201
|
Wang Q, Li H, Tajima K, Verkerke ARP, Taxin ZH, Hou Z, Cole JB, Li F, Wong J, Abe I, Pradhan RN, Yamamuro T, Yoneshiro T, Hirschhorn JN, Kajimura S. Post-translational control of beige fat biogenesis by PRDM16 stabilization. Nature 2022; 609:151-158. [PMID: 35978186 PMCID: PMC9433319 DOI: 10.1038/s41586-022-05067-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 07/04/2022] [Indexed: 02/08/2023]
Abstract
Compelling evidence shows that brown and beige adipose tissue are protective against metabolic diseases1,2. PR domain-containing 16 (PRDM16) is a dominant activator of the biogenesis of beige adipocytes by forming a complex with transcriptional and epigenetic factors and is therefore an attractive target for improving metabolic health3-8. However, a lack of knowledge surrounding the regulation of PRDM16 protein expression hampered us from selectively targeting this transcriptional pathway. Here we identify CUL2-APPBP2 as the ubiquitin E3 ligase that determines PRDM16 protein stability by catalysing its polyubiquitination. Inhibition of CUL2-APPBP2 sufficiently extended the half-life of PRDM16 protein and promoted beige adipocyte biogenesis. By contrast, elevated CUL2-APPBP2 expression was found in aged adipose tissues and repressed adipocyte thermogenesis by degrading PRDM16 protein. Importantly, extended PRDM16 protein stability by adipocyte-specific deletion of CUL2-APPBP2 counteracted diet-induced obesity, glucose intolerance, insulin resistance and dyslipidaemia in mice. These results offer a cell-autonomous route to selectively activate the PRDM16 pathway in adipose tissues.
Collapse
Affiliation(s)
- Qiang Wang
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Huixia Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Kazuki Tajima
- Department of Endocrinology and Metabolism, National Hospital Organization, Yokohama Medical Center, Yokohama, Japan
| | - Anthony R P Verkerke
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Zachary H Taxin
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Zhishuai Hou
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Joanne B Cole
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Fei Li
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jake Wong
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ichitaro Abe
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | - Tadashi Yamamuro
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Takeshi Yoneshiro
- Division of Metabolic Medicine, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Joel N Hirschhorn
- Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
202
|
Min HY, Hwang J, Choi Y, Jo YH. Overexpressing the hydroxycarboxylic acid receptor 1 in mouse brown adipose tissue restores glucose tolerance and insulin sensitivity in diet-induced obese mice. Am J Physiol Endocrinol Metab 2022; 323:E231-E241. [PMID: 35830691 PMCID: PMC9423771 DOI: 10.1152/ajpendo.00084.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/05/2022] [Accepted: 07/09/2022] [Indexed: 11/22/2022]
Abstract
Interscapular brown adipose tissue (BAT) plays an important role in controlling glucose homeostasis. Increased glucose entry and glycolysis in BAT result in lactate production and release. The adipose tissue expresses the lactate receptor hydrocarboxylic acid receptor 1 (HCAR1), markedly downregulated in male diet-induced obese (DIO) and ob/ob mice. In this study, we examined the role of HCAR1 in BAT in controlling glucose homeostasis in male DIO mice. We overexpressed HCAR1 in BAT by injecting adeno-associated viruses (AAVs) expressing HCAR1 into the BAT pads of male DIO C57BL/6J mice. Overexpressing HCAR1 in BAT resulted in augmented glucose uptake by BAT in response to treatment with the HCAR1 agonist. HCAR1 overexpression elevated BAT temperature associated with increased thermogenic gene expression in BAT. HCAR1 overexpression prevented body weight gain in male DIO mice. Importantly, mice overexpressing HCAR1 in BAT exhibited improved glucose tolerance and insulin sensitivity. HCAR1 overexpression upregulated the Slc2a4 gene expression and promoted GLUT4 trafficking to the plasma membrane. In addition, mice overexpressing HCAR1 displayed a decrease in hormone-sensitive lipase (HSL) phosphorylation and increased lipogenic enzyme gene expression in BAT. Unlike DIO mice, overexpressing HCAR1 in BAT of mice fed a low-fat diet did not change body weight gain and glucose homeostasis. Taken together, our results support the interpretation that HCAR1 expressed in BAT promotes glucose entry and reduces lipolysis in BAT of male DIO mice. As activation of HCAR1 in BAT restores body weight, glucose tolerance, and insulin sensitivity in male DIO mice, our study suggests that interoceptive lactate detection via HCAR1 in BAT can regulate glucose and lipid substrate utilization and/or availability to promote healthy metabolism.NEW & NOTEWORTHY HCAR1 expressed in BAT can promote glucose entry and reduce lipolysis, resulting in body weight loss and increased insulin sensitivity. Hence, targeting HCAR1 in BAT would provide an alternative way to control body weight and euglycemia in individuals with obesity.
Collapse
Affiliation(s)
- Hyeon-Young Min
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York City, New York
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, New York City, New York
| | - Jiyeon Hwang
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York City, New York
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, New York City, New York
| | - Yuna Choi
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York City, New York
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, New York City, New York
| | - Young-Hwan Jo
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York City, New York
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine, New York City, New York
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York City, New York
| |
Collapse
|
203
|
Re GF, Li H, Yang JQ, Li Y, Zhang Z, Wu X, Zhou R, Kong D, Luo H, Kuang YQ, Wang KH. Exercise modulates central and peripheral inflammatory responses and ameliorates methamphetamine-induced anxiety-like symptoms in mice. Front Mol Neurosci 2022; 15:955799. [PMID: 36106141 PMCID: PMC9465459 DOI: 10.3389/fnmol.2022.955799] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 07/28/2022] [Indexed: 11/20/2022] Open
Abstract
Anxiety-like symptoms are common symptoms of methamphetamine (METH) users, especially in the acute withdrawal period, which is an important factor for the high relapse rate during METH acute withdrawal. Exercise has been demonstrated to relieve anxiety-like symptoms during METH withdrawal, but the underlying mechanisms of this anti-anxiety effect are still unclear. Activated microglia and abnormal neuroinflammation play an important role in the pathogenesis of anxiety-like symptoms after METH withdrawal. Moreover, peripheral immune factors were also significantly associated with anxiety symptoms. However, the effects of treadmill exercise on microglial function and neuroinflammation in the striatum and hippocampus during acute METH withdrawal have not been reported. In the current study, we found severe peripheral immune dysfunction in METH users during acute withdrawal, which may in part contribute to anxiety symptoms during METH acute withdrawal. We also showed that 2 weeks of METH exposure induced anxiety-like symptoms in the acute withdrawal period. Additionally, METH exposure resulted in increased microglial activation and proinflammatory cytokines released in the mouse striatum and hippocampus during acute withdrawal. We next evaluated the effects of treadmill exercise in countering anxiety-like symptoms induced by METH acute withdrawal. The results showed that anxiety-like symptoms induced by acute METH withdrawal were attenuated by coadministration of treadmill exercise. In addition, treadmill exercise counteracted METH-induced microglial activation in the mouse striatum and various subregions of the hippocampus. Furthermore, treadmill exercise also reversed the increase in proinflammatory cytokines induced by acute METH withdrawal in the mouse striatum, hippocampus and serum. Our findings suggest that the anti-anxiety effect of treadmill exercise may be mediated by reducing microglial activation and regulating central and peripheral inflammatory responses.
Collapse
Affiliation(s)
- Guo-Fen Re
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hong Li
- Yunnan Narcotics Control Bureau, Kunming, China
| | - Ji-Qun Yang
- The Third People’s Hospital of Kunming, Kunming, China
| | - Yue Li
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zunyue Zhang
- School of Medicine, Yunnan University, Kunming, China
| | - Xiaocong Wu
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruiyi Zhou
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Deshenyue Kong
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Huayou Luo
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- Department of Gastrointestinal and Hernia Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yi-Qun Kuang
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, China
- *Correspondence: Yi-Qun Kuang,
| | - Kun-Hua Wang
- School of Medicine, Yunnan University, Kunming, China
- National Health Commission (NHC) Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
- *Correspondence: Yi-Qun Kuang,
| |
Collapse
|
204
|
Zhou B, Claflin KE, Flippo KH, Sullivan AI, Asghari A, Tadinada SM, Jensen-Cody SO, Abel T, Potthoff MJ. Central FGF21 production regulates memory but not peripheral metabolism. Cell Rep 2022; 40:111239. [PMID: 36001982 PMCID: PMC9472585 DOI: 10.1016/j.celrep.2022.111239] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/25/2022] [Accepted: 07/28/2022] [Indexed: 11/25/2022] Open
Abstract
Fibroblast growth factor 21 (FGF21) is a liver-derived endocrine hormone that functions to regulate energy homeostasis and macronutrient intake. Recently, FGF21 was reported to be produced and secreted from hypothalamic tanycytes, to regulate peripheral lipid metabolism; however, rigorous investigation of FGF21 expression in the brain has yet to be accomplished. Using a mouse model that drives CRE recombinase in FGF21-expressing cells, we demonstrate that FGF21 is not expressed in the hypothalamus, but instead is produced from the retrosplenial cortex (RSC), an essential brain region for spatial learning and memory. Furthermore, we find that central FGF21 produced in the RSC enhances spatial memory but does not regulate energy homeostasis or sugar intake. Finally, our data demonstrate that administration of FGF21 prolongs the duration of long-term potentiation in the hippocampus and enhances activation of hippocampal neurons. Thus, endogenous and pharmacological FGF21 appear to function in the hippocampus to enhance spatial memory. Zhou et al. reveal that the endocrine hormone FGF21 is expressed in the brain. Central FGF21 expression occurs in distinct areas, including the retrosplenial cortex, but not the hypothalamus. Interestingly, brain-derived FGF21 regulates spatial memory formation, but not metabolism, and the converse is true for liver-derived FGF21.
