoing study, we studied the effects of 6–12 months of leptin replacement on apoCIII in lipodystrophy patients as an exploratory outcome.ApoCIII was higher in lipodystrophy patients (geometric mean [25th and 75th percentiles]) (23.9 mg/dL [14.6, 40.3]) compared with controls (14.9 mg/dL [12.3, 17.7]) (P.0001). ApoCIII and triglyceride levels were positively correlated in patients with lipodystrophy (R = 0.72, P.0001) and healthy controls (R = 0.6, P.0001). Leptin replacement (6–12 mo) did not significantly alter apoCIII (before leptin: 23.4 mg/dL [14.5, 40.1]; after leptin: 21.4 mg/dL [16.7, 28.3]; P = .34).Leptin replacement in lipodystrophy did not alter serum apoCIII levels. Elevated apoCIII may play a role in the hypertriglyceridemia of lipodystrophy independent of leptin deficiency and replacement.
- Published
- 2016
- Full Text
- View/download PDF
14. Nonalcoholic fatty liver disease and gastric bypass surgery regulate serum and hepatic levels of pyruvate kinase isoenzyme M2
- Author
-
Luca Meoli, Courtney Panciotti, Sudha B. Biddinger, Nitin Gupta, Nima Saeidi, Kathleen E. Corey, and Nicholas Stylopoulos
- Subjects
0301 basic medicine ,Nonalcoholic steatohepatitis ,Adult ,Male ,medicine.medical_specialty ,Thyroid Hormones ,Physiology ,Endocrinology, Diabetes and Metabolism ,Pyruvate Kinase ,Gastric Bypass ,medicine.disease_cause ,Isozyme ,digestive system ,03 medical and health sciences ,Mice ,Non-alcoholic Fatty Liver Disease ,Physiology (medical) ,Internal medicine ,Nonalcoholic fatty liver disease ,Medicine ,Animals ,Humans ,Obesity ,Metabolic Syndrome ,business.industry ,Gastric bypass surgery ,nutritional and metabolic diseases ,Membrane Proteins ,Middle Aged ,medicine.disease ,digestive system diseases ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Jejunum ,Liver ,Female ,Steatohepatitis ,Metabolic syndrome ,business ,Carrier Proteins ,Pyruvate kinase ,Research Article - Abstract
Treatment of nonalcoholic fatty liver disease (NAFLD) focuses on the underlying metabolic syndrome, and Roux-en-Y gastric bypass surgery (RYGB) remains one of the most effective options. In rodents and human patients, RYGB induces an increase in the gene and protein expression levels of the M2 isoenzyme of pyruvate kinase (PKM2) in the jejunum. Since PKM2 can be secreted in the circulation, our hypothesis was that the circulating levels of PKM2 increase after RYGB. Our data, however, revealed an unexpected finding and a potential new role of PKM2 for the natural history of metabolic syndrome and NAFLD. Contrary to our initial hypothesis, RYGB-treated patients had decreased PKM2 blood levels compared with a well-matched group of patients with severe obesity before RYGB. Interestingly, PKM2 serum concentration correlated with body mass index before but not after the surgery. This prompted us to evaluate other potential mechanisms and sites of PKM2 regulation by the metabolic syndrome and RYGB. We found that in patients with NAFLD and nonalcoholic steatohepatitis (NASH), the liver had increased PKM2 expression levels, and the enzyme appears to be specifically localized in Kupffer cells. The study of murine models of metabolic syndrome and NASH replicated this pattern of expression, further suggesting a metabolic link between hepatic PKM2 and NAFLD. Therefore, we conclude that PKM2 serum and hepatic levels increase in both metabolic syndrome and NAFLD and decrease after RYGB. Thus, PKM2 may represent a new target for monitoring and treatment of NAFLD.
- Published
- 2018
15. Fructose and glucose can regulate mammalian target of rapamycin complex 1 and lipogenic gene expression via distinct pathways
- Author
-
Ji Miao, Sudha B. Biddinger, Ivana Semova, Anthony N. Hollenberg, Hong Kang, Matthew D. Hirschey, David Masson, Yue Hu, Satyapal Chahar, Xiaowei Sun, Division of Endocrinology, Children's Hospital Boston, Harvard Medical School, Harvard Medical School [Boston] ( HMS ), Beth Israel Deaconess Medical Center, Lipides - Nutrition - Cancer [Dijon - U1231] ( LNC ), Université de Bourgogne ( UB ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale ( INSERM ), and Duke University Medical Center
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biochemistry ,metabolic syndrome ,fructose ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Internal medicine ,Gene expression ,medicine ,Animals ,glucose ,Molecular Biology ,[ SDV.BBM ] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Triglycerides ,lipogenesis ,Mice, Knockout ,Fatty liver ,dyslipidemia ,mammalian target of rapamycin (mTOR) ,Lipid metabolism ,Fructose ,Cell Biology ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Endocrinology ,Metabolism ,chemistry ,Gene Expression Regulation ,Liver ,Lipogenesis ,Metabolic syndrome ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
International audience; Although calorically equivalent to glucose, fructose appears to be more lipogenic, promoting dyslipidemia, fatty liver disease, cardiovascular disease, and diabetes. To better understand how fructose induces lipogenesis, we compared the effects of fructose and glucose on mammalian target of rapamycin complex 1 (mTORC1), which appeared to have both positive and negative effects on lipogenic gene expression. We found that fructose acutely and transiently suppressed mTORC1 signalingin vitroandin vivoThe constitutive activation of mTORC1 reduced hepatic lipogenic gene expression and produced hypotriglyceridemia after 1 week of fructose feeding. In contrast, glucose did not suppress mTORC1, and the constitutive activation of mTORC1 failed to suppress plasma triglycerides after 1 week of glucose feeding. Thus, these data reveal fundamental differences in the signaling pathways used by fructose and glucose to regulate lipid metabolism.
- Published
- 2018
- Full Text
- View/download PDF
16. Role of Insulin in the Regulation of Proprotein Convertase Subtilisin/Kexin Type 9
- Author
-
Mark J. Graham, Praveen V. Manthena, Alisha V. Ling, Dong-Ju Shin, Ji Miao, Jingwen Liu, Rosanne M. Crooke, Sudha B. Biddinger, and Mary E. Haas
- Subjects
biology ,PCSK9 ,Insulin ,medicine.medical_treatment ,Subtilisin ,Proprotein convertase ,IRS2 ,Insulin receptor ,Biochemistry ,LDL receptor ,biology.protein ,medicine ,Kexin ,Cardiology and Cardiovascular Medicine - Abstract
Objective— Proprotein convertase subtilisin/kexin type 9 (PCSK9), which binds the low-density lipoprotein receptor and targets it for degradation, has emerged as an important regulator of serum cholesterol levels and cardiovascular disease risk. Although much work is currently focused on developing therapies for inhibiting PCSK9, the endogenous regulation of PCSK9, particularly by insulin, remains unclear. The objective of these studies was to determine the effects of insulin on PCSK9 in vitro and in vivo. Approach and Results— Using rat hepatoma cells and primary rat hepatocytes, we found that insulin increased PCSK9 expression and increased low-density lipoprotein receptor degradation in a PCSK9-dependent manner. In parallel, hepatic Pcsk9 mRNA and plasma PCSK9 protein levels were reduced by 55% to 75% in mice with liver-specific knockout of the insulin receptor; 75% to 88% in mice made insulin-deficient with streptozotocin; and 65% in ob/ob mice treated with antisense oligonucleotides against the insulin receptor. However, antisense oligonucleotide–mediated knockdown of insulin receptor in lean, wild-type mice had little effect. In addition, we found that fasting was able to reduce PCSK9 expression by 80% even in mice that lack hepatic insulin signaling. Conclusions— Taken together, these data indicate that although insulin induces PCSK9 expression, it is not the sole or even dominant regulator of PCSK9 under all conditions.
- Published
- 2015
- Full Text
- View/download PDF
17. FoxO1 Is Required for Most of the Metabolic and Hormonal Perturbations Produced by Hepatic Insulin Receptor Deletion in Male Mice
- Author
-
Mary E Gearing, Alisha V. Ling, Rebecca Clements, Ivana Semova, Dong-Ju Shin, Sudha B. Biddinger, and Zon Weng Lai
- Subjects
0301 basic medicine ,Blood Glucose ,Leptin ,Male ,medicine.medical_specialty ,medicine.medical_treatment ,Gene Expression ,FOXO1 ,Carbohydrate metabolism ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Endocrinology ,Internal medicine ,medicine ,Animals ,Insulin ,Insulin-Like Growth Factor I ,Research Articles ,Triglycerides ,Mice, Knockout ,Leptin receptor ,biology ,Fatty acid metabolism ,Chemistry ,Forkhead Box Protein O1 ,Lipogenesis ,Fatty Acids ,Gluconeogenesis ,Lipids ,Receptor, Insulin ,Mice, Inbred C57BL ,Insulin receptor ,030104 developmental biology ,Liver ,biology.protein ,Receptors, Leptin ,Oxidation-Reduction ,Hormone - Abstract
Insulin coordinates the complex response to feeding, affecting numerous metabolic and hormonal pathways. Forkhead box protein O1 (FoxO1) is one of several signaling molecules downstream of insulin; FoxO1 drives gluconeogenesis and is suppressed by insulin. To determine the role of FoxO1 in mediating other actions of insulin, we studied mice with hepatic deletion of the insulin receptor, FoxO1, or both. We found that mice with deletion of the insulin receptor alone showed not only hyperglycemia but also a 70% decrease in plasma insulin-like growth factor 1 and delayed growth during the first 2 months of life, a 24-fold increase in the soluble leptin receptor and a 19-fold increase in plasma leptin levels. Deletion of the insulin receptor also produced derangements in fatty acid metabolism, with a decrease in the expression of the lipogenic enzymes, hepatic diglycerides, and plasma triglycerides; in parallel, it increased expression of the fatty acid oxidation enzymes. Mice with deletion of both insulin receptor and FoxO1 showed a much more modest phenotype, with normal or near-normal glucose levels, growth, leptin levels, hepatic diglycerides, and fatty acid oxidation gene expression; however, lipogenic gene expression remained low. Taken together, these data reveal the pervasive role of FoxO1 in mediating the effects of insulin on not only glucose metabolism but also other hormonal signaling pathways and even some aspects of lipid metabolism.
