20 results on '"Peter-James H. Zushin"'
Search Results
2. G protein-coupled receptor 151 regulates glucose metabolism and hepatic gluconeogenesis
- Author
-
Ewa Bielczyk-Maczynska, Meng Zhao, Peter-James H. Zushin, Theresia M. Schnurr, Hyun-Jung Kim, Jiehan Li, Pratima Nallagatla, Panjamaporn Sangwung, Chong Y. Park, Cameron Cornn, Andreas Stahl, Katrin J. Svensson, and Joshua W. Knowles
- Subjects
Science - Abstract
Rare variants in the G-protein coupled receptor 151 (GPR151) gene in humans are associated with lower odds ratio for type 2 diabetes, but the mechanism behind this association has remained elusive. Here, the authors show that GPR151 regulates the process of hepatic gluconeogenesis and affects whole-body glucose metabolism.
- Published
- 2022
- Full Text
- View/download PDF
3. Generation of two human iPSC lines with Exon 3 mutations in BCL2-Associated Athanogene 3 (BAG3) from dilated cardiomyopathy patients
- Author
-
Peter-James H. Zushin, Yang Zhou, Audrey Li, Euan A. Ashley, Matthew T. Wheeler, and Joseph C. Wu
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Dilated cardiomyopathies (DCM) are one of the main causes of heart failure as one ages. BAG3 is a chaperone protein that is heavily implicated in the development of DCM and speed of progression toward heart failure. Here we generate two human iPSC lines from individuals with mutations in exon 3 of BAG3 and provide validation of their pluripotency and ability to differentiate toward the three primary germ layers. These two cell lines can help our understanding of BAG3 and its role in DCM by providing a good model for BAG3 inactivation and insufficiency.
- Published
- 2023
- Full Text
- View/download PDF
4. CoQ Regulates Brown Adipose Tissue Respiration and Uncoupling Protein 1 Expression
- Author
-
Ching-Fang Chang, Amanda L. Gunawan, Irene Liparulo, Peter-James H. Zushin, Ambre M. Bertholet, Yuriy Kirichok, and Andreas Stahl
- Subjects
Coenzyme Q ,brown adipose tissue ,mitochondrial function ,thermogenesis ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Coenzyme Q (CoQ, aka ubiquinone) is a key component of the mitochondrial electron transport chain (ETC) and membrane-incorporated antioxidant. CoQ10 deficiencies encompass a heterogeneous spectrum of clinical phenotypes and can be caused by hereditary mutations in the biosynthesis pathway or result from pharmacological interventions such as HMG-CoA Reductase inhibitors, and statins, which are widely used to treat hypercholesterolemia and prevent cardiovascular disease. How CoQ deficiency affects individual tissues and cell types, particularly mitochondrial-rich ones such as brown adipose tissue (BAT), has remained poorly understood. Here we show that pharmacological and genetic models of BAT CoQ deficiency show altered respiration that can only in part be explained by classical roles of CoQ in the respiration chain. Instead, we found that CoQ strongly impacts brown and beige adipocyte respiration via the regulation of uncoupling protein 1 (UCP1) expression. CoQ deficiency in BAT robustly decreases UCP1 protein levels and uncoupled respiration unexpectedly, resulting in increased inner mitochondrial membrane potential and decreased ADP/ATP ratios. Suppressed UCP1 expression was also observed in a BAT-specific in vivo model of CoQ deficiency and resulted in enhanced cold sensitivity. These findings demonstrate an as yet unappreciated role of CoQ in the transcriptional regulation of key thermogenic genes and functions.
- Published
- 2022
- Full Text
- View/download PDF
5. 4β-Hydroxycholesterol is a prolipogenic factor that promotes SREBP1c expression and activity through the liver X receptor
- Author
-
Ofer Moldavski, Peter-James H. Zushin, Charles A. Berdan, Robert J. Van Eijkeren, Xuntian Jiang, Mingxing Qian, Daniel S. Ory, Douglas F. Covey, Daniel K. Nomura, Andreas Stahl, Ethan J. Weiss, and Roberto Zoncu
- Subjects
oxysterol ,SREBP1c ,liver-X-Receptor ,de-novo-lipogenesis ,lipid droplets ,insulin ,Biochemistry ,QD415-436 - Abstract
Oxysterols are oxidized derivatives of cholesterol that play regulatory roles in lipid biosynthesis and homeostasis. How oxysterol signaling coordinates different lipid classes such as sterols and triglycerides remains incompletely understood. Here, we show that 4β-hydroxycholesterol (HC) (4β-HC), a liver and serum abundant oxysterol of poorly defined functions, is a potent and selective inducer of the master lipogenic transcription factor, SREBP1c, but not the related steroidogenic transcription factor SREBP2. By correlating tracing of lipid synthesis with lipogenic gene expression profiling, we found that 4β-HC acts as a putative agonist for the liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of the LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c and triggered de novo lipogenesis both in primary hepatocytes and in the mouse liver. In addition, 4β-HC acted in parallel to insulin-PI3K–dependent signaling to stimulate triglyceride synthesis and lipid-droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
- Published
- 2021
- Full Text
- View/download PDF
6. Phosphorylation of Beta-3 adrenergic receptor at serine 247 by ERK MAP kinase drives lipolysis in obese adipocytes
- Author
-
Shangyu Hong, Wei Song, Peter-James H. Zushin, Bingyang Liu, Mark P. Jedrychowski, Amir I. Mina, Zhaoming Deng, Dimitrije Cabarkapa, Jessica A. Hall, Colin J. Palmer, Hassan Aliakbarian, John Szpyt, Steven P. Gygi, Ali Tavakkoli, Lydia Lynch, Norbert Perrimon, and Alexander S. Banks
- Subjects
Internal medicine ,RC31-1245 - Abstract