Collapse
Affiliation(s)
- Bolu Zhou
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Kristin E Claflin
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Kyle H Flippo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Andrew I Sullivan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Arvand Asghari
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Satya M Tadinada
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Sharon O Jensen-Cody
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, 169 Newton Road, 3322 PBDB, Iowa City, IA 52242, USA; Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA.
| |
Collapse
|
205
|
Gupta A, Balakrishnan B, Karki S, Slayton M, Jash S, Banerjee S, Grahn THM, Jambunathan S, Disney S, Hussein H, Kong D, Lowell BB, Natarajan P, Reddy UK, Gokce N, Sharma VM, Puri V. Human CIDEC transgene improves lipid metabolism and protects against high-fat diet-induced glucose intolerance in mice. J Biol Chem 2022; 298:102347. [PMID: 35963433 PMCID: PMC9472082 DOI: 10.1016/j.jbc.2022.102347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/08/2022] [Accepted: 07/20/2022] [Indexed: 11/12/2022] Open
Abstract
Cell death–inducing DNA fragmentation factor-like effector C (CIDEC) expression in adipose tissue positively correlates with insulin sensitivity in obese humans. Further, E186X, a single-nucleotide CIDEC variant is associated with lipodystrophy, hypertriglyceridemia, and insulin resistance. To establish the unknown mechanistic link between CIDEC and maintenance of systemic glucose homeostasis, we generated transgenic mouse models expressing CIDEC (Ad-CIDECtg) and CIDEC E186X variant (Ad-CIDECmut) transgene specifically in the adipose tissue. We found that Ad-CIDECtg but not Ad-CIDECmut mice were protected against high-fat diet-induced glucose intolerance. Furthermore, we revealed the role of CIDEC in lipid metabolism using transcriptomics and lipidomics. Serum triglycerides, cholesterol, and low-density lipoproteins were lower in high-fat diet-fed Ad-CIDECtg mice compared to their littermate controls. Mechanistically, we demonstrated that CIDEC regulates the enzymatic activity of adipose triglyceride lipase via interacting with its activator, CGI-58, to reduce free fatty acid release and lipotoxicity. In addition, we confirmed that CIDEC is indeed a vital regulator of lipolysis in adipose tissue of obese humans, and treatment with recombinant CIDEC decreased triglyceride breakdown in visceral human adipose tissue. Our study unravels a central pathway whereby adipocyte-specific CIDEC plays a pivotal role in regulating adipose lipid metabolism and whole-body glucose homeostasis. In summary, our findings identify human CIDEC as a potential ‘drug’ or a ‘druggable’ target to reverse obesity-induced lipotoxicity and glucose intolerance.
Collapse
Affiliation(s)
- Abhishek Gupta
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Bijinu Balakrishnan
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Shakun Karki
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Mark Slayton
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Sukanta Jash
- Alpert Medical school of Brown University, Brown University, RI, USA
| | - Sayani Banerjee
- Alpert Medical school of Brown University, Brown University, RI, USA
| | - Tan Hooi Min Grahn
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University Hospital, Lund, Sweden
| | | | - Sarah Disney
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Hebaallaha Hussein
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Dong Kong
- Division of Endocrinology, Department of Pediatrics, F.M. Kirby Neurobiology Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA; Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | | | - Umesh K Reddy
- Department of Biology, West Virginia State University, Institute, WV, USA
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Vishva M Sharma
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| | - Vishwajeet Puri
- Department of Biomedical Sciences and Diabetes Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA.
| |
Collapse
|
206
|
Stork BA, Dean A, Ortiz AR, Saha P, Putluri N, Planas-Silva MD, Mahmud I, Rajapakshe K, Coarfa C, Knapp S, Lorenzi PL, Kemp BE, Turk BE, Scott JW, Means AR, York B. Calcium/calmodulin-dependent protein kinase kinase 2 regulates hepatic fuel metabolism. Mol Metab 2022; 62:101513. [PMID: 35562082 PMCID: PMC9157561 DOI: 10.1016/j.molmet.2022.101513] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/04/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE The liver is the primary internal metabolic organ that coordinates whole body energy homeostasis in response to feeding and fasting. Genetic ablation or pharmacological inhibition of calcium/calmodulin-dependent protein kinase kinase 2 (CaMKK2) has been shown to significantly improve hepatic health and peripheral insulin sensitivity upon overnutrition with high fat diet. However, the precise molecular underpinnings that explain this metabolic protection have remained largely undefined. METHODS To characterize the role of CaMKK2 in hepatic metabolism, we developed and challenged liver-specific CaMKK2 knockout (CaMKK2LKO) mice with high fat diet and performed glucose and insulin tolerance tests to evaluate peripheral insulin sensitivity. We used a combination of RNA-Sequencing, glucose and fatty acid istotopic tracer studies, a newly developed Seahorse assay for measuring the oxidative capacity of purified peroxisomes, and a degenerate peptide libarary to identify putative CaMKK2 substrates that mechanistically explain the protective effects of hepatic CaMKK2 ablation. RESULTS Consistent with previous findings, we show that hepatic CaMKK2 ablation significantly improves indices of peripheral insulin sensitivity. Mechanistically, we found that CaMKK2 phosphorylates and regulates GAPDH to promote glucose metabolism and PEX3 to blunt peroxisomal fatty acid catabolism in the liver. CONCLUSION CaMKK2 is a central metabolic fuel sensor in the liver that significantly contributes to whole body systems metabolism.
Collapse
Affiliation(s)
- Brittany A Stork
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Adam Dean
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Andrea R Ortiz
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Pradip Saha
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Nagireddy Putluri
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Iqbal Mahmud
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Kimal Rajapakshe
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Cristian Coarfa
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Stefan Knapp
- Institut für Pharmazeutische Chemie, Goethe University Frankfurt am Main, Max-von-Laue-Str. 9, 60438 Frankfurt am Main, Germany; Structural Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438, Frankfurt am Main, Germany
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, Victoria, 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, 3000, Australia
| | - Benjamin E Turk
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - John W Scott
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Fitzroy, Victoria, 3065, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3052, Australia
| | - Anthony R Means
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Brian York
- Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
207
|
Zhang W, Jia X, Xu Y, Xie Q, Zhu M, Zhang H, Zhao Z, Hao J, Li H, Du J, Liu Y, Liu WH, Ma X, Hung W, Feng H, Li H. Effects of Coix Seed Extract, Bifidobacterium BPL1, and Their Combination on the Glycolipid Metabolism in Obese Mice. Front Nutr 2022; 9:939423. [PMID: 35923203 PMCID: PMC9341295 DOI: 10.3389/fnut.2022.939423] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Coix seed extract (CSE) and probiotics have been reported to regulate glycolipid metabolism via different modes of action. We tested the effects of CSE, Bifidobacterium BPL1, and their combination to determine their effects on glycolipid metabolism in obese mice. Male C57BL/6J mice were fed a high-fat diet for 8 weeks to establish an obesity model. Obese mice were selected and divided into four groups: the model control group and three intervention groups. After 10 weeks of continuous gavage intervention, the mice in the intervention groups exhibited lower body weight (lower about 2.31 g, vs. HFD mice 42.23 g) and epididymal (lower about 0.37 g, vs. HFD mice 2.5 g) and perirenal fat content (lower about 0.47 g, vs. HFD mice 0.884 g); decreased fasting blood glucose, total cholesterol, triglycerides, and VLDL; and increased HLDL, respiratory exchange ratio, energy expenditure, and amount of exercise performed. CSE, BPL1 and their combination can effectively control the weight gain in obese mice, reduce fat content, and regulate blood lipids and abnormal blood sugar. These results may be related to reduce the chronic inflammatory states, improve energy metabolism, exercise, relieve insulin sensitivity, and reduce lipid synthesis via the intervention of CSE, BPL1 and their combination. Compared with the single use of CSE alone, the combination of CSE + BPL1 can better exert the regulation function of intestinal flora, and change in the abundance of bacteria that could improve the level of inflammatory factors, such as increasing Bifidobacterium, reducing Lactococcus. Compared with the use of BPL1 alone, the combination of CSE and BPL1 can better regulate pancreatic islet and improve blood sugar. CSE may act directly on body tissues to exert anti-inflammatory effects. BPL1 and CSE + BPL1 may improve the structure and function of the intestinal flora, and reduce tissue inflammation.