- Published
- 2017
18. Hepatocyte ABCA1 deletion impairs liver insulin signaling and lipogenesis
- Author
-
Praveen Sethupathy, Timothy A. Dinh, C. Lisa Kurtz, Sudha B. Biddinger, Peter E. Phelan, Alexander Bashore, Mingxia Liu, John S. Parks, Timothy F. Osborne, Chia Chi C. Key, Lijun Ma, Xuewei Zhu, Barry I. Freedman, Elena Boudyguina, and Soonkyu Chung
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Carbohydrate metabolism ,plasma membrane ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,Insulin resistance ,hepatosteatosis ,Internal medicine ,medicine ,hepatocyte ,Animals ,lcsh:QH301-705.5 ,Mice, Knockout ,Western-type diet ,biology ,mTORC ,Lipogenesis ,selective insulin resistance ,medicine.disease ,Sterol regulatory element-binding protein ,lipid raft ,Insulin receptor ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Cholesterol ,lcsh:Biology (General) ,Liver ,ABCA1 ,Hepatocyte ,vesicle trafficking ,biology.protein ,Hepatocytes ,Signal transduction ,Insulin Resistance ,Gene Deletion ,ATP Binding Cassette Transporter 1 ,Signal Transduction - Abstract
Plasma membrane (PM) free cholesterol (FC) is emerging as an important modulator of signal transduction. Here, we show that hepatocyte-specific knockout (HSKO) of the cellular FC exporter, ATP-binding cassette transporter A1 (ABCA1), leads to decreased PM FC content and defective trafficking of lysosomal FC to the PM. Compared with controls, chow-fed HSKO mice had reduced hepatic (1) insulin-stimulated Akt phosphorylation, (2) activation of the lipogenic transcription factor Sterol Regulatory Element Binding Protein (SREBP)-1c, and (3) lipogenic gene expression. Consequently, Western-type diet-fed HSKO mice were protected from steatosis. Surprisingly, HSKO mice had intact glucose metabolism; they showed normal gluconeogenic gene suppression in response to re-feeding and normal glucose and insulin tolerance. We conclude that: (1) ABCA1 maintains optimal hepatocyte PM FC, through intracellular FC trafficking, for efficient insulin signaling; and (2) hepatocyte ABCA1 deletion produces a form of selective insulin resistance so that lipogenesis is suppressed but glucose metabolism remains normal.
- Published
- 2017
19. Calorically Restricted Diets Decrease PCSK9 in Overweight Adolescents
- Author
-
David S. Ludwig, Carly E. Milliren, Amy E. Levenson, Henry A. Feldman, Sudha B. Biddinger, Cara B. Ebbeling, and S.D. de Ferranti
- Subjects
0301 basic medicine ,Blood Glucose ,Counseling ,Male ,medicine.medical_specialty ,Pediatric Obesity ,Time Factors ,Adolescent ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Medicine (miscellaneous) ,Down-Regulation ,030204 cardiovascular system & hematology ,Overweight ,Article ,03 medical and health sciences ,0302 clinical medicine ,Weight loss ,Internal medicine ,Glycemic load ,Weight Loss ,medicine ,Humans ,Insulin ,Medical nutrition therapy ,Child ,Diet, Fat-Restricted ,Nutrition and Dietetics ,business.industry ,PCSK9 ,Age Factors ,Glycemic Load ,medicine.disease ,Obesity ,030104 developmental biology ,Endocrinology ,Treatment Outcome ,Female ,medicine.symptom ,Insulin Resistance ,Proprotein Convertase 9 ,Cardiology and Cardiovascular Medicine ,business ,Energy Intake ,Dyslipidemia ,Biomarkers ,Boston - Abstract
Background and aims Nutritional therapy is the first line approach to treatment of hyperlipidemia in childhood. Proprotein convertase subtilisin kexin type 9 (PCSK9) is a key regulator of plasma cholesterol levels and a target of novel lipid-lowering pharmacotherapies. We examined the effects of an intensive nutritional intervention on PCSK9 levels in overweight adolescents with cardiovascular disease (CVD) risk factors. Methods and results Twenty seven obese and overweight adolescents with CVD risk factors were assigned to either a low fat or low glycemic load diet. During an 8-week " Intensive Phase ," assigned meals were delivered to the home, and all participants received weekly in-person home nutrition counseling and phone calls. The subjects then underwent a 4-month " Maintenance Phase " without food provision and with no in-person contact. Anthropometric measurements, laboratory data, and serum PCSK9 protein levels were measured at baseline, 8 weeks, and 6 months. PCSK9 decreased by 16.5% at 8 weeks (201.2 ± 56.3 vs 165.6 ± 58.4 ng/mL; p Maintenance Phase . Change in PCSK9 was associated with change in fasting insulin, HOMA-IR, and AUC insulin, independent of weight loss. Conclusions PCSK9 decreased in youth participating in an intensive dietary intervention. Change in HOMA-IR was associated with change in PCSK9, independent of weight loss, suggesting an important relationship with insulin sensitivity. ClinicalTrials.gov Identifier: NCT01080339.
- Published
- 2017
20. PKB/Akt phosphorylation of ERRγ contributes to insulin-mediated inhibition of hepatic gluconeogenesis
- Author
-
Yong-Hoon Kim, Ho Zoon Chae, Dongryeol Ryu, Hueng Sik Choi, Brian A. Hemmings, Inkyu Lee, Chul-Ho Lee, Kyung Seok Kim, Eun Kyung Yoo, Seung Hoi Koo, Sudha B. Biddinger, Jeong Sun Kim, Keum Jin Yang, Yanning Wang, Jongsun Park, Debby Hynx, and Don Kyu Kim
- Subjects
medicine.medical_specialty ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Type 2 diabetes ,medicine.disease ,Endocrinology ,Insulin resistance ,Nuclear receptor ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Transcriptional regulation ,Phosphorylation ,Receptor - Abstract
Aims/hypothesis Insulin resistance, a major contributor to the pathogenesis of type 2 diabetes, leads to increased hepatic glucose production (HGP) owing to an impaired ability of insulin to suppress hepatic gluconeogenesis. Nuclear receptor oestrogen-related receptor γ (ERRγ) is a major transcriptional regulator of hepatic gluconeogenesis. In this study, we investigated insulin-dependent post-translational modifications (PTMs) altering the transcriptional activity of ERRγ for the regulation of hepatic gluconeogenesis.
- Published
- 2014
- Full Text
- View/download PDF
21. MicroRNA-29 Fine-tunes the Expression of Key FOXA2-Activated Lipid Metabolism Genes and Is Dysregulated in Animal Models of Insulin Resistance and Diabetes
- Author
-
Ji Miao, Bailey C.E. Peck, Shengli Ding, Kasey C. Vickers, C. Lisa Kurtz, Praveen Sethupathy, Vandana Turaga, Stuart R Landstreet, Sudha B. Biddinger, P. Kay Lund, Carine Beysen, Emily E. Fannin, and Scott M. Turner
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Type 2 diabetes ,Biology ,Energy homeostasis ,Mice ,Insulin resistance ,Downregulation and upregulation ,Internal medicine ,Diabetes mellitus ,Cell Line, Tumor ,microRNA ,Internal Medicine ,medicine ,Animals ,Humans ,Pioglitazone ,Lipid metabolism ,Genetics/Genomes/Proteomics/Metabolomics ,medicine.disease ,3. Good health ,Rats ,Rats, Zucker ,Up-Regulation ,MicroRNAs ,Endocrinology ,Diabetes Mellitus, Type 2 ,Models, Animal ,Hepatocyte Nuclear Factor 3-beta ,Female ,Thiazolidinediones ,Insulin Resistance ,medicine.drug - Abstract
MicroRNAs (miRNAs) have emerged as biomarkers of metabolic status, etiological factors in complex disease, and promising drug targets. Recent reports suggest that miRNAs are critical regulators of pathways underlying the pathophysiology of type 2 diabetes. In this study, we demonstrate by deep sequencing and real-time quantitative PCR that hepatic levels of Foxa2 mRNA and miR-29 are elevated in a mouse model of diet-induced insulin resistance. We also show that Foxa2 and miR-29 are significantly upregulated in the livers of Zucker diabetic fatty (fa/fa) rats and that the levels of both returned to normal upon treatment with the insulin-sensitizing agent pioglitazone. We present evidence that miR-29 expression in human hepatoma cells is controlled in part by FOXA2, which is known to play a critical role in hepatic energy homeostasis. Moreover, we demonstrate that miR-29 fine-tunes FOXA2-mediated activation of key lipid metabolism genes, including PPARGC1A, HMGCS2, and ABHD5. These results suggest that miR-29 is an important regulatory factor in normal metabolism and may represent a novel therapeutic target in type 2 diabetes and related metabolic syndromes.