Objective: The inappropriate release of free fatty acids from obese adipose tissue stores has detrimental effects on metabolism, but key molecular mechanisms controlling FFA release from adipocytes remain undefined. Although obesity promotes systemic inflammation, we find activation of the inflammation-associated Mitogen Activated Protein kinase ERK occurs specifically in adipose tissues of obese mice, and provide evidence that adipocyte ERK activation may explain exaggerated adipose tissue lipolysis observed in obesity. Methods and Results: We provide genetic and pharmacological evidence that inhibition of the MEK/ERK pathway in human adipose tissue, mice, and flies all effectively limit adipocyte lipolysis. In complementary findings, we show that genetic and obesity-mediated activation of ERK enhances lipolysis, whereas adipose tissue specific knock-out of ERK2, the exclusive ERK1/2 protein in adipocytes, dramatically impairs lipolysis in explanted mouse adipose tissue. In addition, acute inhibition of MEK/ERK signaling also decreases lipolysis in adipose tissue and improves insulin sensitivity in obese mice. Mice with decreased rates of adipose tissue lipolysis in vivo caused by either MEK or ATGL pharmacological inhibition were unable to liberate sufficient White Adipose Tissue (WAT) energy stores to fuel thermogenesis from brown fat during a cold temperature challenge. To identify a molecular mechanism controlling these actions, we performed unbiased phosphoproteomic analysis of obese adipose tissue at different time points following acute pharmacological MEK/ERK inhibition. MEK/ERK inhibition decreased levels of adrenergic signaling and caused de-phosphorylation of the β3-adrenergic receptor (β3AR) on serine 247. To define the functional implications of this phosphorylation, we showed that CRISPR/Cas9 engineered cells expressing wild type β3AR exhibited β3AR phosphorylation by ERK2 and enhanced lipolysis, but this was not seen when serine 247 of β3AR was mutated to alanine. Conclusion: Taken together, these data suggest that ERK activation in adipocytes and subsequent phosphorylation of the β3AR on S247 are critical regulatory steps in the enhanced adipocyte lipolysis of obesity. Keywords: ERK, Beta adrenergic receptor, Lipolysis, Free fatty acid, Obesity, Insulin resistance, Adipose, Fat
- Published
- 2018
- Full Text
- View/download PDF
7. Cdkal1, a type 2 diabetes susceptibility gene, regulates mitochondrial function in adipose tissue
- Author
-
Colin J. Palmer, Raphael J. Bruckner, Joao A. Paulo, Lawrence Kazak, Jonathan Z. Long, Amir I. Mina, Zhaoming Deng, Katherine B. LeClair, Jessica A. Hall, Shangyu Hong, Peter-James H. Zushin, Kyle L. Smith, Steven P. Gygi, Susan Hagen, David E. Cohen, and Alexander S. Banks
- Subjects
Cdkal1 ,GWAS ,Diabetes ,Adipose ,Mitochondria ,ANT1 ,Internal medicine ,RC31-1245 - Abstract
Objectives: Understanding how loci identified by genome wide association studies (GWAS) contribute to pathogenesis requires new mechanistic insights. Variants within CDKAL1 are strongly linked to an increased risk of developing type 2 diabetes and obesity. Investigations in mouse models have focused on the function of Cdkal1 as a tRNALys modifier and downstream effects of Cdkal1 loss on pro-insulin translational fidelity in pancreatic β−cells. However, Cdkal1 is broadly expressed in other metabolically relevant tissues, including adipose tissue. In addition, the Cdkal1 homolog Cdk5rap1 regulates mitochondrial protein translation and mitochondrial function in skeletal muscle. We tested whether adipocyte-specific Cdkal1 deletion alters systemic glucose homeostasis or adipose mitochondrial function independently of its effects on pro-insulin translation and insulin secretion. Methods: We measured mRNA levels of type 2 diabetes GWAS genes, including Cdkal1, in adipose tissue from lean and obese mice. We then established a mouse model with adipocyte-specific Cdkal1 deletion. We examined the effects of adipose Cdkal1 deletion using indirect calorimetry on mice during a cold temperature challenge, as well as by measuring cellular and mitochondrial respiration in vitro. We also examined brown adipose tissue (BAT) mitochondrial morphology by electron microscopy. Utilizing co-immunoprecipitation followed by mass spectrometry, we performed interaction mapping to identify new CDKAL1 binding partners. Furthermore, we tested whether Cdkal1 loss in adipose tissue affects total protein levels or accurate Lys incorporation by tRNALys using quantitative mass spectrometry. Results: We found that Cdkal1 mRNA levels are reduced in adipose tissue of obese mice. Using adipose-specific Cdkal1 KO mice (A-KO), we demonstrated that mitochondrial function is impaired in primary differentiated brown adipocytes and in isolated mitochondria from A-KO brown adipose tissue. A-KO mice displayed decreased energy expenditure during 4 °C cold challenge. Furthermore, mitochondrial morphology was highly abnormal in A-KO BAT. Surprisingly, we found that lysine codon representation was unchanged in Cdkal1 A-KO adipose tissue. We identified novel protein interactors of CDKAL1, including SLC25A4/ANT1, an inner mitochondrial membrane ADP/ATP translocator. ANT proteins can account for the UCP1-independent basal proton leak in BAT mitochondria. Cdkal1 A-KO mice had increased ANT1 protein levels in their white adipose tissue. Conclusions: Cdkal1 is necessary for normal mitochondrial morphology and function in adipose tissue. These results suggest that the type 2 diabetes susceptibility gene CDKAL1 has novel functions in regulating mitochondrial activity.