Collapse
Affiliation(s)
- Wei Zhang
- School of Public Health, Xiamen University, Xiamen, China
| | - Xiuzhen Jia
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Yuhan Xu
- School of Public Health, Xiamen University, Xiamen, China
| | - Qiaoling Xie
- School of Public Health, Xiamen University, Xiamen, China
| | - Meizhen Zhu
- School of Public Health, Xiamen University, Xiamen, China
| | - Hesong Zhang
- School of Public Health, Xiamen University, Xiamen, China
| | - Zifu Zhao
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Jingyu Hao
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Haoqiu Li
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Jinrui Du
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Yan Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Wei-Hsien Liu
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Xia Ma
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Weilian Hung
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Haotian Feng
- Inner Mongolia Dairy Technology Research Institute Co., Ltd., Hohhot, China
- Yili Innovation Center, Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Hongwei Li
- School of Public Health, Xiamen University, Xiamen, China
| |
Collapse
|
208
|
Greenwell AA, Saed CT, Tabatabaei Dakhili SA, Ho KL, Gopal K, Chan JSF, Kaczmar OO, Dyer SA, Eaton F, Lopaschuk GD, Al Batran R, Ussher JR. An isoproteic cocoa butter-based ketogenic diet fails to improve glucose homeostasis and promote weight loss in obese mice. Am J Physiol Endocrinol Metab 2022; 323:E8-E20. [PMID: 35575232 DOI: 10.1152/ajpendo.00435.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
High-fat and very low-carbohydrate based ketogenic diets have gained considerable popularity as a nonpharmacological strategy for obesity, due to their potential to enhance weight loss and improve glucose homeostasis. However, the effectiveness of a ketogenic diet toward metabolic health is equivocal. To better understand the impact of ketogenic diets in obesity, male and female mice were fed a 60% cocoa butter-based high-fat diet for 16-wk to induce obesity, following which mice were transitioned to either an 85% cocoa butter fat-based ketogenic diet, a 10% cocoa butter fat-based low-fat diet, or maintained on a high-fat diet for an additional 8-wk. All experimental diets were matched for sucrose and protein content and contained an identical micronutrient profile, with complex carbohydrates being the primary carbohydrate source in the low-fat diet. The transition to a ketogenic diet was ineffective at promoting significant body fat loss and improving glucose homeostasis in obese male and female mice. Alternatively, obese male and female mice transitioned to a low-fat and high-complex carbohydrate diet exhibited beneficial body composition changes and improved glucose tolerance that may, in part, be attributed to a mild decrease in food intake and a mild increase in energy expenditure. Our findings support the consumption of a diet low in saturated fat and rich in complex carbohydrates as a potential dietary intervention for the treatment of obesity and obesity-induced impairments in glycemia. Furthermore, our results suggest that careful consideration should be taken when considering a ketogenic diet as a nonpharmacological strategy for obesity.NEW & NOTEWORTHY It has been demonstrated that ketogenic diets may be a nutritional strategy for alleviating hyperglycemia and promoting weight loss in obesity. However, there are a number of inconsistencies with many of these studies, especially with regard to the macronutrient and micronutrient compositions of the diets being compared. Our work demonstrates that a ketogenic diet that is both micronutrient-matched and isoproteic with its comparator diets fails to improve glycemia or promote weight loss in obese mice.
Collapse
Affiliation(s)
- Amanda A Greenwell
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Christina T Saed
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Seyed Amirhossein Tabatabaei Dakhili
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Kim L Ho
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Keshav Gopal
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Jordan S F Chan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Oksana O Kaczmar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Scott A Dyer
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Farah Eaton
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D Lopaschuk
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Rami Al Batran
- Faculty of Pharmacy, Université de Montréal, Montreal, Quebec, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
209
|
Deletion of LDLRAP1 Induces Atherosclerotic Plaque Formation, Insulin Resistance, and Dysregulated Insulin Response in Adipose Tissue. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1092-1108. [PMID: 35460615 PMCID: PMC9253916 DOI: 10.1016/j.ajpath.2022.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/18/2022] [Accepted: 03/28/2022] [Indexed: 12/25/2022]
Abstract
Dyslipidemia, vascular inflammation, obesity, and insulin resistance often overlap and exacerbate each other. Mutations in low density lipoprotein receptor adaptor protein-1 (LDLRAP1) lead to LDLR malfunction and are associated with the autosomal recessive hypercholesterolemia disorder in humans. However, direct causality on atherogenesis in a defined preclinical model has not been reported. The objective of this study was to test the hypothesis that deletion of LDLRAP1 will lead to hypercholesteremia and atherosclerosis. LDLRAP1-/- mice fed a high-fat Western diet had significantly increased plasma cholesterol and triglyceride concentrations accompanied with significantly increased plaque burden compared with wild-type controls. Unexpectedly, LDLRAP1-/- mice gained significantly more weight compared with controls. Even on a chow diet, LDLRAP1-/- mice were insulin-resistant, and calorimetric studies suggested an altered metabolic profile. The study showed that LDLRAP1 is highly expressed in visceral adipose tissue, and LDLRAP1-/- adipocytes are significantly larger, have reduced glucose uptake and AKT phosphorylation, but have increased CD36 expression. Visceral adipose tissue from LDLRAP1-/- mice was hypoxic and had gene expression signatures of dysregulated lipid storage and energy homeostasis. These data are the first to indicate that lack of LDLRAP1 directly leads to atherosclerosis in mice and also plays an unanticipated metabolic regulatory role in adipose tissue. LDLRAP1 may link atherosclerosis and hypercholesterolemia with common comorbidities of obesity and insulin resistance.
Collapse
|
210
|
Bernier M, Enamorado IN, Gómez-Cabrera MC, Calvo-Rubio M, González-Reyes JA, Price NL, Cortés-Rodríguez AB, Rodríguez-Aguilera JC, Rodríguez-López S, Mitchell SJ, Murt KN, Kalafut K, Williams KM, Ward CW, Stains JP, Brea-Calvo G, Villalba JM, Cortassa S, Aon MA, de Cabo R. Age-dependent impact of two exercise training regimens on genomic and metabolic remodeling in skeletal muscle and liver of male mice. NPJ AGING 2022; 8:8. [PMID: 35927269 PMCID: PMC9237062 DOI: 10.1038/s41514-022-00089-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 05/11/2022] [Indexed: 11/09/2022]
Abstract
Skeletal muscle adapts to different exercise training modalities with age; however, the impact of both variables at the systemic and tissue levels is not fully understood. Here, adult and old C57BL/6 male mice were assigned to one of three groups: sedentary, daily high-intensity intermittent training (HIIT), or moderate intensity continuous training (MICT) for 4 weeks, compatible with the older group's exercise capacity. Improvements in body composition, fasting blood glucose, and muscle strength were mostly observed in the MICT old group, while effects of HIIT training in adult and old animals was less clear. Skeletal muscle exhibited structural and functional adaptations to exercise training, as revealed by electron microscopy, OXPHOS assays, respirometry, and muscle protein biomarkers. Transcriptomics analysis of gastrocnemius muscle combined with liver and serum metabolomics unveiled an age-dependent metabolic remodeling in response to exercise training. These results support a tailored exercise prescription approach aimed at improving health and ameliorating age-associated loss of muscle strength and function in the elderly.
Collapse
Affiliation(s)
- Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Ignacio Navas Enamorado
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
- Translational Medicine Section, Akouos, Inc., 645 Summer St, Boston, MA, 02210, USA
| | - Mari Carmen Gómez-Cabrera
- Freshage Research Group, Department of Physiology, Faculty of Medicine, University of Valencia, and CIBERFES, Fundación Investigación Hospital Clínico Universitario/INCLIVA, Valencia, Spain
| | - Miguel Calvo-Rubio
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
- Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, 3ª planta, 14014, Córdoba, Spain
| | - Jose Antonio González-Reyes
- Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, 3ª planta, 14014, Córdoba, Spain
| | - Nathan L Price
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | | | | | - Sandra Rodríguez-López
- Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, 3ª planta, 14014, Córdoba, Spain
| | - Sarah J Mitchell
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Kelsey N Murt
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Krystle Kalafut
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Katrina M Williams
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Christopher W Ward
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Gloria Brea-Calvo
- Centro Andaluz de Biología del Desarrollo and CIBERER, Instituto de Salud Carlos III, Universidad Pablo de Olavide - CSIC - JA, Sevilla, 41013, Spain
| | - Jose M Villalba
- Departamento de Biología Celular, Fisiología e Inmunología, Campus de Excelencia Internacional Agroalimentario, ceiA3, Universidad de Córdoba, Campus de Rabanales, Edificio Severo Ochoa, 3ª planta, 14014, Córdoba, Spain
| | - Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Miguel A Aon
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, 21224, USA.
| |
Collapse
|
211
|
Metabolic Impact of MKP-2 Upregulation in Obesity Promotes Insulin Resistance and Fatty Liver Disease. Nutrients 2022; 14:nu14122475. [PMID: 35745205 PMCID: PMC9228271 DOI: 10.3390/nu14122475] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
The mechanisms connecting obesity with type 2 diabetes, insulin resistance, nonalcoholic fatty liver disease, and cardiovascular diseases remain incompletely understood. The function of MAPK phosphatase-2 (MKP-2), a type 1 dual-specific phosphatase (DUSP) in whole-body metabolism, and how this contributes to the development of diet-induced obesity, type 2 diabetes (T2D), and insulin resistance is largely unknown. We investigated the physiological contribution of MKP-2 in whole-body metabolism and whether MKP-2 is altered in obesity and human fatty liver disease using MKP-2 knockout mice models and human liver tissue derived from fatty liver disease patients. We demonstrate that, for the first time, MKP-2 expression was upregulated in liver tissue in humans with obesity and fatty liver disease and in insulin-responsive tissues in mice with obesity. MKP-2-deficient mice have enhanced p38 MAPK, JNK, and ERK activities in insulin-responsive tissues compared with wild-type mice. MKP-2 deficiency in mice protects against diet-induced obesity and hepatic steatosis and was accompanied by improved glucose homeostasis and insulin sensitivity. Mkp-2−/− mice are resistant to diet-induced obesity owing to reduced food intake and associated lower respiratory exchange ratio. This was associated with enhanced circulating insulin-like growth factor-1 (IGF-1) and stromal cell-derived factor 1 (SDF-1) levels in Mkp-2−/− mice. PTEN, a negative regulator of Akt, was downregulated in livers of Mkp-2−/− mice, resulting in enhanced Akt activity consistent with increased insulin sensitivity. These studies identify a novel role for MKP-2 in the regulation of systemic metabolism and pathophysiology of obesity-induced insulin resistance and fatty liver disease.