- Published
- 2014
22. Hepatic insulin receptor deficiency impairs the SREBP-2 response to feeding and statins
- Author
-
Yanning Wang, Praveen V. Manthena, Sudha B. Biddinger, Enpeng Zhao, Joel T. Haas, Irwin J. Kurland, Ji Miao, and Bhavapriya Vaitheesvaran
- Subjects
Transcriptional Activation ,medicine.medical_specialty ,Mice, 129 Strain ,liver insulin receptor knockout ,medicine.medical_treatment ,Gene Expression ,Mice, Transgenic ,QD415-436 ,Biochemistry ,Mice ,chemistry.chemical_compound ,Endocrinology ,Ezetimibe ,Internal medicine ,cholesterol biosynthesis ,medicine ,Animals ,Lovastatin ,Research Articles ,hepatic insulin signaling ,Oligonucleotide Array Sequence Analysis ,biology ,Cholesterol ,Lipogenesis ,Insulin ,Fasting ,Cell Biology ,Receptor, Insulin ,Biosynthetic Pathways ,Sterol regulatory element-binding protein ,Mice, Inbred C57BL ,Insulin receptor ,medicine.anatomical_structure ,Liver ,chemistry ,sterol regulatory element binding protein ,Hepatocyte ,biology.protein ,Azetidines ,lipids (amino acids, peptides, and proteins) ,Sterol regulatory element-binding protein 2 ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,Transcriptome ,Sterol Regulatory Element Binding Protein 2 ,medicine.drug - Abstract
The liver plays a central role in metabolism and mediating insulin action. To dissect the effects of insulin on the liver in vivo, we have studied liver insulin receptor knockout (LIRKO) mice. Because LIRKO livers lack insulin receptors, they are unable to respond to insulin. Surprisingly, the most profound derangement observed in LIRKO livers by microarray analysis is a suppression of the cholesterologenic genes. Sterol regulatory element binding protein (SREBP)-2 promotes cholesterologenic gene transcription, and is inhibited by intracellular cholesterol. LIRKO livers show a slight increase in hepatic cholesterol, a 40% decrease in Srebp-2, and a 50-90% decrease in the cholesterologenic genes at the mRNA and protein levels. In control mice, SREBP-2 and cholesterologenic gene expression are suppressed by fasting and restored by refeeding; in LIRKO mice, this response is abolished. Similarly, the ability of statins to induce Srebp-2 and the cholesterologenic genes is lost in LIRKO livers. In contrast, ezetimibe treatment robustly induces Srepb-2 and its targets in LIRKO livers, raising the possibility that insulin may regulate SREBP-2 indirectly, by altering the accumulation or distribution of cholesterol within the hepatocyte. Taken together, these data indicate that cholesterol synthesis is a key target of insulin action in the liver.
- Published
- 2014
- Full Text
- View/download PDF
23. Trimethylamine N-Oxide Binds and Activates PERK to Promote Metabolic Dysfunction
- Author
-
Bingdong Sha, Federico E. Rey, Mark J. Graham, Mary E Gearing, Ayana Henderson, Michael C. Petriello, Andrew J. Morris, Ji Miao, Emily P. Balskus, Kymberleigh A. Romano, Jiahui Tao, Walter J. Sandoval-Espinola, Mareike Schell, Rosanne M. Crooke, Sudha B. Biddinger, Sifan Chen, and André Kleinridders
- Subjects
0301 basic medicine ,endocrine system ,Indoles ,Physiology ,Mice, Obese ,FOXO1 ,Trimethylamine N-oxide ,Methylamines ,Mice ,eIF-2 Kinase ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Animals ,Humans ,EIF2AK3 ,Molecular Biology ,Transcription factor ,Metabolic Syndrome ,Mice, Knockout ,biology ,Chemistry ,Kinase ,Endoplasmic reticulum ,Hep G2 Cells ,Cell Biology ,Gastrointestinal Microbiome ,Cell biology ,Mice, Inbred C57BL ,Insulin receptor ,HEK293 Cells ,030104 developmental biology ,Oxygenases ,biology.protein ,Unfolded protein response ,Insulin Resistance ,030217 neurology & neurosurgery - Abstract
Summary The gut-microbe-derived metabolite trimethylamine N-oxide (TMAO) is increased by insulin resistance and associated with several sequelae of metabolic syndrome in humans, including cardiovascular, renal, and neurodegenerative disease. The mechanism by which TMAO promotes disease is unclear. We now reveal the endoplasmic reticulum stress kinase PERK (EIF2AK3) as a receptor for TMAO: TMAO binds to PERK at physiologically relevant concentrations; selectively activates the PERK branch of the unfolded protein response; and induces the transcription factor FoxO1, a key driver of metabolic disease, in a PERK-dependent manner. Furthermore, interventions to reduce TMAO, either by manipulation of the gut microbiota or by inhibition of the TMAO synthesizing enzyme, flavin-containing monooxygenase 3, can reduce PERK activation and FoxO1 levels in the liver. Taken together, these data suggest TMAO and PERK may be central to the pathogenesis of the metabolic syndrome.
- Published
- 2019
- Full Text
- View/download PDF
24. PCSK9 Is Increased in Youth With Type 1 Diabetes
- Author
-
Sarah D. de Ferranti, Elaine M. Urbina, Franziska K. Bishop, Amy S. Shah, Lawrence M. Dolan, R. Paul Wadwa, Sudha B. Biddinger, Amy E. Levenson, David M. Maahs, Philip R. Khoury, and Thomas R. Kimball
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Statin ,Apolipoprotein B ,medicine.drug_class ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Type 2 diabetes ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,Advanced and Specialized Nursing ,Type 1 diabetes ,biology ,business.industry ,PCSK9 ,e-Letters: Observations ,nutritional and metabolic diseases ,medicine.disease ,030104 developmental biology ,Endocrinology ,biology.protein ,lipids (amino acids, peptides, and proteins) ,business ,Dyslipidemia ,Lipoprotein - Abstract
Type 1 diabetes (T1D) is associated with a 10-fold increase in cardiovascular disease (CVD) risk, due in part to dyslipidemia. Patients with poorly controlled T1D show an increase in total and LDL cholesterol as well as elevated apolipoprotein B (ApoB) levels. Moreover, even patients with well-controlled T1D can show increased levels of ApoB. Dyslipidemia is particularly important in females with T1D, as the atheroprotective lipoprotein profile and reduced risk of CVD usually observed in females without diabetes are lost in females with diabetes (1,2). Proprotein convertase subtilisin/kexin type 9 (PCSK9) has recently emerged as a novel regulator of plasma cholesterol levels: gain-of-function PCSK9 mutations increase LDL cholesterol and CVD risk, whereas inhibitors of PCSK9 lower LDL cholesterol. Although PCSK9 is increased in individuals with obesity and type 2 diabetes (3,4), it is not known whether PCSK9 levels are also changed in individuals with T1D. Here, we measured PCSK9 levels in a subset of a previously described cohort of T1D and control subjects (5). For the current study, subjects with serum available for PCSK9 measurement were matched for age and sex between each group using the frequency matching method. Subjects who reported taking statin medications at the time of the study were excluded from the analyses ( n = 3, all from T1D …
- Published
- 2016
25. Effect of Leptin Replacement on PCSK9 in ob/ob Mice and Female Lipodystrophic Patients
- Author
-
Ji Miao, Rebecca J. Brown, Ranganath Muniyappa, Sudha B. Biddinger, Mary E. Haas, Amy E. Levenson, and Sarah D. de Ferranti
- Subjects
0301 basic medicine ,Leptin ,Male ,Lipodystrophy ,Gene Expression ,Mice, Obese ,030204 cardiovascular system & hematology ,Metreleptin ,chemistry.chemical_compound ,0302 clinical medicine ,Endocrinology ,Child ,Original Research ,Reverse Transcriptase Polymerase Chain Reaction ,digestive, oral, and skin physiology ,Serine Endopeptidases ,Cholesterol ,Liver ,Child, Preschool ,Kexin ,lipids (amino acids, peptides, and proteins) ,Female ,Proprotein Convertases ,Proprotein Convertase 9 ,hormones, hormone substitutes, and hormone antagonists ,medicine.medical_specialty ,Adolescent ,Blotting, Western ,Enzyme-Linked Immunosorbent Assay ,Biology ,03 medical and health sciences ,Young Adult ,Sex Factors ,Internal medicine ,medicine ,Animals ,Humans ,Obesity ,Triglycerides ,PCSK9 ,Cholesterol, LDL ,medicine.disease ,030104 developmental biology ,chemistry ,Lipoprotein - Abstract
Leptin treatment has beneficial effects on plasma lipids in patients with lipodystrophy, but the underlying mechanism is unknown. Proprotein convertase subtilisin/kexin type 9 (PCSK9) decreases low-density lipoprotein (LDL) clearance, promotes hypercholesterolemia, and has recently emerged as a novel therapeutic target. To determine the effect of leptin on PCSK9, we treated male and female ob/ob mice with leptin for 4 days via sc osmotic pumps (∼24 μg/d). Leptin reduced body weight and food intake in all mice, but the effects of leptin on plasma PCSK9 and lipids differed markedly between the sexes. In male mice, leptin suppressed PCSK9 but had no effect on plasma triglycerides or cholesterol. In female mice, leptin suppressed plasma triglycerides and cholesterol but had no effect on plasma PCSK9. In parallel, we treated female lipodystrophic patients (8 females, ages 5–23 y) with sc metreleptin injections (∼4.4 mg/d) for 4–6 months. In this case, leptin reduced plasma PCSK9 by 26% (298 ± 109 vs 221 ± 102 ng/mL; n = 8; P = .008), and the change in PCSK9 was correlated with a decrease in LDL cholesterol (r2 = 0.564, P = .03). In summary, in leptin-deficient ob/ob mice, the effects of leptin on PCSK9 and plasma lipids appeared to be independent of one another and strongly modified by sex. On the other hand, in lipodystrophic females, leptin treatment reduced plasma PCSK9 in parallel with LDL cholesterol.