- Published
- 2017
- Full Text
- View/download PDF
8. Human iPSC‐Derived Proinflammatory Macrophages cause Insulin Resistance in an Isogenic White Adipose Tissue Microphysiological System
- Author
-
Lin Qi, Koji Matsuo, Ashley Pereira, Yue Tung Lee, Fenmiao Zhong, Yuchen He, Peter‐James H. Zushin, Marko Gröger, Aditi Sharma, Holger Willenbring, Edward C. Hsiao, and Andreas Stahl
- Subjects
Biomaterials ,General Materials Science ,General Chemistry ,Biotechnology - Published
- 2023
9. Coenzyme Q regulates UCP1 expression and thermogenesis through the integrated stress responses
- Author
-
Ching-Fang Chang, Amanda L. Gunawan, Peter-James H. Zushin, Greg A. Timblin, Ambre M. Bertholet, Biao Wang, Kaoru Sajio, Yuriy Kirichok, and Andreas Stahl
- Subjects
food and beverages - Abstract
Coenzyme Q (CoQ) is an essential component of mitochondrial respiration1 and required for thermogenic activity in brown adipose tissues2 (BAT). CoQ deficiency leads to a wide range of pathological manifestations3 but mechanistic consequences of CoQ deficiency in specific tissues such as BAT remain poorly understood. Here we show that pharmacological or genetic CoQ deficiency (50-75% reduction) in BAT leads to accumulation of cytosolic mitochondrial RNAs (mtRNAs) and activation of the eIF2α kinase PKR resulting in the induction of the integrated stress response (ISR) and suppression of UCP1 expression in an ATF4-dependent fashion. Surprisingly, despite diminished UCP1 levels, BAT CoQ deficiency increases whole-body metabolic rates at room temperature and thermoneutrality resulting in decreased weight gain on high fat diets (HFD). This mitohormesis like effect of BAT CoQ insufficiency is dependent on the ATF4-FGF21 axis in BAT revealing an unexpected role for CoQ in the modulation of whole-body energy expenditure with wide-ranging implications for primary and secondary CoQ deficiencies.
- Published
- 2022
10. An exercise-inducible metabolite that suppresses feeding and obesity
- Author
-
Veronica L. Li, Yang He, Kévin Contrepois, Hailan Liu, Joon T. Kim, Amanda L. Wiggenhorn, Julia T. Tanzo, Alan Sheng-Hwa Tung, Xuchao Lyu, Peter-James H. Zushin, Robert S. Jansen, Basil Michael, Kang Yong Loh, Andrew C. Yang, Christian S. Carl, Christian T. Voldstedlund, Wei Wei, Stephanie M. Terrell, Benjamin C. Moeller, Rick M. Arthur, Gareth A. Wallis, Koen van de Wetering, Andreas Stahl, Bente Kiens, Erik A. Richter, Steven M. Banik, Michael P. Snyder, Yong Xu, and Jonathan Z. Long
- Subjects
General Science & Technology ,Phenylalanine ,Cardiovascular ,Oral and gastrointestinal ,Article ,Mice ,Eating ,Physical Conditioning, Animal ,Diabetes Mellitus ,Animals ,Obesity ,Lactic Acid ,Metabolic and endocrine ,Adiposity ,Nutrition ,Cancer ,Multidisciplinary ,Animal ,Body Weight ,Diabetes ,Feeding Behavior ,Physical Conditioning ,Stroke ,Disease Models, Animal ,Glucose ,Diabetes Mellitus, Type 2 ,Ecological Microbiology ,Disease Models ,Energy Metabolism ,Type 2 - Abstract
Exercise confers protection against obesity, type 2 diabetes and other cardiometabolic diseases1-5. However, the molecular and cellular mechanisms that mediate the metabolic benefits of physical activity remain unclear6. Here we show that exercise stimulates the production of N-lactoyl-phenylalanine (Lac-Phe), a blood-borne signalling metabolite that suppresses feeding and obesity. The biosynthesis of Lac-Phe from lactate and phenylalanineoccurs in CNDP2+ cells, including macrophages,monocytes and otherimmune and epithelial cells localized to diverse organs. In diet-induced obese mice, pharmacological-mediated increases in Lac-Phe reduces food intake without affecting movement or energy expenditure. Chronic administration of Lac-Phe decreases adiposity and body weight and improves glucose homeostasis. Conversely, genetic ablation of Lac-Phe biosynthesis in mice increases food intake and obesity following exercise training. Last, large activity-inducible increases in circulating Lac-Phe are alsoobserved in humans and racehorses, establishing this metabolite as a molecular effector associated with physical activity across multiple activity modalities and mammalian species. These data define a conserved exercise-inducible metabolite that controls food intake and influences systemic energy balance.