Collapse
|
212
|
Shammas MK, Huang X, Wu BP, Fessler E, Song I, Randolph NP, Li Y, Bleck CK, Springer DA, Fratter C, Barbosa IA, Powers AF, Quirós PM, Lopez-Otin C, Jae LT, Poulton J, Narendra DP. OMA1 mediates local and global stress responses against protein misfolding in CHCHD10 mitochondrial myopathy. J Clin Invest 2022; 132:157504. [PMID: 35700042 PMCID: PMC9282932 DOI: 10.1172/jci157504] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
Mitochondrial stress triggers a response in the cell’s mitochondria and nucleus, but how these stress responses are coordinated in vivo is poorly understood. Here, we characterize a family with myopathy caused by a dominant p.G58R mutation in the mitochondrial protein CHCHD10. To understand the disease etiology, we developed a knockin (KI) mouse model and found that mutant CHCHD10 aggregated in affected tissues, applying a toxic protein stress to the inner mitochondrial membrane. Unexpectedly, the survival of CHCHD10-KI mice depended on a protective stress response mediated by the mitochondrial metalloendopeptidase OMA1. The OMA1 stress response acted both locally within mitochondria, causing mitochondrial fragmentation, and signaled outside the mitochondria, activating the integrated stress response through cleavage of DAP3-binding cell death enhancer 1 (DELE1). We additionally identified an isoform switch in the terminal complex of the electron transport chain as a component of this response. Our results demonstrate that OMA1 was critical for neonatal survival conditionally in the setting of inner mitochondrial membrane stress, coordinating local and global stress responses to reshape the mitochondrial network and proteome.
Collapse
Affiliation(s)
- Mario K Shammas
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, United States of America
| | - Xiaoping Huang
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, United States of America
| | - Beverly P Wu
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, United States of America
| | - Evelyn Fessler
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Insung Song
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, United States of America
| | - Nicholas P Randolph
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, United States of America
| | - Yan Li
- Proteomics Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, United States of America
| | - Christopher Ke Bleck
- Electron Microscopy Core Facility, National Heart, Lung, and Blood Institute, Bethesda, United States of America
| | - Danielle A Springer
- Mouse Phenotyping Core, National Heart, Lung, and Blood Institute, Bethesda, United States of America
| | - Carl Fratter
- Oxford Medical Genetics Laboratories, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Ines A Barbosa
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | | | - Pedro M Quirós
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain
| | - Carlos Lopez-Otin
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, Oviedo, Spain
| | - Lucas T Jae
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Joanna Poulton
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Derek P Narendra
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, Bethesda, United States of America
| |
Collapse
|
213
|
Targeting parvalbumin promotes M2 macrophage polarization and energy expenditure in mice. Nat Commun 2022; 13:3301. [PMID: 35676256 PMCID: PMC9177846 DOI: 10.1038/s41467-022-30757-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/17/2022] [Indexed: 11/08/2022] Open
Abstract
Exercise benefits M2 macrophage polarization, energy homeostasis and protects against obesity partially through exercise-induced circulating factors. Here, by unbiased quantitative proteomics on serum samples from sedentary and exercised mice, we identify parvalbumin as a circulating factor suppressed by exercise. Parvalbumin functions as a non-competitive CSF1R antagonist to inhibit M2 macrophage activation and energy expenditure in adipose tissue. More importantly, serum concentrations of parvalbumin positively correlate with obesity in mouse and human, while treating mice with a recombinant parvalbumin blocker prevents its interaction with CSF1R and promotes M2 macrophage polarization and ameliorates diet-induced obesity. Thus, although further studies are required to assess the significance of parvalbumin in mediating the effects of exercise, our results implicate parvalbumin as a potential therapeutic strategy against obesity in mice.
Collapse
|
214
|
Verzijl CRC, Oldoni F, Loaiza N, Wolters JC, Rimbert A, Tian E, Yang W, Struik D, Smit M, Kloosterhuis NJ, Fernandez AJ, Samara NL, Ten Hagen KG, Dalal K, Chernish A, McCluggage P, Tabak LA, Jonker JW, Kuivenhoven JA. A novel role for GalNAc-T2 dependent glycosylation in energy homeostasis. Mol Metab 2022; 60:101472. [PMID: 35304331 PMCID: PMC9019398 DOI: 10.1016/j.molmet.2022.101472] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 02/25/2022] [Accepted: 03/07/2022] [Indexed: 10/27/2022] Open
Abstract
OBJECTIVE GALNT2, encoding polypeptide N-acetylgalactosaminyltransferase 2 (GalNAc-T2), was initially discovered as a regulator of high-density lipoprotein metabolism. GalNAc-T2 is known to exert these effects through post-translational modification, i.e., O-linked glycosylation of secreted proteins with established roles in plasma lipid metabolism. It has recently become clear that loss of GALNT2 in rodents, cattle, nonhuman primates, and humans should be regarded as a novel congenital disorder of glycosylation that affects development and body weight. The role of GALNT2 in metabolic abnormalities other than plasma lipids, including insulin sensitivity and energy homeostasis, is poorly understood. METHODS GWAS data from the UK Biobank was used to study variation in the GALNT2 locus beyond changes in high-density lipoprotein metabolism. Experimental data were obtained through studies in Galnt2-/- mice and wild-type littermates on both control and high-fat diet. RESULTS First, we uncovered associations between GALNT2 gene variation, adiposity, and body mass index in humans. In mice, we identify the insulin receptor as a novel substrate of GalNAc-T2 and demonstrate that Galnt2-/- mice exhibit decreased adiposity, alterations in insulin signaling and a shift in energy substrate utilization in the inactive phase. CONCLUSIONS This study identifies a novel role for GALNT2 in energy homeostasis, and our findings suggest that the local effects of GalNAc-T2 are mediated through posttranslational modification of the insulin receptor.
Collapse
Affiliation(s)
- Cristy R C Verzijl
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Federico Oldoni
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Natalia Loaiza
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Justina C Wolters
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Antoine Rimbert
- Nantes Université, CNRS, INSERM, l'institut du thorax, F-44000, Nantes, France
| | - E Tian
- Developmental Glycobiology Section, NIDCR, National Institutes of Health, Bethesda, MD, United States
| | - Weiming Yang
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Dicky Struik
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Marieke Smit
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Niels J Kloosterhuis
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Amy J Fernandez
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nadine L Samara
- Structural Biochemistry Unit, National Institutes of Health, Bethesda, MD, United States; Developmental Glycobiology Section, NIDCR, National Institutes of Health, Bethesda, MD, United States
| | - Kelly G Ten Hagen
- Developmental Glycobiology Section, NIDCR, National Institutes of Health, Bethesda, MD, United States
| | - Kruti Dalal
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Aliona Chernish
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Peggy McCluggage
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Lawrence A Tabak
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Johan W Jonker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
215
|
Dantas WS, Zunica ERM, Heintz EC, Vandanmagsar B, Floyd ZE, Yu Y, Fujioka H, Hoppel CL, Belmont KP, Axelrod CL, Kirwan JP. Mitochondrial uncoupling attenuates sarcopenic obesity by enhancing skeletal muscle mitophagy and quality control. J Cachexia Sarcopenia Muscle 2022; 13:1821-1836. [PMID: 35304976 PMCID: PMC9178352 DOI: 10.1002/jcsm.12982] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/28/2022] [Accepted: 02/21/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Sarcopenic obesity is a highly prevalent disease with poor survival and ineffective medical interventions. Mitochondrial dysfunction is purported to be central in the pathogenesis of sarcopenic obesity by impairing both organelle biogenesis and quality control. We have previously identified that a mitochondrial-targeted furazano[3,4-b]pyrazine named BAM15 is orally available and selectively lowers respiratory coupling efficiency and protects against diet-induced obesity in mice. Here, we tested the hypothesis that mitochondrial uncoupling simultaneously attenuates loss of muscle function and weight gain in a mouse model of sarcopenic obesity. METHODS Eighty-week-old male C57BL/6J mice with obesity were randomized to 10 weeks of high fat diet (CTRL) or BAM15 (BAM15; 0.1% w/w in high fat diet) treatment. Body weight and food intake were measured weekly. Body composition, muscle function, energy expenditure, locomotor activity, and glucose tolerance were determined after treatment. Skeletal muscle was harvested and evaluated for histology, gene expression, protein signalling, and mitochondrial structure and function. RESULTS BAM15 decreased body weight (54.0 ± 2.0 vs. 42.3 ± 1.3 g, P < 0.001) which was attributable to increased energy expenditure (10.1 ± 0.1 vs. 11.3 ± 0.4 kcal/day, P < 0.001). BAM15 increased muscle mass (52.7 ± 0.4 vs. 59.4 ± 1.0%, P < 0.001), strength (91.1 ± 1.3 vs. 124.9 ± 1.2 g, P < 0.0001), and locomotor activity (347.0 ± 14.4 vs. 432.7 ± 32.0 m, P < 0.001). Improvements in physical function were mediated in part by reductions in skeletal muscle inflammation (interleukin 6 and gp130, both P < 0.05), enhanced mitochondrial function, and improved endoplasmic reticulum homeostasis. Specifically, BAM15 activated mitochondrial quality control (PINK1-ubiquitin binding and LC3II, P < 0.01), increased mitochondrial activity (citrate synthase and complex II activity, all P < 0.05), restricted endoplasmic reticulum (ER) misfolding (decreased oligomer A11 insoluble/soluble ratio, P < 0.0001) while limiting ER stress (decreased PERK signalling, P < 0.0001), apoptotic signalling (decreased cytochrome C release and Caspase-3/9 activation, all P < 0.001), and muscle protein degradation (decreased 14-kDa actin fragment insoluble/soluble ratio, P < 0.001). CONCLUSIONS Mitochondrial uncoupling by agents such as BAM15 may mitigate age-related decline in muscle mass and function by molecular and cellular bioenergetic adaptations that confer protection against sarcopenic obesity.