- Published
- 2016
26. Insulin Dissociates the Effects of Liver X Receptor on Lipogenesis, Endoplasmic Reticulum Stress, and Inflammation
- Author
-
Rosanne M. Crooke, Jianguo Wang, Xiaowei Sun, Jean-Paul Pais de Barros, Abhiruchi J Mehta, David Masson, Ji Miao, Mary E. Haas, Sudha B. Biddinger, Mark J. Graham, Masanori Aikawa, Division of Endocrinology, Children's Hospital Boston, Harvard Medical School, Harvard Medical School [Boston] (HMS), Isis Pharmaceuticals, IBIS THERAPEUTICS-ISIS Therapeutics, Lipides - Nutrition - Cancer (U866) (LNC), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Bourgogne (UB)-Ecole Nationale Supérieure de Biologie Appliquée à la Nutrition et à l'Alimentation de Dijon (ENSBANA)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement, Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School [Boston] (HMS)-Brigham and Women's Hospital [Boston], ANR-11-LABX-0021,Lipstic,Lipoprotéines et santé : prévention et traitement des maladies inflammatoires non vasculaires et du(2011), Harvard Medical School [Boston] ( HMS ), Lipides - Nutrition - Cancer (U866) ( LNC ), Université de Bourgogne ( UB ) -Institut National de la Santé et de la Recherche Médicale ( INSERM ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Ecole Nationale Supérieure de Biologie Appliquée à la Nutrition et à l'Alimentation de Dijon ( ENSBANA ), Harvard Medical School [Boston] ( HMS ) -Brigham and Women's Hospital [Boston], and ANR-11-LABX-0021/11-LABX-0021,Lipstic,Lipoprotéines et santé : prévention et traitement des maladies inflammatoires non vasculaires et du ( 2011 )
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Lipid-metabolism ,Resistance ,Biochemistry ,Hepatitis ,MESH: Hepatitis ,MESH: Endoplasmic Reticulum Stress ,polycyclic compounds ,Insulin ,Gene-expression ,Phospholipids ,Liver X Receptors ,Mice, Knockout ,biology ,MESH : Gene Expression Regulation ,Fatty-acid synthesis ,food and beverages ,Endoplasmic Reticulum Stress ,Orphan Nuclear Receptors ,Cultured-cells ,Lipids ,MESH: Gene Expression Regulation ,MESH : Endoplasmic Reticulum Stress ,Messenger-rna ,Liver ,MESH: Orphan Nuclear Receptors ,Gene Knockdown Techniques ,Lipogenesis ,Female ,lipids (amino acids, peptides, and proteins) ,Signal Transduction ,liver X receptor ,medicine.medical_specialty ,Lxr-alpha ,Mice, Transgenic ,digestive system ,Phospholipid transfer protein ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,Insulin resistance ,MESH : Hepatitis ,Lysophosphatidylcholine acyltransferase ,Internal medicine ,medicine ,Animals ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Liver X receptor ,Molecular Biology ,[ SDV.BBM ] Life Sciences [q-bio]/Biochemistry, Molecular Biology ,Crosses, Genetic ,Endoplasmic reticulum ,Element-binding protein-1c ,MESH : Liver ,Cell Biology ,medicine.disease ,MESH : Orphan Nuclear Receptors ,Receptor, Insulin ,Mice, Inbred C57BL ,Insulin receptor ,030104 developmental biology ,Endocrinology ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,Nuclear receptor ,biology.protein ,Unfolded protein response ,Insulin Resistance ,MESH: Liver - Abstract
IF 4.258; International audience; Diabetes is characterized by increased lipogenesis as well as increased endoplasmic reticulum (ER) stress and inflammation. The nuclear hormone receptor liver X receptor (LXR) is induced by insulin and is a key regulator of lipid metabolism. It promotes lipogenesis and cholesterol efflux, but suppresses endoplasmic reticulum stress and inflammation. The goal of these studies was to dissect the effects of insulin on LXR action. We used antisense oligonucleotides to knock down Lxr alpha in mice with hepatocytespecific deletion of the insulin receptor and their controls. We found, surprisingly, that knock-out of the insulin receptor and knockdown of Lxr alpha produced equivalent, non-additive effects on the lipogenic genes. Thus, insulin was unable to induce the lipogenic genes in the absence of Lxr alpha, and LXR alpha was unable to induce the lipogenic genes in the absence of insulin. However, insulin was not required for LXR alpha to modulate the phospholipid profile, or to suppress genes in the ER stress or inflammation pathways. These data show that insulin is required specifically for the lipogenic effects of LXR alpha and that manipulation of the insulin signaling pathway could dissociate the beneficial effects of LXR on cholesterol efflux, inflammation, and ER stress from the negative effects on lipogenesis.
- Published
- 2016
- Full Text
- View/download PDF
27. Hepatic Insulin Signaling Is Required for Obesity-Dependent Expression of SREBP-1c mRNA but Not for Feeding-Dependent Expression
- Author
-
Ji Miao, Bhavapriya Vaitheesvaran, Yanning Wang, Sudha B. Biddinger, Matthew D. Hirschey, Robert V. Farese, Joel T. Haas, Fabienne Foufelle, Rosanne M. Crooke, Dipanjan Chanda, Irwin J. Kurland, Enpeng Zhao, Mary E. Haas, and Mark J. Graham
- Subjects
Male ,Physiology ,medicine.medical_treatment ,Gene Expression ,Mice, Obese ,mTORC1 ,Type 2 diabetes ,Mice ,0302 clinical medicine ,Insulin ,Cells, Cultured ,0303 health sciences ,Gene knockdown ,biology ,Ketones ,Liver ,Gene Knockdown Techniques ,030220 oncology & carcinogenesis ,Lipogenesis ,Female ,RNA Interference ,Signal transduction ,Sterol Regulatory Element Binding Protein 1 ,Signal Transduction ,medicine.medical_specialty ,Mice, Transgenic ,Fructose ,Article ,03 medical and health sciences ,Internal medicine ,Dietary Carbohydrates ,medicine ,Animals ,Obesity ,Molecular Biology ,Triglycerides ,030304 developmental biology ,Cell Nucleus ,Membrane Proteins ,Cell Biology ,medicine.disease ,Receptor, Insulin ,Fatty Liver ,Mice, Inbred C57BL ,Insulin receptor ,Glucose ,Endocrinology ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,Hepatocytes ,biology.protein ,Steatosis - Abstract
SummaryDissecting the role of insulin in the complex regulation of triglyceride metabolism is necessary for understanding dyslipidemia and steatosis. Liver insulin receptor knockout (LIRKO) mice show that in the physiological context of feeding, hepatic insulin signaling is not required for the induction of mTORC1, an upstream activator of the lipogenic regulator, SREBP-1c. Feeding induces SREBP-1c mRNA in LIRKO livers, though not to the extent observed in controls. A high fructose diet also partially induces SREBP-1c and lipogenic gene expression in LIRKO livers. Insulin signaling becomes more important in the pathological context of obesity, as knockdown of the insulin receptor in ob/ob mice, a model of Type 2 diabetes, using antisense oligonucleotides, abolishes the induction of SREBP-1c and its targets by obesity and ameliorates steatosis. Thus, insulin-independent signaling pathways can partially compensate for insulin in the induction of SREBP-1c by feeding but the further induction by obesity/Type 2 diabetes is entirely dependent upon insulin.
- Published
- 2012
- Full Text
- View/download PDF
28. PKCδ regulates hepatic insulin sensitivity and hepatosteatosis in mice and humans
- Author
-
George L. King, Michael Leitges, Marcelo A. Mori, Olivier Bezy, Allison B. Goldfine, C. Ronald Kahn, Mary-Elizabeth Patti, Brice Emanuelli, Jussi Pihlajamäki, Jonathan Winnay, Thien T. Tran, Ryo Suzuki, Sudha B. Biddinger, and Joel T. Haas
- Subjects
Blood Glucose ,Male ,Aging ,medicine.medical_specialty ,Mice, 129 Strain ,medicine.medical_treatment ,Biology ,Diabetes Mellitus, Experimental ,Mice ,Insulin resistance ,Species Specificity ,Internal medicine ,Diabetes mellitus ,Glucose Intolerance ,Gene expression ,medicine ,Animals ,Humans ,Insulin ,Obesity ,Triglycerides ,Metabolic Syndrome ,Mice, Knockout ,Lipogenesis ,Gluconeogenesis ,Fasting ,General Medicine ,medicine.disease ,Dietary Fats ,Fatty Liver ,Mice, Inbred C57BL ,Protein Kinase C-delta ,Insulin receptor ,Endocrinology ,Liver ,Enzyme Induction ,biology.protein ,Female ,Insulin Resistance ,Metabolic syndrome ,Research Article - Abstract
C57BL/6J and 129S6/Sv (B6 and 129) mice differ dramatically in their susceptibility to developing diabetes in response to diet- or genetically induced insulin resistance. A major locus contributing to this difference has been mapped to a region on mouse chromosome 14 that contains the gene encoding PKCδ. Here, we found that PKCδ expression in liver was 2-fold higher in B6 versus 129 mice from birth and was further increased in B6 but not 129 mice in response to a high-fat diet. PRKCD gene expression was also elevated in obese humans and was positively correlated with fasting glucose and circulating triglycerides. Mice with global or liver-specific inactivation of the Prkcd gene displayed increased hepatic insulin signaling and reduced expression of gluconeogenic and lipogenic enzymes. This resulted in increased insulin-induced suppression of hepatic gluconeogenesis, improved glucose tolerance, and reduced hepatosteatosis with aging. Conversely, mice with liver-specific overexpression of PKCδ developed hepatic insulin resistance characterized by decreased insulin signaling, enhanced lipogenic gene expression, and hepatosteatosis. Therefore, changes in the expression and regulation of PKCδ between strains of mice and in obese humans play an important role in the genetic risk of hepatic insulin resistance, glucose intolerance, and hepatosteatosis; and thus PKCδ may be a potential target in the treatment of metabolic syndrome.