- Published
- 2022
11. 4β-hydroxycholesterol is a pro-lipogenic factor that promotes SREBP1c expression and activity through Liver X-receptor
- Author
-
Daniel S. Ory, Peter-James H. Zushin, Xuntian Jiang, Mingxing Qian, Daniel K. Nomura, Ofer Moldavski, Ethan J. Weiss, Charles A. Berdan, Robert J. van Eijkeren, Douglas F. Covey, Andreas Stahl, and Roberto Zoncu
- Subjects
Oxysterol ,Chemistry ,Lipid biosynthesis ,Lipogenesis ,polycyclic compounds ,Transcriptional regulation ,Regulator ,lipids (amino acids, peptides, and proteins) ,Liver X receptor ,Beta (finance) ,Transcription factor ,Cell biology - Abstract
Oxysterols are oxidized derivatives of cholesterol that play signaling roles in lipid biosynthesis and homeostasis. Here we show that 4β-hydroxycholesterol (4β-HC), a liver and serum abundant oxysterol of poorly defined function, is a potent and selective inducer of the master lipogenic transcription factor, Sterol Regulatory Element Binding Protein 1c (SREBP1c), but not the related steroidogenic transcription factor SREBP2. Mechanistically, 4β-HC acts as a putative agonist for Liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c, and triggeredde novolipogenesis both in primary hepatocytes and in mouse liver. 4β-HC-acted in parallel to insulin-PI3K-dependent signaling to stimulate triglyceride synthesis and lipid droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
- Published
- 2020
12. Chronic arsenic exposure impairs adaptive thermogenesis in male C57BL/6J mice
- Author
-
Felicia Castriota, Rachael V. Phillips, Andreas Stahl, Martyn T. Smith, Jen-Chywan Wang, Alan Hubbard, Sylvia S. Sanchez, Peter-James H. Zushin, and Michele A. La Merrill
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Sodium arsenite ,Siloxanes ,Physiology ,Arsenites ,Endocrinology, Diabetes and Metabolism ,Adipose Tissue, White ,Subcutaneous Fat ,chemistry.chemical_element ,Receptors, Cell Surface ,White adipose tissue ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Adipose Tissue, Brown ,Physiology (medical) ,Internal medicine ,Brown adipose tissue ,Mitochondrial Precursor Protein Import Complex Proteins ,medicine ,Glucose homeostasis ,Animals ,Beta oxidation ,Arsenic ,Fatty acid metabolism ,Membrane Transport Proteins ,Thermogenesis ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Sodium Compounds ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Mitochondrial biogenesis ,030220 oncology & carcinogenesis ,Methacrylates ,Energy Metabolism ,Research Article - Abstract
The global prevalence of type 2 diabetes (T2D) has doubled since 1980. Human epidemiological studies support arsenic exposure as a risk factor for T2D, although the precise mechanism is unclear. We hypothesized that chronic arsenic ingestion alters glucose homeostasis by impairing adaptive thermogenesis, i.e., body heat production in cold environments. Arsenic is a pervasive environmental contaminant, with more than 200 million people worldwide currently exposed to arsenic-contaminated drinking water. Male C57BL/6J mice exposed to sodium arsenite in drinking water at 300 μg/L for 9 wk experienced significantly decreased metabolic heat production when acclimated to chronic cold tolerance testing, as evidenced by indirect calorimetry, despite no change in physical activity. Arsenic exposure increased total fat mass and subcutaneous inguinal white adipose tissue (iWAT) mass. RNA sequencing analysis of iWAT indicated that arsenic dysregulated mitochondrial processes, including fatty acid metabolism. Western blotting in WAT confirmed that arsenic significantly decreased TOMM20, a correlate of mitochondrial abundance; PGC1A, a master regulator of mitochondrial biogenesis; and, CPT1B, the rate-limiting step of fatty acid oxidation (FAO). Our findings show that chronic arsenic exposure impacts the mitochondrial proteins of thermogenic tissues involved in energy expenditure and substrate regulation, providing novel mechanistic evidence for arsenic’s role in T2D development.
- Published
- 2020
13. Obesity-linked PPARγ S273 phosphorylation promotes insulin resistance through Growth Differentiation Factor 3
- Author
-
Daniel G. Tenen, Joanna R. DiSpirito, Christopher Jacobs, Zhaoming Deng, Pratik Aryal, Barbara B. Kahn, Deepti Ramachandran, Chester W. Brown, Amir Mina, Peter-James H. Zushin, Bruce M. Spiegelman, Jessica A. Hall, Hyun Cheol Roh, Julia F. Charles, T. Belchior de Oliveira, Linus T.-Y. Tsai, Bingyang Liu, Gerald I. Shulman, Evan D. Rosen, Shangyu Hong, Colin J Palmer, and Alexander S. Banks
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Physiology ,Glucose uptake ,Peroxisome proliferator-activated receptor ,Adipose tissue ,Article ,Serine ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Insulin resistance ,Internal medicine ,Growth Differentiation Factor 3 ,medicine ,Animals ,Humans ,Obesity ,Phosphorylation ,Receptor ,Molecular Biology ,Alleles ,Cells, Cultured ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,Chemistry ,Cell Biology ,medicine.disease ,3. Good health ,Mice, Inbred C57BL ,PPAR gamma ,Insulin receptor ,030104 developmental biology ,Endocrinology ,Adipogenesis ,biology.protein ,Thiazolidinediones ,Ectopic expression ,Insulin Resistance ,030217 neurology & neurosurgery - Abstract
Overnutrition and obesity promote adipose tissue dysfunction, often leading to systemic insulin resistance. The thiazolidinediones (TZDs) are a potent class of insulin-sensitizing drugs and ligands of PPARγ that improve insulin sensitivity, but their use is limited due to significant side effects. Recently, we demonstrated a mechanism by which TZDs improve insulin sensitivity distinct from receptor agonism and adipogenesis: reversal of obesity-linked phosphorylation of PPARγ at Serine 273. However, the role of this modification has not been tested genetically. Here we demonstrate that mice encoding an allele of PPARγ which cannot be phosphorylated at S273 are protected from insulin resistance, without exhibiting differences in body weight or TZD-associated side effects. Indeed, hyperinsulinemic-euglycemic clamp experiments confirm improved insulin sensitivity, as evidenced by increased whole-body glucose uptake. RNA-seq experiments reveal PPARγ S273 phosphorylation specifically enhances transcription of Gdf3, a BMP family member. Ectopic expression of Gdf3 is sufficient to induce insulin resistance in lean, healthy mice. We find that Gdf3 can impact metabolism by inhibition of BMP signaling. Together, these results highlight the diabetogenic role of PPARγ S273 phosphorylation and focuses attention on a putative target, Gdf3.