Collapse
Affiliation(s)
- Wagner S Dantas
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Elizabeth R M Zunica
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Elizabeth C Heintz
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Bolormaa Vandanmagsar
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Z Elizabeth Floyd
- Ubiquitin Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Yongmei Yu
- Ubiquitin Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Hisashi Fujioka
- Cryo-Electron Microscopy Core, Case Western Reserve University, Cleveland, OH, USA.,Center for Mitochondrial Diseases, Case Western Reserve University of School of Medicine, Cleveland, OH, USA
| | - Charles L Hoppel
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA.,Center for Mitochondrial Diseases, Case Western Reserve University of School of Medicine, Cleveland, OH, USA.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Kathryn P Belmont
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Christopher L Axelrod
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
216
|
Tovy A, Reyes JM, Zhang L, Huang YH, Rosas C, Daquinag AC, Guzman A, Ramabadran R, Chen CW, Gu T, Gupta S, Ortinau L, Park D, Cox AR, Rau RE, Hartig SM, Kolonin MG, Goodell MA. Constitutive loss of DNMT3A causes morbid obesity through misregulation of adipogenesis. eLife 2022; 11:e72359. [PMID: 35635747 PMCID: PMC9150890 DOI: 10.7554/elife.72359] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
DNA Methyltransferase 3 A (DNMT3A) is an important facilitator of differentiation of both embryonic and hematopoietic stem cells. Heterozygous germline mutations in DNMT3A lead to Tatton-Brown-Rahman Syndrome (TBRS), characterized by obesity and excessive height. While DNMT3A is known to impact feeding behavior via the hypothalamus, here we investigated a role in adipocyte progenitors utilizing heterozygous knockout mice that recapitulate cardinal TBRS phenotypes. These mice become morbidly obese due to adipocyte enlargement and tissue expansion. Adipose tissue in these mice exhibited defects in preadipocyte maturation and precocious activation of inflammatory gene networks, including interleukin-6 signaling. Adipocyte progenitor cell lines lacking DNMT3A exhibited aberrant differentiation. Furthermore, mice in which Dnmt3a was specifically ablated in adipocyte progenitors showed enlarged fat depots and increased progenitor numbers, partly recapitulating the TBRS obesity phenotypes. Loss of DNMT3A led to constitutive DNA hypomethylation, such that the DNA methylation landscape of young adipocyte progenitors resemble that of older wild-type mice. Together, our results demonstrate that DNMT3A coordinates both the central and local control of energy storage required to maintain normal weight and prevent inflammatory obesity.
Collapse
Affiliation(s)
- Ayala Tovy
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Jaime M Reyes
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Linda Zhang
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of MedicineHoustonUnited States
| | - Yung-Hsin Huang
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
| | - Carina Rosas
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Alexes C Daquinag
- Institute of Molecular Medicine, McGovern Medical School at the University of Texas Health Science CenterHoustonUnited States
| | - Anna Guzman
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Raghav Ramabadran
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Chun-Wei Chen
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
| | - Tianpeng Gu
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Sinjini Gupta
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Laura Ortinau
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Metabolic and Degenerative Disease, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at HoustonHoustonUnited States
| | - Dongsu Park
- Department of Molecular and Human Genetics, Baylor College of MedicineHoustonUnited States
- Center for Metabolic and Degenerative Disease, Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at HoustonHoustonUnited States
| | - Aaron R Cox
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Rachel E Rau
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Pediatrics, Baylor College of Medicine and Texas Children's HospitalHoustonUnited States
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Baylor College of MedicineHoustonUnited States
| | - Mikhail G Kolonin
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of MedicineHoustonUnited States
| | - Margaret A Goodell
- Stem Cells and Regenerative Medicine Center, Baylor College of MedicineHoustonUnited States
- Department of Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| |
Collapse
|
217
|
Quattrocelli M, Wintzinger M, Miz K, Panta M, Prabakaran AD, Barish GD, Chandel NS, McNally EM. Intermittent prednisone treatment in mice promotes exercise tolerance in obesity through adiponectin. J Exp Med 2022; 219:e20211906. [PMID: 35363257 PMCID: PMC8980841 DOI: 10.1084/jem.20211906] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/21/2021] [Accepted: 02/24/2022] [Indexed: 12/26/2022] Open
Abstract
The fat-muscle communication regulates metabolism and involves circulating signals like adiponectin. Modulation of this cross-talk could benefit muscle bioenergetics and exercise tolerance in conditions like obesity. Chronic daily intake of exogenous glucocorticoids produces or exacerbates metabolic stress, often leading to obesity. In stark contrast to the daily intake, we discovered that intermittent pulses of glucocorticoids improve dystrophic muscle metabolism. However, the underlying mechanisms, particularly in the context of obesity, are still largely unknown. Here we report that in mice with diet-induced obesity, intermittent once-weekly prednisone increased total and high-molecular weight adiponectin levels and improved exercise tolerance and energy expenditure. These effects were dependent upon adiponectin, as shown by genetic ablation of the adipokine. Upregulation of Adipoq occurred through the glucocorticoid receptor (GR), as this effect was blocked by inducible GR ablation in adipocytes. The treatment increased the muscle metabolic response of adiponectin through the CAMKK2-AMPK cascade. Our study demonstrates that intermittent glucocorticoids produce healthful metabolic remodeling in diet-induced obesity.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Michelle Wintzinger
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Karen Miz
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Manoj Panta
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Ashok D. Prabakaran
- Division of Molecular Cardiovascular Biology, Heart Institute, Cincinnati Children’s Hospital Medical Center, and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Grant D. Barish
- Division of Endocrinology, Metabolism and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Navdeep S. Chandel
- Department of Medicine and Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL
| |
Collapse
|
218
|
Picarda E, Galbo PM, Zong H, Rajan MR, Wallenius V, Zheng D, Börgeson E, Singh R, Pessin J, Zang X. The immune checkpoint B7-H3 (CD276) regulates adipocyte progenitor metabolism and obesity development. SCIENCE ADVANCES 2022; 8:eabm7012. [PMID: 35476450 PMCID: PMC9045715 DOI: 10.1126/sciadv.abm7012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/11/2022] [Indexed: 05/09/2023]
Abstract
The immune checkpoint B7-H3 (CD276) is a member of the B7 family that has been studied in the tumor microenvironment and immunotherapy, but its potential role in metabolism remains largely unknown. Here, we show that B7-H3 is highly expressed in mouse and human adipose tissue at steady state, with the highest levels in adipocyte progenitor cells. B7-H3 is rapidly down-regulated upon the initiation of adipocyte differentiation. Combined RNA sequencing and metabolic studies reveal that B7-H3 stimulates glycolytic and mitochondrial activity of adipocyte progenitors. Loss of B7-H3 in progenitors results in impaired oxidative metabolism program and increased lipid accumulation in derived adipocytes. Consistent with these observations, mice knocked out for B7-H3 develop spontaneous obesity, metabolic dysfunction, and adipose tissue inflammation. Our results reveal an unexpected metabolic role for B7-H3 in adipose tissue and open potential new avenues for the treatment of metabolic diseases by targeting the B7-H3 pathway.
Collapse
Affiliation(s)
- Elodie Picarda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Phillip M. Galbo
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Haihong Zong
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Meenu Rohini Rajan
- Institute of Medicine, Department of Molecular and Clinical Medicine, The Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Ville Wallenius
- Department of Gastrosurgical Research and Education, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Departments of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emma Börgeson
- Institute of Medicine, Department of Molecular and Clinical Medicine, The Wallenberg Laboratory and Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Vaestra Goetaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Rajat Singh
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jeffrey Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
219
|
Xia H, Scholtes C, Dufour CR, Ouellet C, Ghahremani M, Giguère V. Insulin action and resistance are dependent on a GSK3β-FBXW7-ERRα transcriptional axis. Nat Commun 2022; 13:2105. [PMID: 35440636 PMCID: PMC9019090 DOI: 10.1038/s41467-022-29722-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/30/2022] [Indexed: 12/15/2022] Open
Abstract
Insulin resistance, a harbinger of the metabolic syndrome, is a state of compromised hormonal response resulting from the dysregulation of a wide range of insulin-controlled cellular processes. However, how insulin affects cellular energy metabolism via long-term transcriptional regulation and whether boosting mitochondrial function alleviates insulin resistance remains to be elucidated. Herein we reveal that insulin directly enhances the activity of the nuclear receptor ERRα via a GSK3β/FBXW7 signaling axis. Liver-specific deletion of GSK3β or FBXW7 and mice harboring mutations of ERRα phosphosites (ERRα3SA) co-targeted by GSK3β/FBXW7 result in accumulated ERRα proteins that no longer respond to fluctuating insulin levels. ERRα3SA mice display reprogrammed liver and muscle transcriptomes, resulting in compromised energy homeostasis and reduced insulin sensitivity despite improved mitochondrial function. This crossroad of insulin signaling and transcriptional control by a nuclear receptor offers a framework to better understand the complex cellular processes contributing to the development of insulin resistance.