- Published
- 2011
- Full Text
- View/download PDF
29. Diabetes and Insulin in Regulation of Brain Cholesterol Metabolism
- Author
-
Kevin Y. Lee, Ryo Suzuki, C. Ronald Kahn, Sudha B. Biddinger, Enxuan Jing, Thomas J. Montine, Jeffrey G. McDonald, and Suzanne Craft
- Subjects
medicine.medical_specialty ,Physiology ,medicine.medical_treatment ,Genetic Vectors ,Hypothalamus ,Biology ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Mice, Inbred NOD ,Internal medicine ,Diabetes mellitus ,Diabetes Mellitus ,medicine ,Animals ,Insulin ,Molecular Biology ,030304 developmental biology ,Neurons ,Regulation of gene expression ,Analysis of Variance ,0303 health sciences ,Gene knockdown ,Cholesterol ,Lentivirus ,Cell Biology ,medicine.disease ,Biosynthetic Pathways ,Mice, Inbred C57BL ,Endocrinology ,medicine.anatomical_structure ,Gene Expression Regulation ,chemistry ,Gene Knockdown Techniques ,Neuroglia ,lipids (amino acids, peptides, and proteins) ,Sterol regulatory element-binding protein 2 ,030217 neurology & neurosurgery ,Sterol Regulatory Element Binding Protein 2 - Abstract
SummaryThe brain is the most cholesterol-rich organ in the body, most of which comes from in situ synthesis. Here we demonstrate that in insulin-deficient diabetic mice, there is a reduction in expression of the major transcriptional regulator of cholesterol metabolism, SREBP-2, and its downstream genes in the hypothalamus and other areas of the brain, leading to a reduction in brain cholesterol synthesis and synaptosomal cholesterol content. These changes are due, at least in part, to direct effects of insulin to regulate these genes in neurons and glial cells and can be corrected by intracerebroventricular injections of insulin. Knockdown of SREBP-2 in cultured neurons causes a decrease in markers of synapse formation and reduction of SREBP-2 in the hypothalamus of mice using shRNA results in increased feeding and weight gain. Thus, insulin and diabetes can alter brain cholesterol metabolism, and this may play an important role in the neurologic and metabolic dysfunction observed in diabetes and other disease states.
- Published
- 2010
- Full Text
- View/download PDF
30. High Circulating Leptin Receptors with Normal Leptin Sensitivity in Liver-specific Insulin Receptor Knock-out (LIRKO) Mice
- Author
-
Christos S. Mantzoros, Tatsuya Kondo, C. Ronald Kahn, Rolf Gebhardt, Sudha B. Biddinger, Juergen Kratzsch, Efi Kokkotou, and Shmuel E. Cohen
- Subjects
Leptin ,medicine.medical_specialty ,medicine.medical_treatment ,Adipose tissue ,Receptors, Cell Surface ,Suppressor of Cytokine Signaling Proteins ,Polymerase Chain Reaction ,Biochemistry ,Mice ,Internal medicine ,Hyperinsulinemia ,medicine ,Animals ,Insulin ,Protein Isoforms ,RNA, Messenger ,SOCS3 ,Molecular Biology ,Mice, Knockout ,Leptin receptor ,biology ,Chemistry ,Gene Expression Profiling ,digestive, oral, and skin physiology ,Cell Biology ,medicine.disease ,Receptor, Insulin ,Insulin receptor ,Endocrinology ,Gene Expression Regulation ,Liver ,Suppressor of Cytokine Signaling 3 Protein ,biology.protein ,Receptors, Leptin ,hormones, hormone substitutes, and hormone antagonists ,Homeostasis ,Signal Transduction - Abstract
Liver-specific insulin receptor knock-out (LIRKO) mice display hyperinsulinemia, abnormal glucose metabolism, and progressive liver dysfunction. In addition, circulating leptin levels appear to be increased more than 10-fold. However, food intake, body weight, and adipose mass are not significantly altered in LIRKO mice compared with wild-type littermates. Using a ligand immunofunctional assay, we found that the apparent increase in circulating leptin in LIRKO mice is because of an 80-fold increased serum level of soluble leptin receptor. Gene expression analysis by microarray and real time PCR reveals the liver as the source of soluble leptin receptor in LIRKO mice, with an increase in expression of the short (Ob-Ra), long (Ob-Rb), and soluble (Ob-Re) forms of the leptin receptor. Direct control of leptin receptor expression by insulin could also be demonstrated in isolated hepatocytes from normal mice. Despite the markedly increased levels of leptin receptor in their circulation, LIRKO mice exhibit normal or even enhanced leptin sensitivity, as assessed by their physiological and molecular responses to exogenous leptin administration and their lower base-line hypothalamic levels of SOCS3 mRNA. Thus, insulin signaling in the liver plays an important role in control of leptin receptor expression and shedding. In the LIRKO mouse, this is lost, leading to markedly increased leptin receptors into the circulation. These high levels of circulating leptin receptor bind leptin and likely alter its clearance, but do not inhibit leptin action and may actually potentiate leptin action. In this manner, insulin signaling in liver plays an important role in leptin homeostasis and fine modulation of leptin action.
- Published
- 2007
- Full Text
- View/download PDF
31. The Role of Proprotein Convertase Subtilisin/Kexin Type 9 in Nephrotic Syndrome-Associated Hypercholesterolemia
- Author
-
Wan Hui Liao, Sudha B. Biddinger, Sarah D. de Ferranti, Valérie Schumacher, Amy E. Levenson, Xiaowei Sun, Joseph M. Rutkowski, Philipp E. Scherer, David J. Salant, and Mary E. Haas
- Subjects
medicine.medical_specialty ,Nephrotic Syndrome ,Hypercholesterolemia ,030232 urology & nephrology ,030204 cardiovascular system & hematology ,law.invention ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,law ,Physiology (medical) ,Internal medicine ,medicine ,Animals ,Humans ,Mice, Knockout ,Kidney ,Cholesterol ,business.industry ,Podocytes ,PCSK9 ,Subtilisin ,Proprotein convertase ,medicine.disease ,Lipids ,Recombinant Proteins ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Liver ,Recombinant DNA ,Kexin ,Proprotein Convertase 9 ,Cardiology and Cardiovascular Medicine ,business ,Nephrotic syndrome - Abstract
Background: In nephrotic syndrome, damage to the podocytes of the kidney produces severe hypercholesterolemia for which novel treatments are urgently needed. PCSK9 (proprotein convertase subtilisin/kexin type 9) has emerged as an important regulator of plasma cholesterol levels and therapeutic target. Here, we tested the role of PCSK9 in mediating the hypercholesterolemia of nephrotic syndrome. Methods: PCSK9 and plasma lipids were studied in nephrotic syndrome patients before and after remission of disease, mice with genetic ablation of the podocyte (Podocyte Apoptosis Through Targeted Activation of Caspase-8, Pod-ATTAC mice) and mice treated with nephrotoxic serum (NTS), which triggers immune-mediated podocyte damage. In addition, mice with hepatic deletion of Pcsk9 were treated with NTS to determine the contribution of PCSK9 to the dyslipidemia of nephrotic syndrome. Results: Patients with nephrotic syndrome showed a decrease in plasma cholesterol and plasma PCSK9 on remission of their disease ( P Pcsk9 ameliorated the effects of NTS on plasma lipids. Thus, in the presence of NTS, mice lacking hepatic Pcsk9 showed a 40% to 50% decrease in plasma cholesterol and triglycerides. Moreover, the ability of NTS treatment to increase the percentage of low-density lipoprotein–associated cholesterol (from 9% in vehicle-treated Flox mice to 47% after NTS treatment), was lost in mice with hepatic deletion of Pcsk9 (5% in both the presence and absence of NTS). Conclusions: Podocyte damage triggers marked inductions in plasma PCSK9, and knockout of Pcsk9 ameliorates dyslipidemia in a mouse model of nephrotic syndrome. These data suggest that PCSK9 inhibitors may be beneficial in patients with nephrotic syndrome–associated hypercholesterolemia.
- Published
- 2015
32. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis
- Author
-
Sudha B. Biddinger, Alisha V. Ling, Mark J. Graham, Clary B. Clish, Praveen V. Manthena, Mary E Gearing, Rosanne M. Crooke, Ryan M. Esquejo, Ji Miao, Kevin Croce, and David Vicent
- Subjects
Male ,medicine.medical_treatment ,General Physics and Astronomy ,FOXO1 ,030204 cardiovascular system & hematology ,Rats, Sprague-Dawley ,Mice ,chemistry.chemical_compound ,0302 clinical medicine ,Hyperlipidemia ,Insulin ,0303 health sciences ,Multidisciplinary ,Forkhead Box Protein O1 ,Forkhead Transcription Factors ,3. Good health ,Liver ,Gene Knockdown Techniques ,Oxygenases ,medicine.medical_specialty ,Blotting, Western ,Hyperlipidemias ,Flavin-containing monooxygenase ,In Vitro Techniques ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Insulin resistance ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Humans ,Hypoglycemic Agents ,Obesity ,RNA, Messenger ,Triglycerides ,030304 developmental biology ,Cholesterol ,Gene Expression Profiling ,Cholesterol, HDL ,Cholesterol, LDL ,General Chemistry ,Atherosclerosis ,medicine.disease ,Rats ,Endocrinology ,Diabetes Mellitus, Type 2 ,chemistry ,Hyperglycemia ,Metabolic control analysis ,Hepatocytes ,Insulin Resistance - Abstract
Despite the well-documented association between insulin resistance and cardiovascular disease, the key targets of insulin relevant to the development of cardiovascular disease are not known. Here, using non-biased profiling methods, we identify the enzyme flavin-containing monooxygenase 3 (Fmo3) to be a target of insulin. FMO3 produces trimethylamine N-oxide (TMAO), which has recently been suggested to promote atherosclerosis in mice and humans. We show that FMO3 is suppressed by insulin in vitro, increased in obese/insulin resistant male mice and increased in obese/insulin-resistant humans. Knockdown of FMO3 in insulin-resistant mice suppresses FoxO1, a central node for metabolic control, and entirely prevents the development of hyperglycaemia, hyperlipidemia and atherosclerosis. Taken together, these data indicate that FMO3 is required for FoxO1 expression and the development of metabolic dysfunction., The hepatic enzyme FMO3 has been linked to atherosclerosis. Here the authors show that FMO3 is upregulated in various models of diabetes and link FMO3 with key transcriptional regulators of hepatic glucose and cholesterol synthesis, thus proposing a mechanistic connection between diabetes and atherosclerosis.