- Published
- 2020
- Full Text
- View/download PDF
14. Phosphorylation of Beta-3 adrenergic receptor at serine 247 by ERK MAP kinase drives lipolysis in obese adipocytes
- Author
-
Steven P. Gygi, Wei Song, Shangyu Hong, Colin J. Palmer, Peter-James H. Zushin, Dimitrije Cabarkapa, Alexander S. Banks, Jessica A. Hall, John Szpyt, Norbert Perrimon, Bingyang Liu, Mark P. Jedrychowski, Zhaoming Deng, Hassan Aliakbarian, Amir I. Mina, Ali Tavakkoli, and Lydia Lynch
- Subjects
0301 basic medicine ,Beta-3 adrenergic receptor ,MAPK/ERK pathway ,lcsh:Internal medicine ,medicine.medical_specialty ,MAP Kinase Signaling System ,Lipolysis ,Adipocytes, White ,Adipose ,Adipose tissue ,White adipose tissue ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Beta adrenergic receptor ,Adipocyte ,Internal medicine ,Serine ,medicine ,Animals ,Humans ,Obesity ,Phosphorylation ,lcsh:RC31-1245 ,Molecular Biology ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Free fatty acid ,Insulin resistance ,3T3 Cells ,Cell Biology ,Mice, Inbred C57BL ,ERK ,Drosophila melanogaster ,030104 developmental biology ,Endocrinology ,chemistry ,Fat ,Receptors, Adrenergic, beta-3 ,Original Article ,Thermogenesis - Abstract
Objective The inappropriate release of free fatty acids from obese adipose tissue stores has detrimental effects on metabolism, but key molecular mechanisms controlling FFA release from adipocytes remain undefined. Although obesity promotes systemic inflammation, we find activation of the inflammation-associated Mitogen Activated Protein kinase ERK occurs specifically in adipose tissues of obese mice, and provide evidence that adipocyte ERK activation may explain exaggerated adipose tissue lipolysis observed in obesity. Methods and Results We provide genetic and pharmacological evidence that inhibition of the MEK/ERK pathway in human adipose tissue, mice, and flies all effectively limit adipocyte lipolysis. In complementary findings, we show that genetic and obesity-mediated activation of ERK enhances lipolysis, whereas adipose tissue specific knock-out of ERK2, the exclusive ERK1/2 protein in adipocytes, dramatically impairs lipolysis in explanted mouse adipose tissue. In addition, acute inhibition of MEK/ERK signaling also decreases lipolysis in adipose tissue and improves insulin sensitivity in obese mice. Mice with decreased rates of adipose tissue lipolysis in vivo caused by either MEK or ATGL pharmacological inhibition were unable to liberate sufficient White Adipose Tissue (WAT) energy stores to fuel thermogenesis from brown fat during a cold temperature challenge. To identify a molecular mechanism controlling these actions, we performed unbiased phosphoproteomic analysis of obese adipose tissue at different time points following acute pharmacological MEK/ERK inhibition. MEK/ERK inhibition decreased levels of adrenergic signaling and caused de-phosphorylation of the β3-adrenergic receptor (β3AR) on serine 247. To define the functional implications of this phosphorylation, we showed that CRISPR/Cas9 engineered cells expressing wild type β3AR exhibited β3AR phosphorylation by ERK2 and enhanced lipolysis, but this was not seen when serine 247 of β3AR was mutated to alanine. Conclusion Taken together, these data suggest that ERK activation in adipocytes and subsequent phosphorylation of the β3AR on S247 are critical regulatory steps in the enhanced adipocyte lipolysis of obesity., Graphical abstract, Highlights • Obese mice have increased ERK activity in white adipose tissues. • Blocking ERK reduces lipolysis in flies, in mice and in human adipose tissue. • Genetic activation of ERK promotes lipolysis. • Identification of in vivo ERK substrates by proteomics in obese adipose tissue. • ERK phosphorylates β3AR at Ser247 to enhance lipolysis in obesity.