Collapse
Affiliation(s)
- Hui Xia
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Charlotte Scholtes
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Catherine R Dufour
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Carlo Ouellet
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Majid Ghahremani
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Vincent Giguère
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada.
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, H3G 1Y6, Canada.
| |
Collapse
|
220
|
Gosis BS, Wada S, Thorsheim C, Li K, Jung S, Rhoades JH, Yang Y, Brandimarto J, Li L, Uehara K, Jang C, Lanza M, Sanford NB, Bornstein MR, Jeong S, Titchenell PM, Biddinger SB, Arany Z. Inhibition of nonalcoholic fatty liver disease in mice by selective inhibition of mTORC1. Science 2022; 376:eabf8271. [PMID: 35420934 PMCID: PMC9811404 DOI: 10.1126/science.abf8271] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) remain without effective therapies. The mechanistic target of rapamycin complex 1 (mTORC1) pathway is a potential therapeutic target, but conflicting interpretations have been proposed for how mTORC1 controls lipid homeostasis. We show that selective inhibition of mTORC1 signaling in mice, through deletion of the RagC/D guanosine triphosphatase-activating protein folliculin (FLCN), promotes activation of transcription factor E3 (TFE3) in the liver without affecting other mTORC1 targets and protects against NAFLD and NASH. Disease protection is mediated by TFE3, which both induces lipid consumption and suppresses anabolic lipogenesis. TFE3 inhibits lipogenesis by suppressing proteolytic processing and activation of sterol regulatory element-binding protein-1c (SREBP-1c) and by interacting with SREBP-1c on chromatin. Our data reconcile previously conflicting studies and identify selective inhibition of mTORC1 as a potential approach to treat NASH and NAFLD.
Collapse
Affiliation(s)
- Bridget S Gosis
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shogo Wada
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chelsea Thorsheim
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina Li
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunhee Jung
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Joshua H Rhoades
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yifan Yang
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeffrey Brandimarto
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Li Li
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kahealani Uehara
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cholsoon Jang
- Department of Biological Chemistry, University of California, Irvine, CA, USA
| | - Matthew Lanza
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan B Sanford
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc R Bornstein
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sunhye Jeong
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Titchenell
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sudha B Biddinger
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zoltan Arany
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
221
|
Mishra I, Xie WR, Bournat JC, He Y, Wang C, Silva ES, Liu H, Ku Z, Chen Y, Erokwu BO, Jia P, Zhao Z, An Z, Flask CA, He Y, Xu Y, Chopra AR. Protein tyrosine phosphatase receptor δ serves as the orexigenic asprosin receptor. Cell Metab 2022; 34:549-563.e8. [PMID: 35298903 PMCID: PMC8986618 DOI: 10.1016/j.cmet.2022.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/25/2021] [Accepted: 02/22/2022] [Indexed: 12/20/2022]
Abstract
Asprosin is a fasting-induced glucogenic and centrally acting orexigenic hormone. The olfactory receptor Olfr734 is known to be the hepatic receptor for asprosin that mediates its effects on glucose production, but the receptor for asprosin's orexigenic function has been unclear. Here, we have identified protein tyrosine phosphatase receptor δ (Ptprd) as the orexigenic receptor for asprosin. Asprosin functions as a high-affinity Ptprd ligand in hypothalamic AgRP neurons, regulating the activity of this circuit in a cell-autonomous manner. Genetic ablation of Ptprd results in a strong loss of appetite, leanness, and an inability to respond to the orexigenic effects of asprosin. Ablation of Ptprd specifically in AgRP neurons causes resistance to diet-induced obesity. Introduction of the soluble Ptprd ligand-binding domain in the circulation of mice suppresses appetite and blood glucose levels by sequestering plasma asprosin. Identification of Ptprd as the orexigenic asprosin receptor creates a new avenue for the development of anti-obesity therapeutics.
Collapse
Affiliation(s)
- Ila Mishra
- Harrington Discovery Institute, Cleveland, OH, USA
| | - Wei Rose Xie
- Harrington Discovery Institute, Cleveland, OH, USA
| | - Juan C Bournat
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Yang He
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chunmei Wang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | - Hailan Liu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Zhiqiang Ku
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yinghua Chen
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, USA
| | - Bernadette O Erokwu
- Departments of Radiology, Biomedical Engineering, and Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhiqiang An
- Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Chris A Flask
- Departments of Radiology, Biomedical Engineering, and Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Yanlin He
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Yong Xu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; USDA-ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Atul R Chopra
- Harrington Discovery Institute, Cleveland, OH, USA; Department of Medicine, Case Western Reserve University, Cleveland, OH, USA; Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
222
|
Lin Y, Bai M, Wang S, Chen L, Li Z, Li C, Cao P, Chen Y. Lactate Is a Key Mediator That Links Obesity to Insulin Resistance via Modulating Cytokine Production From Adipose Tissue. Diabetes 2022; 71:637-652. [PMID: 35044451 DOI: 10.2337/db21-0535] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 01/03/2022] [Indexed: 11/13/2022]
Abstract
Numerous evidence indicates that inflammation in adipose tissue is the primary cause of systemic insulin resistance induced by obesity. Obesity-associated changes in circulating LPS level and hypoxia/HIF-1α activation have been proposed to be involved in boosting obesity-induced inflammation. However, there is poor understanding of what triggers obesity-induced inflammation. In this study, we pinpoint lactate as a key trigger to mediate obesity-induced inflammation and systemic insulin resistance. Specific deletion of Slc16a1 that encodes MCT1, the primary lactate transporter in adipose tissues, robustly elevates blood levels of proinflammatory cytokines and aggravates systemic insulin resistance without alteration of adiposity in mice fed high-fat diet. Slc16a1 deletion in adipocytes elevates intracellular lactate level while reducing circulating lactate concentration. Mechanistically, lactate retention due to Slc16a1 deletion initiates adipocyte apoptosis and cytokine release. The locally recruited macrophages amplify the inflammation by release of proinflammatory cytokines to the circulation, leading to insulin resistance in peripheral tissues. This study, therefore, indicates that lactate within adipocytes has a key biological function linking obesity to insulin resistance, and harnessing lactate in adipocytes can be a promising strategy to break this link.
Collapse
Affiliation(s)
- Yijun Lin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Meijuan Bai
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shuo Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lingling Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zixuan Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Chenchen Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Peijuan Cao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| |
Collapse
|
223
|
Agrud A, Subburaju S, Goel P, Ren J, Kumar AS, Caldarone BJ, Dai W, Chavez J, Fukumura D, Jain RK, Kloner RA, Vasudevan A. Gabrb3 endothelial cell-specific knockout mice display abnormal blood flow, hypertension, and behavioral dysfunction. Sci Rep 2022; 12:4922. [PMID: 35318369 PMCID: PMC8941104 DOI: 10.1038/s41598-022-08806-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/04/2022] [Indexed: 11/17/2022] Open
Abstract
Our recent studies uncovered a novel GABA signaling pathway in embryonic forebrain endothelial cells that works independently from neuronal GABA signaling and revealed that disruptions in endothelial GABAA receptor-GABA signaling from early embryonic stages can directly contribute to the origin of psychiatric disorders. In the GABAA receptor β3 subunit endothelial cell conditional knockout (Gabrb3ECKO) mice, the β3 subunit is deleted selectively from endothelial cells, therefore endothelial GABAA receptors become inactivated and dysfunctional. There is a reduction in vessel densities and increased vessel morphology in the Gabrb3ECKO telencephalon that persists in the adult neocortex. Gabrb3ECKO mice show behavioral deficits such as impaired reciprocal social interactions, communication deficits, heightened anxiety, and depression. Here, we characterize the functional changes in Gabrb3ECKO mice by evaluating cortical blood flow, examine the consequences of loss of endothelial Gabrb3 on cardiac tissue, and define more in-depth altered behaviors. Red blood cell velocity and blood flow were increased in the cortical microcirculation of the Gabrb3ECKO mice. The Gabrb3ECKO mice had a reduction in vessel densities in the heart, similar to the brain; exhibited wavy, myocardial fibers, with elongated 'worm-like' nuclei in their cardiac histology, and developed hypertension. Additional alterations in behavioral function were observed in the Gabrb3ECKO mice such as increased spontaneous exploratory activity and rearing in an open field, reduced short term memory, decreased ambulatory activity in CLAMS testing, and altered prepulse inhibition to startle, an important biomarker of psychiatric diseases such as schizophrenia. Our results imply that vascular Gabrb3 is a key player in the brain as well as the heart, and its loss in both organs can lead to concurrent development of psychiatric and cardiac dysfunction.