- Published
- 2015
- Full Text
- View/download PDF
33. FROM MICE TO MEN: Insights into the Insulin Resistance Syndromes
- Author
-
Sudha B. Biddinger and C. Ronald Kahn
- Subjects
Mice, Knockout ,medicine.medical_specialty ,Physiology ,Disease ,Type 2 diabetes ,Biology ,medicine.disease ,Obesity ,Phenotype ,Receptor, Insulin ,Mice ,Insulin receptor ,Insulin resistance ,Endocrinology ,Internal medicine ,medicine ,biology.protein ,Animals ,Humans ,Insulin ,Insulin Resistance ,Receptor ,Dyslipidemia ,Signal Transduction - Abstract
▪ Abstract The insulin resistance syndrome refers to a constellation of findings, including glucose intolerance, obesity, dyslipidemia, and hypertension, that promote the development of type 2 diabetes, cardiovascular disease, cancer, and other disorders. Defining the pathophysiological links between insulin resistance, the insulin resistance syndrome, and its sequelae is critical to understanding and treating these disorders. Over the past decade, two approaches have provided important insights into how changes in insulin signaling produce the spectrum of phenotypes associated with insulin resistance. First, studies using tissue-specific knockouts or tissue-specific reconstitution of the insulin receptor in vivo in mice have enabled us to deconstruct the insulin resistance syndromes by dissecting the contributions of different tissues to the insulin-resistant state. Second, in vivo and in vitro studies of the complex network of insulin signaling have provided insight into how insulin resistance can develop in some pathways whereas insulin sensitivity is maintained in others. These data, taken together, give us a framework for understanding the relationship between insulin resistance and the insulin resistance syndromes.
- Published
- 2006
- Full Text
- View/download PDF
34. Diet rapidly and reproducibly alters the human gut microbiome
- Author
-
Peter J. Turnbaugh, Yug Varma, Lawrence A. David, Michael A. Fischbach, Benjamin E. Wolfe, Corinne F. Maurice, David B. Gootenberg, Rachel J. Dutton, A. Sloan Devlin, Rachel N. Carmody, Alisha V. Ling, Julie E. Button, and Sudha B. Biddinger
- Subjects
Adult ,Male ,Time Factors ,Firmicutes ,General Science & Technology ,Bilophila ,Article ,Oral and gastrointestinal ,Microbiology ,Bile Acids and Salts ,Feces ,Young Adult ,Vegetarian ,Humans ,Bacteroides ,Microbiome ,Herbivory ,Alistipes ,2. Zero hunger ,Multidisciplinary ,biology ,Bacteria ,Diet, Vegetarian ,Microbiota ,Gastrointestinal Microbiome ,Bacterial ,Gene Expression Regulation, Bacterial ,biology.organism_classification ,Inflammatory Bowel Diseases ,Dietary Fats ,Carnivory ,Diet ,Gastrointestinal Tract ,Bilophila wadsworthia ,Gene Expression Regulation ,Fermentation ,Food Microbiology ,Metagenome ,Enterotype ,Female ,Roseburia - Abstract
Long-term dietary intake influences the structure and activity of the trillions of microorganisms residing in the human gut, but it remains unclear how rapidly and reproducibly the human gut microbiome responds to short-term macronutrient change. Here we show that the short-term consumption of diets composed entirely of animal or plant products alters microbial community structure and overwhelms inter-individual differences in microbial gene expression. The animal-based diet increased the abundance of bile-tolerant microorganisms (Alistipes, Bilophila and Bacteroides) and decreased the levels of Firmicutes that metabolize dietary plant polysaccharides (Roseburia, Eubacterium rectale and Ruminococcus bromii). Microbial activity mirrored differences between herbivorous and carnivorous mammals, reflecting trade-offs between carbohydrate and protein fermentation. Foodborne microbes from both diets transiently colonized the gut, including bacteria, fungi and even viruses. Finally, increases in the abundance and activity of Bilophila wadsworthia on the animal-based diet support a link between dietary fat, bile acids and the outgrowth of microorganisms capable of triggering inflammatory bowel disease. In concert, these results demonstrate that the gut microbiome can rapidly respond to altered diet, potentially facilitating the diversity of human dietary lifestyles.
- Published
- 2014
35. The regulation of ApoB metabolism by insulin
- Author
-
Alan D. Attie, Sudha B. Biddinger, and Mary E. Haas
- Subjects
medicine.medical_specialty ,Apolipoprotein B ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Type 2 diabetes ,Lipoproteins, VLDL ,digestive system ,Article ,Endocrinology ,Insulin resistance ,Diabetes mellitus ,Internal medicine ,medicine ,Animals ,Humans ,Insulin ,Apolipoproteins B ,biology ,Chemistry ,nutritional and metabolic diseases ,medicine.disease ,LRP1 ,Cardiovascular Diseases ,LDL receptor ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Insulin Resistance ,Lipoprotein - Abstract
The leading cause of death in diabetic patients is cardiovascular disease. Apolipoprotein B (ApoB)-containing lipoprotein particles, which are secreted and cleared by the liver, are essential for the development of atherosclerosis. Insulin plays a key role in the regulation of ApoB. Insulin decreases ApoB secretion by promoting ApoB degradation in the hepatocyte. In parallel, insulin promotes clearance of circulating ApoB particles by the liver via the low-density lipoprotein receptor (LDLR), LDLR-related protein 1 (LRP1), and heparan sulfate proteoglycans (HSPGs). Consequently, the insulin-resistant state of type 2 diabetes (T2D) is associated with increased secretion and decreased clearance of ApoB. Here, we review the mechanisms by which insulin controls the secretion and uptake of ApoB in normal and diabetic livers.
- Published
- 2013
36. Transcriptional activation of apolipoprotein CIII expression by glucose may contribute to diabetic dyslipidemia
- Author
-
Emmanuelle Vallez, Luc Van Gaal, Ilse Mertens, Bart Staels, Sven Francque, An Verrijken, Daniel Duran-Sandoval, Jan Albert Kuivenhoven, Isabelle Berard, Janne Prawitt, Folkert Kuipers, Carolina Huaman Samanez, Sudha B. Biddinger, Anne Muhr-Tailleux, Joel T. Haas, Sandrine Caron, Gisèle Mautino, Marja-Riitta Taskinen, Center for Liver, Digestive and Metabolic Diseases (CLDM), Cardiovascular Centre (CVC), Lifestyle Medicine (LM), Vascular Ageing Programme (VAP), ACS - Amsterdam Cardiovascular Sciences, and Experimental Vascular Medicine
- Subjects
Blood Glucose ,Male ,Time Factors ,medicine.medical_treatment ,nuclear receptors ,Receptors, Cytoplasmic and Nuclear ,Type 2 diabetes ,030204 cardiovascular system & hematology ,APOC-III ,Mice ,0302 clinical medicine ,Insulin ,Promoter Regions, Genetic ,Cells, Cultured ,Heat-Shock Proteins ,Liver X Receptors ,Mice, Knockout ,0303 health sciences ,type II diabetes ,INSULIN-RESISTANCE ,PLASMA ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Fatty liver ,RNA-Binding Proteins ,Middle Aged ,Orphan Nuclear Receptors ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,3. Good health ,Up-Regulation ,A-I ,Hepatocyte Nuclear Factor 4 ,RECEPTOR LXR ,RNA Interference ,Cardiology and Cardiovascular Medicine ,Adult ,Transcriptional Activation ,medicine.medical_specialty ,C-III LEVELS ,LOW-DENSITY-LIPOPROTEIN ,Carbohydrate metabolism ,Biology ,Transfection ,lipids ,Diabetes Complications ,03 medical and health sciences ,Insulin resistance ,Internal medicine ,medicine ,Animals ,Humans ,Obesity ,RNA, Messenger ,Liver X receptor ,030304 developmental biology ,Dyslipidemias ,Apolipoprotein C-III ,GENE-TRANSCRIPTION ,medicine.disease ,ELEMENT-BINDING PROTEIN ,Receptor, Insulin ,Rats ,Endocrinology ,Glucose ,Hepatocyte nuclear factor 4 ,Diabetes Mellitus, Type 2 ,Hepatocytes ,Farnesoid X receptor ,Human medicine ,apolipoproteins ,metabolism ,TRIGLYCERIDE-RICH LIPOPROTEINS ,Transcription Factors - Abstract
Objective— Hypertriglyceridemia and fatty liver are common in patients with type 2 diabetes, but the factors connecting alterations in glucose metabolism with plasma and liver lipid metabolism remain unclear. Apolipoprotein CIII (apoCIII), a regulator of hepatic and plasma triglyceride metabolism, is elevated in type 2 diabetes. In this study, we analyzed whether apoCIII is affected by altered glucose metabolism. Methods and Results— Liver-specific insulin receptor–deficient mice display lower hepatic apoCIII mRNA levels than controls, suggesting that factors other than insulin regulate apoCIII in vivo. Glucose induces apoCIII transcription in primary rat hepatocytes and immortalized human hepatocytes via a mechanism involving the transcription factors carbohydrate response element–binding protein and hepatocyte nuclear factor-4α. ApoCIII induction by glucose is blunted by treatment with agonists of farnesoid X receptor and peroxisome proliferator-activated receptor-α but not liver X receptor, ie, nuclear receptors controlling triglyceride metabolism. Moreover, in obese humans, plasma apoCIII protein correlates more closely with plasma fasting glucose and glucose excursion after oral glucose load than with insulin. Conclusion— Glucose induces apoCIII transcription, which may represent a mechanism linking hyperglycemia, hypertriglyceridemia, and cardiovascular disease in type 2 diabetes.