- Published
- 2018
15. Probing Insulin Sensitivity with Metabolically Competent Human Stem Cell‐Derived White Adipose Tissue Microphysiological Systems
- Author
-
Lin Qi, Ching-Fang Chang, Peter-James H. Zushin, Yue Tung Lee, Suneil K. Koliwad, Diana L. Alba, and Andreas Stahl
- Subjects
medicine.medical_specialty ,Adipose Tissue, White ,medicine.medical_treatment ,Glucose uptake ,Adipose tissue ,White ,White adipose tissue ,Regenerative Medicine ,Article ,Biomaterials ,white adipose tissue ,Stem Cell Research - Nonembryonic - Human ,Internal medicine ,Lipid droplet ,Adipocytes ,human-induced pluripotent stem cells ,medicine ,Humans ,Insulin ,insulin sensitivity ,Lipolysis ,General Materials Science ,Obesity ,microphysiological system ,Nanoscience & Nanotechnology ,Metabolic and endocrine ,organ-on-a-chip ,Chemistry ,Stem Cells ,Diabetes ,General Chemistry ,Stem Cell Research ,Endocrinology ,Adipose Tissue ,Insulin Resistance ,Stem cell ,Ex vivo ,Biotechnology - Abstract
Impaired white adipose tissue (WAT) function has been recognized as a critical early event in obesity-driven disorders, but high buoyancy, fragility, and heterogeneity of primary adipocytes have largely prevented their use in drug discovery efforts highlighting the need for human stem cell-based approaches. Here, human stem cells are utilized to derive metabolically functional 3D adipose tissue (iADIPO) in a microphysiological system (MPS). Surprisingly, previously reported WAT differentiation approaches create insulin resistant WAT ill-suited for type-2 diabetes mellitus drug discovery. Using three independent insulin sensitivity assays, i.e., glucose and fatty acid uptake and suppression of lipolysis, as the functional readouts new differentiation conditions yielding hormonally responsive iADIPO are derived. Through concomitant optimization of an iADIPO-MPS, it is abled to obtain WAT with more unilocular and significantly larger (≈40%) lipid droplets compared to iADIPO in 2D culture, increased insulin responsiveness of glucose uptake (≈2-3 fold), fatty acid uptake (≈3-6 fold), and ≈40% suppressing of stimulated lipolysis giving a dynamic range that is competent to current in vivo and ex vivo models, allowing to identify both insulin sensitizers and desensitizers.
- Published
- 2021
16. 4β-Hydroxycholesterol is a prolipogenic factor that promotes SREBP1c expression and activity through the liver X receptor
- Author
-
Douglas F. Covey, Daniel K. Nomura, Ofer Moldavski, Xuntian Jiang, Mingxing Qian, Ethan J. Weiss, Robert J. van Eijkeren, Andreas Stahl, Peter-James H. Zushin, Charles A. Berdan, Daniel S. Ory, and Roberto Zoncu
- Subjects
0301 basic medicine ,insulin ,Biochemistry & Molecular Biology ,4β-HC, 4β-hydroxycholesterol ,LD, lipid droplet ,mTOR, mechanistic Target of Rapamycin ,Oxysterol ,lipid droplets ,LDS, lipid-depleted serum ,QD415-436 ,DNL, de novo lipogenesis ,Medical Biochemistry and Metabolomics ,030204 cardiovascular system & hematology ,HC, hydroxycholesterol ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,EtOAc, ethyl acetate ,ent-4HC, enantiomer of 4β-HC ,Lipid droplet ,Lipid biosynthesis ,Genetics ,polycyclic compounds ,Transcriptional regulation ,Liver X receptor ,Transcription factor ,Chemistry ,Liver Disease ,de-novo-lipogenesis ,liver-X-Receptor ,PI3K, phosphatidylinositol 3-kinase ,Lipid metabolism ,DPBS, dulbecco’s phosphate buffered saline ,Cell Biology ,SREBP1c ,Cell biology ,030104 developmental biology ,Lipogenesis ,lipids (amino acids, peptides, and proteins) ,NAFLD, nonalcoholic fatty liver disease ,Biochemistry and Cell Biology ,Digestive Diseases ,Sterol Regulatory Element Binding Protein 1 ,oxysterol ,THF, tetrahydrofuran ,Research Article - Abstract
Oxysterols are oxidized derivatives of cholesterol that play regulatory roles in lipid biosynthesis and homeostasis. How oxysterol signaling coordinates different lipid classes such as sterols and triglycerides remains incompletely understood. Here, we show that 4β-hydroxycholesterol (HC) (4β-HC), a liver and serum abundant oxysterol of poorly defined functions, is a potent and selective inducer of the master lipogenic transcription factor, SREBP1c, but not the related steroidogenic transcription factor SREBP2. By correlating tracing of lipid synthesis with lipogenic gene expression profiling, we found that 4β-HC acts as a putative agonist for the liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of the LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c and triggered de novo lipogenesis both in primary hepatocytes and in the mouse liver. In addition, 4β-HC acted in parallel to insulin-PI3K–dependent signaling to stimulate triglyceride synthesis and lipid-droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