Collapse
Affiliation(s)
- Anass Agrud
- grid.280933.30000 0004 0452 8371Angiogenesis and Brain Development Laboratory, Huntington Medical Research Institutes (HMRI), 686 S Fair Oaks Avenue, Pasadena, CA 91105 USA
| | - Sivan Subburaju
- grid.280933.30000 0004 0452 8371Angiogenesis and Brain Development Laboratory, Huntington Medical Research Institutes (HMRI), 686 S Fair Oaks Avenue, Pasadena, CA 91105 USA ,grid.38142.3c000000041936754XDepartment of Psychiatry, Harvard Medical School, Boston, MA 02215 USA ,grid.240206.20000 0000 8795 072XDivision of Basic Neuroscience, McLean Hospital, 115 Mill Street, Belmont, MA 02478 USA
| | - Pranay Goel
- grid.280933.30000 0004 0452 8371Angiogenesis and Brain Development Laboratory, Huntington Medical Research Institutes (HMRI), 686 S Fair Oaks Avenue, Pasadena, CA 91105 USA
| | - Jun Ren
- grid.32224.350000 0004 0386 9924Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Ashwin Srinivasan Kumar
- grid.32224.350000 0004 0386 9924Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA ,grid.116068.80000 0001 2341 2786Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Barbara J. Caldarone
- grid.38142.3c000000041936754XMouse Behavior Core, Department of Genetics, Harvard Medical School, Boston, MA USA
| | - Wangde Dai
- grid.280933.30000 0004 0452 8371Huntington Medical Research Institutes, Pasadena, CA USA ,grid.42505.360000 0001 2156 6853Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA USA
| | - Jesus Chavez
- grid.280933.30000 0004 0452 8371Huntington Medical Research Institutes, Pasadena, CA USA ,grid.42505.360000 0001 2156 6853Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA USA
| | - Dai Fukumura
- grid.32224.350000 0004 0386 9924Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Rakesh K. Jain
- grid.32224.350000 0004 0386 9924Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| | - Robert A. Kloner
- grid.280933.30000 0004 0452 8371Huntington Medical Research Institutes, Pasadena, CA USA ,grid.42505.360000 0001 2156 6853Division of Cardiovascular Medicine, Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, CA USA
| | - Anju Vasudevan
- Angiogenesis and Brain Development Laboratory, Huntington Medical Research Institutes (HMRI), 686 S Fair Oaks Avenue, Pasadena, CA, 91105, USA.
| |
Collapse
|
224
|
Li Y, Wang D, Ping X, Zhang Y, Zhang T, Wang L, Jin L, Zhao W, Guo M, Shen F, Meng M, Chen X, Zheng Y, Wang J, Li D, Zhang Q, Hu C, Xu L, Ma X. Local hyperthermia therapy induces browning of white fat and treats obesity. Cell 2022; 185:949-966.e19. [PMID: 35247329 DOI: 10.1016/j.cell.2022.02.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/28/2021] [Accepted: 02/02/2022] [Indexed: 02/08/2023]
Abstract
Beige fat plays key roles in the regulation of systemic energy homeostasis; however, detailed mechanisms and safe strategy for its activation remain elusive. In this study, we discovered that local hyperthermia therapy (LHT) targeting beige fat promoted its activation in humans and mice. LHT achieved using a hydrogel-based photothermal therapy activated beige fat, preventing and treating obesity in mice without adverse effects. HSF1 is required for the effects since HSF1 deficiency blunted the metabolic benefits of LHT. HSF1 regulates Hnrnpa2b1 (A2b1) transcription, leading to increased mRNA stability of key metabolic genes. Importantly, analysis of human association studies followed by functional analysis revealed that the HSF1 gain-of-function variant p.P365T is associated with improved metabolic performance in humans and increased A2b1 transcription in mice and cells. Overall, we demonstrate that LHT offers a promising strategy against obesity by inducing beige fat activation via HSF1-A2B1 transcriptional axis.
Collapse
Affiliation(s)
- Yu Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Dongmei Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xiaodan Ping
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yankang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ting Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Li Jin
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Wenjun Zhao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Mingwei Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Fei Shen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Meiyao Meng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Xin Chen
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Ying Zheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiang Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China; Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China.
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China; Department of Endocrinology and Metabolism, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China; Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology and School of Life Sciences, East China Normal University, Shanghai 200241, China.
| |
Collapse
|
225
|
Chen Y, Fernandez EA, Roger C, Lopez-Mejia IC, Fajas Coll L, Ji H. Adipocyte-Specific CDK7 Ablation Leads to Progressive Loss of Adipose Tissue and Metabolic Dysfunction. FEBS Lett 2022; 596:1434-1444. [PMID: 35294049 DOI: 10.1002/1873-3468.14335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 11/12/2022]
Abstract
Adipose tissue regulates whole-body energy homeostasis. Both lipodystrophy and obesity, the extreme and opposite aspects of adipose tissue dysfunction, result in metabolic disorders: insulin resistance and hepatic steatosis. Cyclin-dependent kinases (CDKs) have been reported to be involved in adipose tissue development and functions. Using adipose tissue-specific knockout mice, here we demonstrate that the deletion of CDK7 in adipose tissue results in progressive lipodystrophy, insulin resistance, impaired adipokine secretion and down-regulation of fat-specific genes, which are aggravated on high-fat diet and during aging. Our studies suggest that CDK7 is a key regulatory component of adipose tissue maintenance and systemic energy homeostasis.
Collapse
Affiliation(s)
- Yizhe Chen
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Eric Aria Fernandez
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Catherine Roger
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | | | - Lluis Fajas Coll
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Institut National de la Santé et de la Recherche Médicale (Inserm), Languedoc Roussillon, France
| | - Honglei Ji
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.,Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
| |
Collapse
|
226
|
Meng H, Gonzales NM, Jung SY, Lu Y, Putluri N, Zhu B, Dacso CC, Lonard DM, O'Malley BW. Defining the mammalian coactivation of hepatic 12-h clock and lipid metabolism. Cell Rep 2022; 38:110491. [PMID: 35263593 PMCID: PMC8958721 DOI: 10.1016/j.celrep.2022.110491] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/05/2021] [Accepted: 02/14/2022] [Indexed: 12/18/2022] Open
Abstract
The 12-h clock coordinates lipid homeostasis, energy metabolism, and stress rhythms via the transcriptional regulator XBP1. However, the biochemical and physiological bases for integrated control of the 12-h clock and diverse metabolic pathways remain unclear. Here, we show that steroid receptor coactivator SRC-3 coactivates XBP1 transcription and regulates hepatic 12-h cistrome and gene rhythmicity. Mice lacking SRC-3 show abnormal 12-h rhythms in hepatic transcription, metabolic functions, systemic energetics, and rate-limiting lipid metabolic processes, including triglyceride, phospholipid, and cardiolipin pathways. Notably, 12-h clock coactivation is not only preserved, with its cistromic activation priming ahead of the zeitgeber cue of light, but concomitant with rhythmic remodeling in the absence of food. These findings reveal that SRC-3 integrates the mammalian 12-h clock, energy metabolism, and membrane and lipid homeostasis and demonstrates a role for the 12-h clock machinery as an active transcriptional mechanism in anticipating physiological and metabolic energy needs and stresses.
Collapse
Affiliation(s)
- Huan Meng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Naomi M Gonzales
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sung Yun Jung
- Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Science Park, Smithville, TX 78957, USA
| | - Nagireddy Putluri
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bokai Zhu
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Clifford C Dacso
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - David M Lonard
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bert W O'Malley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
227
|
Wilson RJ, Lyons SP, Koves TR, Bryson VG, Zhang H, Li T, Crown SB, Ding JD, Grimsrud PA, Rosenberg PB, Muoio DM. Disruption of STIM1-mediated Ca 2+ sensing and energy metabolism in adult skeletal muscle compromises exercise tolerance, proteostasis, and lean mass. Mol Metab 2022; 57:101429. [PMID: 34979330 PMCID: PMC8814391 DOI: 10.1016/j.molmet.2021.101429] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Stromal interaction molecule 1 (STIM1) is a single-pass transmembrane endoplasmic/sarcoplasmic reticulum (E/SR) protein recognized for its role in a store operated Ca2+ entry (SOCE), an ancient and ubiquitous signaling pathway. Whereas STIM1 is known to be indispensable during development, its biological and metabolic functions in mature muscles remain unclear. METHODS Conditional and tamoxifen inducible muscle STIM1 knock-out mouse models were coupled with multi-omics tools and comprehensive physiology to understand the role of STIM1 in regulating SOCE, mitochondrial quality and bioenergetics, and whole-body energy homeostasis. RESULTS This study shows that STIM1 is abundant in adult skeletal muscle, upregulated by exercise, and is present at SR-mitochondria interfaces. Inducible tissue-specific deletion of STIM1 (iSTIM1 KO) in adult muscle led to diminished lean mass, reduced exercise capacity, and perturbed fuel selection in the settings of energetic stress, without affecting whole-body glucose tolerance. Proteomics and phospho-proteomics analyses of iSTIM1 KO muscles revealed molecular signatures of low-grade E/SR stress and broad activation of processes and signaling networks involved in proteostasis. CONCLUSION These results show that STIM1 regulates cellular and mitochondrial Ca2+ dynamics, energy metabolism and proteostasis in adult skeletal muscles. Furthermore, these findings provide insight into the pathophysiology of muscle diseases linked to disturbances in STIM1-dependent Ca2+ handling.