- Published
- 2011
37. Insulin Resistance in the Metabolic Syndrome
- Author
-
Brice Emanuelli and Sudha B. Biddinger
- Subjects
medicine.medical_specialty ,business.industry ,Hypertriglyceridemia ,Fatty liver ,nutritional and metabolic diseases ,Disease ,medicine.disease ,Polycystic ovarian disease ,Insulin resistance ,Internal medicine ,medicine ,Steatohepatitis ,Metabolic syndrome ,business ,National Cholesterol Education Program - Abstract
In 1988, Gerald Reaven coined the term “Syndrome X” to describe a complex of metabolic abnormalities, including glucose intolerance, hypertriglyceridemia and reduced levels of HDL-cholesterol, present in individuals at increased risk for cardiovascular disease [1]. Since then, attempts to quantify cardiovascular disease risk have led to the development of clinical criteria for the diagnosis of this syndrome, now known as the “metabolic syndrome” or “insulin resistance syndrome”. Although these criteria continue to evolve, those put forth by the National Cholesterol Education Program (NCEP), World Health Organization (WHO), European Group for the Study of Insulin Resistance (EGIR), International Diabetes Federation (IDF) and American Association of Clinical Endocrinologists (AACE), all include hyperglycemia, hypertriglyceridemia, low HDL-cholesterol and hypertension (reviewed in [2)] (Table 1). It is clear now that the metabolic syndrome is associated with many diseases in addition to cardiovascular disease. These include cholesterol gallstones, non-alcoholic fatty liver disease, which ranges from benign steatosis to non-alcholic steatohepatitis (NASH), polycystic ovary disease (PCOS) and neurodegenerative disease.
- Published
- 2010
- Full Text
- View/download PDF
38. You deserve what you eat: Lessons learned from the study of the Melanin-Concentrating Hormone (MCH) - deficient mice
- Author
-
Yan Wang, Sudha B. Biddinger, Dimitrios C. Ziogas, and Efi Kokkotou
- Subjects
Blood Glucose ,Leptin ,Male ,medicine.medical_specialty ,Saturated fat ,medicine.medical_treatment ,media_common.quotation_subject ,Biology ,Article ,Proinflammatory cytokine ,Eating ,Mice ,Insulin resistance ,Downregulation and upregulation ,Thinness ,Internal medicine ,medicine ,Animals ,Insulin ,media_common ,Adiposity ,Melanins ,Mice, Knockout ,Hypothalamic Hormones ,Lipogenesis ,Body Weight ,Gastroenterology ,Gluconeogenesis ,Appetite ,Glucose Tolerance Test ,medicine.disease ,Lipid Metabolism ,Obesity ,Dietary Fats ,Immunity, Innate ,Toll-Like Receptor 4 ,Pituitary Hormones ,Endocrinology ,Adipose Tissue ,Gene Expression Regulation ,Liver ,Hormone ,Signal Transduction - Abstract
Objectives Diet plays a crucial role in the development of obesity and insulin resistance via multiple mechanisms. Saturated fatty acids can directly trigger tissue specific proinflammatory pathways via Toll-like receptor-4 (TLR4)-dependent mechanisms. Moreover, diet can change the gut microbiome and increase gut permeability. However, very few studies have addressed the obesity-independent role of diet. Dissecting the effects of diet from those of obesity per se will enhance our understanding of the underlying pathogenesis, and, at the translational level, advance our treatment approaches for obesity and its co-morbidities. Methods Melanin-concentrating hormone (MCH) is an important regulator of appetite and energy balance. MCH-deficient mice are resistant to diet-induced obesity, primarily due to increased locomotor activity. We took advantage of the unique phenotype of these mice to examine the metabolic and inflammatory consequences of a 15-week consumption of a diet high in saturated fat. Results MCH-deficient mice chronically exposed to a high-fat diet gain less weight compared to their wild-type littermates, despite similar food intake, and are protected from hepatosteatosis. They also lack obesity-associated upregulation of serum leptin and insulin levels and have improved total body insulin sensitivity. Nevertheless, we found indistinguishable liver-specific innate immune responses in both genotypes associated with high-fat feeding, which involved activation of TLR4 and its downstream effectors, MyD88, p38 MAP kinase and STAT-3. Conclusions Our findings indicate that high-fat feeding is deleterious to the liver, independently of the obesity status. They also suggest that MCH is not necessary for the TLR4-dependent immune response triggered by the high-fat diet.
- Published
- 2010
39. Regulation of glucose homeostasis through a XBP-1-FoxO1 interaction
- Author
-
Yingjiang Zhou, Jason Chung, Cheng Sun, Morris F. White, Jaemin Lee, Umut Ozcan, Sudha B. Biddinger, Sang Won Park, Simon J. Fisher, Justin Lee, and Candace M. Reno
- Subjects
Blood Glucose ,X-Box Binding Protein 1 ,medicine.medical_specialty ,FOXO1 ,Regulatory Factor X Transcription Factors ,Biology ,Carbohydrate metabolism ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mice ,Insulin resistance ,Internal medicine ,medicine ,Glucose homeostasis ,Animals ,Homeostasis ,Phosphorylation ,Transcription factor ,Forkhead Box Protein O1 ,Endoplasmic reticulum ,Hydrolysis ,Forkhead Transcription Factors ,General Medicine ,medicine.disease ,Receptor, Insulin ,DNA-Binding Proteins ,Insulin receptor ,Disease Models, Animal ,Endocrinology ,Glucose ,Liver ,Mutation ,biology.protein ,Signal transduction ,Insulin Resistance ,Signal Transduction ,Transcription Factors - Abstract
To date, the only known role of the spliced form of X-box-binding protein-1 (XBP-1s) in metabolic processes has been its ability to act as a transcription factor that regulates the expression of genes that increase the endoplasmic reticulum (ER) folding capacity, thereby improving insulin sensitivity. Here we show that XBP-1s interacts with the Forkhead box O1 (FoxO1) transcription factor and directs it toward proteasome-mediated degradation. Given this new insight, we tested modest hepatic overexpression of XBP-1s in vivo in mouse models of insulin deficiency or insulin resistance and found it improved serum glucose concentrations, even without improving insulin signaling or ER folding capacity. The notion that XBP-1s can act independently of its role in the ER stress response is further supported by our finding that in the severely insulin resistant ob/ob mouse strain a DNA-binding-defective mutant of XBP-1s, which does not have the ability to increase ER folding capacity, is still capable of reducing serum glucose concentrations and increasing glucose tolerance. Our results thus provide the first evidence to our knowledge that XBP-1s, through its interaction with FoxO1, can bypass hepatic insulin resistance independent of its effects on ER folding capacity, suggesting a new therapeutic approach for the treatment of type 2 diabetes.
- Published
- 2010
40. Dissecting the role of insulin resistance in the metabolic syndrome
- Author
-
Sudha B. Biddinger and Joel T. Haas
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,FOXO1 ,Biology ,Article ,Insulin resistance ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Insulin ,Molecular Biology ,Metabolic Syndrome ,Nutrition and Dietetics ,Hypertriglyceridemia ,Cell Biology ,medicine.disease ,Insulin receptor ,Endocrinology ,biology.protein ,Metabolic syndrome ,Steatosis ,Cardiology and Cardiovascular Medicine ,Dyslipidemia ,Signal Transduction - Abstract
Purpose of Review—Over twenty years ago, insulin resistance was postulated to play a central role in the pathogenesis of the metabolic syndrome. However, this has been difficult to prove, leading to a great deal of controversy within the field. Recent studies in mice and humans with genetic defects in insulin signaling have allowed us, for the first time, to dissect which features of the metabolic syndrome can be caused by insulin resistance. Recent Findings—Mice with liver specific knockout of the insulin receptor (LIRKO) show that hepatic insulin resistance can produce (1) hyperglycemia; (2) increased Apob secretion and atherosclerosis; and (3) increased biliary cholesterol secretion and cholesterol gallstones. Many of these changes may be due to dis-inhibition of the transcription factor, FoxO1. Yet, neither LIRKO mice nor humans with insulin receptor mutations develop the hypertriglyceridemia or hepatic steatosis associated with the metabolic syndrome. Conclusion—These data point to a central role for insulin resistance in the pathogenesis of the metabolic syndrome, as hyperglycemia, atherosclerosis, and cholesterol gallstones can all be caused by insulin resistance. However, hypertriglyceridemia and hepatic steatosis are not due directly to insulin resistance, and should be considered pathogenically distinct features of the metabolic syndrome.