- Published
- 2021
17. Cdkal1, a type 2 diabetes susceptibility gene, regulates mitochondrial function in adipose tissue
- Author
-
Zhaoming Deng, Katherine B. LeClair, Colin J. Palmer, Steven P. Gygi, Lawrence Kazak, Alexander S. Banks, Shangyu Hong, Kyle L. Smith, Susan J. Hagen, Raphael J. Bruckner, Peter-James H. Zushin, Joao A. Paulo, Amir I. Mina, Jessica A. Hall, David E. Cohen, and Jonathan Z. Long
- Subjects
0301 basic medicine ,Male ,HFD, high-fat diet ,FGF21 ,Adipose ,Adipose tissue ,Mice, Obese ,White adipose tissue ,Mice ,Adipose Tissue, Brown ,Insulin-Secreting Cells ,Brown adipose tissue ,Adipocytes ,Insulin ,GWAS ,CDK5RAP1, CDK5 regulatory subunit associated protein 1 ,Inner mitochondrial membrane ,Adiposity ,2. Zero hunger ,PRDM16 ,Mice, Knockout ,tRNA Methyltransferases ,ANT1 ,Diabetes ,A-KO, adipose-specific Cdkal1 KO ,Thermogenin ,Cell biology ,Mitochondria ,medicine.anatomical_structure ,Biochemistry ,Original Article ,Cdkal1 ,Lys, lysine ,lcsh:Internal medicine ,Adipose tissue macrophages ,Adipose Tissue, White ,Nerve Tissue Proteins ,Biology ,Mitochondrial Proteins ,03 medical and health sciences ,medicine ,Animals ,Genetic Predisposition to Disease ,Obesity ,lcsh:RC31-1245 ,Molecular Biology ,CDKAL1, CDK5 regulatory subunit associated protein 1 like 1 ,Cell Biology ,OCR, Oxygen consumption rate ,Mice, Inbred C57BL ,030104 developmental biology ,Glucose ,Diabetes Mellitus, Type 2 - Abstract
Objectives Understanding how loci identified by genome wide association studies (GWAS) contribute to pathogenesis requires new mechanistic insights. Variants within CDKAL1 are strongly linked to an increased risk of developing type 2 diabetes and obesity. Investigations in mouse models have focused on the function of Cdkal1 as a tRNALys modifier and downstream effects of Cdkal1 loss on pro-insulin translational fidelity in pancreatic β−cells. However, Cdkal1 is broadly expressed in other metabolically relevant tissues, including adipose tissue. In addition, the Cdkal1 homolog Cdk5rap1 regulates mitochondrial protein translation and mitochondrial function in skeletal muscle. We tested whether adipocyte-specific Cdkal1 deletion alters systemic glucose homeostasis or adipose mitochondrial function independently of its effects on pro-insulin translation and insulin secretion. Methods We measured mRNA levels of type 2 diabetes GWAS genes, including Cdkal1, in adipose tissue from lean and obese mice. We then established a mouse model with adipocyte-specific Cdkal1 deletion. We examined the effects of adipose Cdkal1 deletion using indirect calorimetry on mice during a cold temperature challenge, as well as by measuring cellular and mitochondrial respiration in vitro. We also examined brown adipose tissue (BAT) mitochondrial morphology by electron microscopy. Utilizing co-immunoprecipitation followed by mass spectrometry, we performed interaction mapping to identify new CDKAL1 binding partners. Furthermore, we tested whether Cdkal1 loss in adipose tissue affects total protein levels or accurate Lys incorporation by tRNALys using quantitative mass spectrometry. Results We found that Cdkal1 mRNA levels are reduced in adipose tissue of obese mice. Using adipose-specific Cdkal1 KO mice (A-KO), we demonstrated that mitochondrial function is impaired in primary differentiated brown adipocytes and in isolated mitochondria from A-KO brown adipose tissue. A-KO mice displayed decreased energy expenditure during 4 °C cold challenge. Furthermore, mitochondrial morphology was highly abnormal in A-KO BAT. Surprisingly, we found that lysine codon representation was unchanged in Cdkal1 A-KO adipose tissue. We identified novel protein interactors of CDKAL1, including SLC25A4/ANT1, an inner mitochondrial membrane ADP/ATP translocator. ANT proteins can account for the UCP1-independent basal proton leak in BAT mitochondria. Cdkal1 A-KO mice had increased ANT1 protein levels in their white adipose tissue. Conclusions Cdkal1 is necessary for normal mitochondrial morphology and function in adipose tissue. These results suggest that the type 2 diabetes susceptibility gene CDKAL1 has novel functions in regulating mitochondrial activity., Highlights • Cdkal1 is a gene most strongly expressed in tissues with high mitochondrial content. • Cdkal1 is required for normal mitochondrial morphology and function. • Deletion of Cdkal1 in adipose tissue impairs the thermogenic response to a cold challenge. • Cdkal1 interacts with ANT1, a mitochondrial ATP/ADP transporter. • Loss of Cdkal1 does not affect protein translation as predicted for a tRNA modifying enzyme.
- Published
- 2017
18. WAT-on-a-chip: A physiologically relevant microfluidic system incorporating white adipose tissue
- Author
-
Andreas Stahl, Kevin E. Healy, Thiagarajan Sezhian, Willie Mae Reese, Peter-James H. Zushin, Shaheen Jeeawoody, Peter Loskill, Felipe T. Lee-Montiel, and Kevin M. Tharp
- Subjects
0301 basic medicine ,Adipose Tissue, White ,Microfluidics ,Biomedical Engineering ,Adipose tissue ,Context (language use) ,White ,Bioengineering ,White adipose tissue ,Biology ,Biochemistry ,Models, Biological ,Article ,Analytical Chemistry ,03 medical and health sciences ,Mice ,Engineering ,In vivo ,Models ,Lab-On-A-Chip Devices ,Animals ,Humans ,Computer Simulation ,Obesity ,Metabolic and endocrine ,Nutrition ,Convective transport ,Lipid metabolism ,General Chemistry ,3T3 Cells ,Equipment Design ,Microfluidic Analytical Techniques ,Biological ,Cell biology ,Stroke ,030104 developmental biology ,Adipose Tissue ,Blood circulation ,Chemical Sciences ,Biomedical engineering ,Biotechnology - Abstract
Organ-on-a-chip systems possess a promising future as drug screening assays and as testbeds for disease modeling in the context of both single-organ systems and multi-organ-chips. Although it comprises approximately one fourth of the body weight of a healthy human, an organ frequently overlooked in this context is white adipose tissue (WAT). WAT-on-a-chip systems are required to create safety profiles of a large number of drugs due to their interactions with adipose tissue and other organs via paracrine signals, fatty acid release, and drug levels through sequestration. We report a WAT-on-a-chip system with a footprint of less than 1 mm2 consisting of a separate media channel and WAT chamber connected via small micropores. Analogous to the in vivo blood circulation, convective transport is thereby confined to the vasculature-like structures and the tissues protected from shear stresses. Numerical and analytical modeling revealed that the flow rates in the WAT chambers are less than 1/100 of the input flow rate. Using optimized injection parameters, we were able to inject pre-adipocytes, which subsequently formed adipose tissue featuring fully functional lipid metabolism. The physiologically relevant microfluidic environment of the WAT-chip supported long term culture of the functional adipose tissue for more than two weeks. Due to its physiological, highly controlled, and computationally predictable character, the system has the potential to be a powerful tool for the study of adipose tissue associated diseases such as obesity and type 2 diabetes.
- Published
- 2017
19. An ERK/Cdk5 axis controls the diabetogenic actions of PPARγ
- Author
-
Peter-James H. Zushin, Dina Laznik-Bogoslavski, Fiona E. McAllister, Gerald I. Shulman, Michael J. Jurczak, Joao Paulo Camporez, Alexander S. Banks, Bruce M. Spiegelman, and Steven P. Gygi
- Subjects
MAPK/ERK pathway ,Male ,MAP Kinase Signaling System ,MAP Kinase Kinase 2 ,Adipose tissue ,Peroxisome proliferator-activated receptor ,Mice, Obese ,Diet, High-Fat ,Article ,Mice ,Insulin resistance ,medicine ,Adipocytes ,Diabetes Mellitus ,Animals ,Humans ,Phosphorylation ,Extracellular Signal-Regulated MAP Kinases ,Cells, Cultured ,Cell Proliferation ,chemistry.chemical_classification ,Multidisciplinary ,biology ,Chemistry ,Kinase ,Cyclin-dependent kinase 5 ,Cyclin-Dependent Kinase 5 ,medicine.disease ,Mice, Inbred C57BL ,PPAR gamma ,Adipose Tissue ,Mitogen-activated protein kinase ,Cancer research ,biology.protein ,Insulin Resistance - Abstract
Blocking ERK/MAP kinases improves insulin sensitivity thorough a mechanism similar to the actions of the anti-diabetic thiazolidinediones drugs on PPARγ. The nuclear receptor PPARγ is required for glucose metabolism and is a drug target in diabetes. Earlier work has suggested that PPARγ phosphorylation has an adverse effect on insulin sensitivity and that the kinase Cdk5 is the main kinase responsible for phosphorylation at this site. Now Bruce Spiegelman and colleagues report the paradoxical observation that insulin resistance is worsened in mice lacking Cdk5 in adipose tissue. The authors further show that Cdk5 suppresses the MEK/ERK signalling pathway that is central to the regulation of cell growth and proliferation, and that ERK directly phosphorylates PPARγ. MEK/ERK inhibitors can improve insulin resistance in obese wild-type and ob/ob mice. This work suggests that MEK/ERK inhibitors — already approved for cancer treatment — might also be effective against diabetes. Obesity-linked insulin resistance is a major precursor to the development of type 2 diabetes. Previous work has shown that phosphorylation of PPARγ (peroxisome proliferator-activated receptor γ) at serine 273 by cyclin-dependent kinase 5 (Cdk5) stimulates diabetogenic gene expression in adipose tissues1. Inhibition of this modification is a key therapeutic mechanism for anti-diabetic drugs that bind PPARγ, such as the thiazolidinediones and PPARγ partial agonists or non-agonists2. For a better understanding of the importance of this obesity-linked PPARγ phosphorylation, we created mice that ablated Cdk5 specifically in adipose tissues. These mice have both a paradoxical increase in PPARγ phosphorylation at serine 273 and worsened insulin resistance. Unbiased proteomic studies show that extracellular signal-regulated kinase (ERK) kinases are activated in these knockout animals. Here we show that ERK directly phosphorylates serine 273 of PPARγ in a robust manner and that Cdk5 suppresses ERKs through direct action on a novel site in MAP kinase/ERK kinase (MEK). Importantly, pharmacological inhibition of MEK and ERK markedly improves insulin resistance in both obese wild-type and ob/ob mice, and also completely reverses the deleterious effects of the Cdk5 ablation. These data show that an ERK/Cdk5 axis controls PPARγ function and suggest that MEK/ERK inhibitors may hold promise for the treatment of type 2 diabetes.
- Published
- 2014
20. 21 Genetic Modulation of PPARγ Phosphorylation Decreases the Severity of Experimental Colitis in Mice
- Author
-
Gregory McElroy, Alexander S. Banks, Sarah N. Flier, and Peter-James H. Zushin
- Subjects
Hepatology ,Chemistry ,Gastroenterology ,Genetic modulation ,Cancer research ,Experimental colitis ,Phosphorylation - Published
- 2015
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.