- Published
- 2009
41. SIRT3 regulates mitochondrial fatty-acid oxidation by reversible enzyme deacetylation
- Author
-
Asish K. Saha, Eric S. Goetzman, Neil B. Ruderman, Tadahiro Shimazu, Carrie A. Grueter, C. Ronald Kahn, Olga Ilkayeva, Christopher B. Newgard, Matthew D. Hirschey, Robert V. Farese, Enxuan Jing, Yu Li, James R. Bain, Charles A. Harris, Robert Stevens, Eric Verdin, Sudha B. Biddinger, Bjoern Schwer, David B. Lombard, and Frederick W. Alt
- Subjects
Male ,SIRT5 ,SIRT3 ,Calorie restriction ,Mitochondrion ,Mass Spectrometry ,Article ,Cell Line ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Adenosine Triphosphate ,Adipose Tissue, Brown ,Carnitine ,Sirtuin 3 ,Animals ,Humans ,Triglycerides ,030304 developmental biology ,Caloric Restriction ,0303 health sciences ,Multidisciplinary ,biology ,Acyl-CoA Dehydrogenase, Long-Chain ,Fatty Acids ,Acyl CoA dehydrogenase ,Acetylation ,Fasting ,Hypoglycemia ,Mitochondria ,Up-Regulation ,Cold Temperature ,Biochemistry ,Liver ,Mitochondrial matrix ,Sirtuin ,biology.protein ,NAD+ kinase ,Oxidation-Reduction ,030217 neurology & neurosurgery ,Body Temperature Regulation - Abstract
Sirtuins are NAD(+)-dependent protein deacetylases. They mediate adaptive responses to a variety of stresses, including calorie restriction and metabolic stress. Sirtuin 3 (SIRT3) is localized in the mitochondrial matrix, where it regulates the acetylation levels of metabolic enzymes, including acetyl coenzyme A synthetase 2 (refs 1, 2). Mice lacking both Sirt3 alleles appear phenotypically normal under basal conditions, but show marked hyperacetylation of several mitochondrial proteins. Here we report that SIRT3 expression is upregulated during fasting in liver and brown adipose tissues. During fasting, livers from mice lacking SIRT3 had higher levels of fatty-acid oxidation intermediate products and triglycerides, associated with decreased levels of fatty-acid oxidation, compared to livers from wild-type mice. Mass spectrometry of mitochondrial proteins shows that long-chain acyl coenzyme A dehydrogenase (LCAD) is hyperacetylated at lysine 42 in the absence of SIRT3. LCAD is deacetylated in wild-type mice under fasted conditions and by SIRT3 in vitro and in vivo; and hyperacetylation of LCAD reduces its enzymatic activity. Mice lacking SIRT3 exhibit hallmarks of fatty-acid oxidation disorders during fasting, including reduced ATP levels and intolerance to cold exposure. These findings identify acetylation as a novel regulatory mechanism for mitochondrial fatty-acid oxidation and demonstrate that SIRT3 modulates mitochondrial intermediary metabolism and fatty-acid use during fasting.
- Published
- 2008
42. Hepatic insulin resistance directly promotes formation of cholesterol gallstones
- Author
-
Olivier Bezy, Martin C. Carey, Joel T. Haas, Sudha B. Biddinger, Wenwei Zhang, C. Ronald Kahn, Enxuan Jing, Bian B Yu, and Terry G. Unterman
- Subjects
Male ,medicine.medical_specialty ,CYP7B1 ,medicine.drug_class ,Lipoproteins ,Cytochrome P450 Family 7 ,Receptors, Cytoplasmic and Nuclear ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Bile Acids and Salts ,Cholesterol, Dietary ,chemistry.chemical_compound ,Mice ,Insulin resistance ,Cholelithiasis ,Internal medicine ,medicine ,Animals ,ATP Binding Cassette Transporter, Subfamily G, Member 5 ,Metabolic Syndrome ,Mice, Knockout ,Bile acid ,Cholesterol ,Forkhead Box Protein O1 ,ATP Binding Cassette Transporter, Subfamily G, Member 8 ,Forkhead Transcription Factors ,General Medicine ,Gallstones ,medicine.disease ,Receptor, Insulin ,DNA-Binding Proteins ,Insulin receptor ,Endocrinology ,chemistry ,Liver ,Models, Animal ,Steroid Hydroxylases ,biology.protein ,Farnesoid X receptor ,ATP-Binding Cassette Transporters ,Metabolic syndrome ,Insulin Resistance ,Transcription Factors - Abstract
Despite the well-documented association between gallstones and the metabolic syndrome1,2, the mechanistic links between these two disorders remain unknown. Here we demonstrate that mice with isolated hepatic insulin resistance created by liver-specific disruption of the insulin receptor (LIRKO mice) are markedly predisposed towards cholesterol gallstone formation due to at least two distinct mechanisms. Disinhibition of the forkhead transcription factor FoxO1 increases expression of the biliary cholesterol transporters Abcg5 and Abcg8, resulting in an increase in biliary cholesterol secretion. Hepatic insulin resistance also decreases expression of the bile acid synthetic enzymes, particularly Cyp7b1, and produces partial resistance to the farnesoid X receptor (FXR), leading to a lithogenic bile salt profile. As a result, after only one week on a lithogenic diet, 36% of LIRKO mice develop gallstones and by 12 weeks, 100% have gallstones. Thus, hepatic insulin resistance provides the critical link between the metabolic syndrome and increased gallstone susceptibility.
- Published
- 2008
43. Regulation of energy substrate utilization and hepatic insulin sensitivity by phosphatidylcholine transfer protein/StarD2
- Author
-
Alessandro Pocai, Luciano Rossetti, Sudha B. Biddinger, David E. Cohen, Hua Li, Lauren T. Glenz, Erez F. Scapa, Roger Gutierrez-Juarez, Keishi Kanno, Linda A. Jelicks, and Michele K. Wu
- Subjects
Blood Glucose ,medicine.medical_specialty ,Glycogenolysis ,Phospholipid ,Cell Culture Techniques ,Oxidative phosphorylation ,Fatty Acids, Nonesterified ,Biochemistry ,chemistry.chemical_compound ,Mice ,Internal medicine ,Genetics ,medicine ,Animals ,Insulin ,RNA, Messenger ,Phospholipid Transfer Proteins ,Molecular Biology ,Fatty acid synthesis ,Crosses, Genetic ,Triglycerides ,chemistry.chemical_classification ,Mice, Knockout ,Triglyceride ,Phosphatidylcholine transfer protein ,Fatty acid ,Glucose Tolerance Test ,Lipids ,Endocrinology ,chemistry ,Gluconeogenesis ,Gene Expression Regulation ,Liver ,Glucose Clamp Technique ,Hepatocytes ,Carrier Proteins ,Energy Metabolism ,Biotechnology - Abstract
Phosphatidylcholine transfer protein (PC-TP, also known as StarD2) is a highly specific intracellular lipid binding protein with accentuated expression in oxidative tissues. Here we show that decreased plasma concentrations of glucose and free fatty acids in fasting PC-TP-deficient (Pctp(-/-)) mice are attributable to increased hepatic insulin sensitivity. In hyperinsulinemic-euglycemic clamp studies, Pctp(-/-) mice exhibited profound reductions in hepatic glucose production, gluconeogenesis, glycogenolysis, and glucose cycling. These changes were explained in part by the lack of PC-TP expression in liver per se and in part by marked alterations in body fat composition. Reduced respiratory quotients in Pctp(-/-) mice were indicative of preferential fatty acid utilization for energy production in oxidative tissues. In the setting of decreased hepatic fatty acid synthesis, increased clearance rates of dietary triglycerides and increased hepatic triglyceride production rates reflected higher turnover in Pctp(-/-) mice. Collectively, these data support a key biological role for PC-TP in the regulation of energy substrate utilization.
- Published
- 2008
44. Hepatic insulin resistance is sufficient to produce dyslipidemia and susceptibility to atherosclerosis
- Author
-
Martin C. Carey, C. Ronald Kahn, Sudha B. Biddinger, Joel T. Haas, Christian Rask-Madsen, Gregory Stephanopoulos, Erez F. Scapa, Antonio Hernandez-Ono, David E. Cohen, Chhavi Agarwal, Henry N. Ginsberg, Ryo Suzuki, George L. King, and Jose O. Aleman
- Subjects
Very low-density lipoprotein ,medicine.medical_specialty ,Apolipoprotein B ,Physiology ,Lipoproteins ,Hypercholesterolemia ,HUMDISEASE ,030209 endocrinology & metabolism ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Insulin resistance ,Internal medicine ,Insulin ,Medicine ,Animals ,Molecular Biology ,030304 developmental biology ,Dyslipidemias ,Hypertriglyceridemia ,2. Zero hunger ,Mice, Knockout ,0303 health sciences ,biology ,business.industry ,Cholesterol ,Liver Diseases ,Gluconeogenesis ,Cell Biology ,medicine.disease ,Atherosclerosis ,Receptor, Insulin ,Insulin receptor ,Endocrinology ,Diabetes Mellitus, Type 2 ,chemistry ,SIGNALING ,Hyperglycemia ,biology.protein ,lipids (amino acids, peptides, and proteins) ,Disease Susceptibility ,Metabolic syndrome ,Insulin Resistance ,business ,Dyslipidemia ,Lipoprotein - Abstract
SummaryInsulin resistance plays a central role in the development of the metabolic syndrome, but how it relates to cardiovascular disease remains controversial. Liver insulin receptor knockout (LIRKO) mice have pure hepatic insulin resistance. On a standard chow diet, LIRKO mice have a proatherogenic lipoprotein profile with reduced high-density lipoprotein (HDL) cholesterol and very low-density lipoprotein (VLDL) particles that are markedly enriched in cholesterol. This is due to increased secretion and decreased clearance of apolipoprotein B-containing lipoproteins, coupled with decreased triglyceride secretion secondary to increased expression of Pgc-1β (Ppargc-1b), which promotes VLDL secretion, but decreased expression of Srebp-1c (Srebf1), Srebp-2 (Srebf2), and their targets, the lipogenic enzymes and the LDL receptor. Within 12 weeks on an atherogenic diet, LIRKO mice show marked hypercholesterolemia, and 100% of LIRKO mice, but 0% of controls, develop severe atherosclerosis. Thus, insulin resistance at the level of the liver is sufficient to produce the dyslipidemia and increased risk of atherosclerosis associated with the metabolic syndrome.
- Published
- 2007
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.