Collapse
Affiliation(s)
- Rebecca J Wilson
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA
| | - Scott P Lyons
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA
| | - Timothy R Koves
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Geriatrics, Duke University School of Medicine, Durham, NC 27705, USA
| | - Victoria G Bryson
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Hengtao Zhang
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - TianYu Li
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Scott B Crown
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA
| | - Jin-Dong Ding
- Department of Medicine, Division of Ophthalmology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Paul A Grimsrud
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, NC 27705, USA
| | - Paul B Rosenberg
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Cardiology, Duke University School of Medicine, Durham, NC 27705, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute, and Sarah W. Stedman Nutrition and Metabolism Center, Duke University School of Medicine, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University School of Medicine, Durham, NC 27705, USA; Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27705, USA.
| |
Collapse
|
228
|
Bartoli F, Debant M, Chuntharpursat-Bon E, Evans EL, Musialowski KE, Parsonage G, Morley LC, Futers TS, Sukumar P, Bowen TS, Kearney MT, Lichtenstein L, Roberts LD, Beech DJ. Endothelial Piezo1 sustains muscle capillary density and contributes to physical activity. J Clin Invest 2022; 132:141775. [PMID: 35025768 PMCID: PMC8884896 DOI: 10.1172/jci141775] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 01/11/2022] [Indexed: 11/17/2022] Open
Abstract
Piezo1 forms mechanically activated nonselective cation channels that contribute to endothelial response to fluid flow. Here we reveal an important role in the control of capillary density. Conditional endothelial cell-specific deletion of Piezo1 in adult mice depressed physical performance. Muscle microvascular endothelial cell apoptosis and capillary rarefaction were evident and sufficient to account for the effect on performance. There was selective upregulation of thrombospondin-2 (TSP2), an inducer of endothelial cell apoptosis, with no effect on TSP1, a related important player in muscle physiology. TSP2 was poorly expressed in muscle endothelial cells but robustly expressed in muscle pericytes, in which nitric oxide (NO) repressed the Tsp2 gene without an effect on Tsp1. In endothelial cells, Piezo1 was required for normal expression of endothelial NO synthase. The data suggest an endothelial cell-pericyte partnership of muscle in which endothelial Piezo1 senses blood flow to sustain capillary density and thereby maintain physical capability.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - T. Scott Bowen
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | | | | | | | |
Collapse
|
229
|
Reho JJ, Nakagawa P, Mouradian GC, Grobe CC, Saravia FL, Burnett CML, Kwitek AE, Kirby JR, Segar JL, Hodges MR, Sigmund CD, Grobe JL. Methods for the Comprehensive in vivo Analysis of Energy Flux, Fluid Homeostasis, Blood Pressure, and Ventilatory Function in Rodents. Front Physiol 2022; 13:855054. [PMID: 35283781 PMCID: PMC8914175 DOI: 10.3389/fphys.2022.855054] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/03/2022] [Indexed: 01/22/2023] Open
Abstract
Cardiovascular disease represents the leading cause of death in the United States, and metabolic diseases such as obesity represent the primary impediment to improving cardiovascular health. Rodent (mouse and rat) models are widely used to model cardiometabolic disease, and as a result, there is increasing interest in the development of accurate and precise methodologies with sufficiently high resolution to dissect mechanisms controlling cardiometabolic physiology in these small organisms. Further, there is great utility in the development of centralized core facilities furnished with high-throughput equipment configurations and staffed with professional content experts to guide investigators and ensure the rigor and reproducibility of experimental endeavors. Here, we outline the array of specialized equipment and approaches that are employed within the Comprehensive Rodent Metabolic Phenotyping Core (CRMPC) and our collaborating laboratories within the Departments of Physiology, Pediatrics, Microbiology & Immunology, and Biomedical Engineering at the Medical College of Wisconsin (MCW), for the detailed mechanistic dissection of cardiometabolic function in mice and rats. We highlight selected methods for the analysis of body composition and fluid compartmentalization, electrolyte accumulation and flux, energy accumulation and flux, physical activity, ingestive behaviors, ventilatory function, blood pressure, heart rate, autonomic function, and assessment and manipulation of the gut microbiota. Further, we include discussion of the advantages and disadvantages of these approaches for their use with rodent models, and considerations for experimental designs using these methods.
Collapse
Affiliation(s)
- John J. Reho
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Pablo Nakagawa
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Gary C. Mouradian
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Connie C. Grobe
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Fatima L. Saravia
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Colin M. L. Burnett
- Department of Internal Medicine, University of Iowa Hospitals & Clinics, Iowa City, IA, United States
| | - Anne E. Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States
| | - John R. Kirby
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Jeffrey L. Segar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Matthew R. Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Curt D. Sigmund
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States,Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI, United States,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, United States,Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI, United States,*Correspondence: Justin L. Grobe,
| |
Collapse
|
230
|
Williams AS, Koves TR, Pettway YD, Draper JA, Slentz DH, Grimsrud PA, Ilkayeva OR, Muoio DM. Nicotinamide riboside supplementation confers marginal metabolic benefits in obese mice without remodeling the muscle acetyl-proteome. iScience 2022; 25:103635. [PMID: 35028529 PMCID: PMC8741497 DOI: 10.1016/j.isci.2021.103635] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/22/2021] [Accepted: 12/14/2021] [Indexed: 12/20/2022] Open
Abstract
Nicotinamide riboside supplements (NRS) have been touted as a nutraceutical that promotes cardiometabolic and musculoskeletal health by enhancing nicotinamide adenine dinucleotide (NAD+) biosynthesis, mitochondrial function, and/or the activities of NAD-dependent sirtuin deacetylase enzymes. This investigation examined the impact of NRS on whole body energy homeostasis, skeletal muscle mitochondrial function, and corresponding shifts in the acetyl-lysine proteome, in the context of diet-induced obesity using C57BL/6NJ mice. The study also included a genetically modified mouse model that imposes greater demand on sirtuin flux and associated NAD+ consumption, specifically within muscle tissues. In general, whole body glucose control was marginally improved by NRS when administered at the midpoint of a chronic high-fat diet, but not when given as a preventative therapy upon initiation of the diet. Contrary to anticipated outcomes, the study produced little evidence that NRS increases tissue NAD+ levels, augments mitochondrial function, and/or mitigates diet-induced hyperacetylation of the skeletal muscle proteome.
Collapse
Affiliation(s)
- Ashley S. Williams
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Timothy R. Koves
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
- Divison of Geriatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Yasminye D. Pettway
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - James A. Draper
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Dorothy H. Slentz
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Paul A. Grimsrud
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Olga R. Ilkayeva
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah M. Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
- Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
231
|
Cao X, Shi T, Zhang C, Jin W, Song L, Zhang Y, Liu J, Yang F, Rotimi CN, Xu A, Yang J. ACE2 pathway regulates thermogenesis and energy metabolism. eLife 2022; 11:72266. [PMID: 35014608 PMCID: PMC8776250 DOI: 10.7554/elife.72266] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 01/09/2022] [Indexed: 12/02/2022] Open
Abstract
Identification of key regulators of energy homeostasis holds important therapeutic promise for metabolic disorders, such as obesity and diabetes. ACE2 cleaves angiotensin II (Ang II) to generate Ang-(1-7) which acts mainly through the Mas1 receptor. Here, we identify ACE2 pathway as a critical regulator in the maintenance of thermogenesis and energy expenditure. We found that ACE2 is highly expressed in brown adipose tissue (BAT) and that cold stimulation increases ACE2 and Ang-(1-7) levels in BAT and serum. Ace2 knockout mice (Ace2-/y) and Mas1 knockout mice (Mas1-/-) displayed impaired thermogenesis. Mice transplanted with brown adipose tissue from Mas1-/- display metabolic abnormalities consistent with those seen in the Ace2 and Mas1 knockout mice. In contrast, impaired thermogenesis of Leprdb/db obese diabetic mice and high-fat diet-induced obese mice were ameliorated by overexpression of Ace2 or continuous infusion of Ang-(1-7). Activation of ACE2 pathway was associated with improvement of metabolic parameters, including blood glucose, lipids, and energy expenditure in multiple animal models. Consistently, ACE2 pathway remarkably enhanced the browning of white adipose tissue. Mechanistically, we showed that ACE2 pathway activated Akt/FoxO1 and PKA pathway, leading to induction of UCP1 and activation of mitochondrial function. Our data propose that adaptive thermogenesis requires regulation of ACE2 pathway and highlight novel potential therapeutic targets for the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Xi Cao
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Tingting Shi
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chuanhai Zhang
- Department of Physiology, University of Texas Meical Center at Dallas, Dallas, United States
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lini Song
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yichen Zhang
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jingyi Liu
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Fangyuan Yang
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Charles N Rotimi
- National Human Genome Research Institute, National Institutes of Health, Bethesda, United States
| | - Aimin Xu
- Department of Medicine, University of Hong Kong, Hong Kong, Hong Kong
| | - Jinkui Yang
- Beijing Diabetes Institute, Beijing Key Laboratory of Diabetes Research and Care, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
232
|
Ni B, Chen S, Farrar JS, Celi FS. Metabolic Phenotyping in Mice with NASH Using Indirect Calorimetry. Methods Mol Biol 2022; 2455:223-232. [PMID: 35212997 DOI: 10.1007/978-1-0716-2128-8_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Obesity caused by caloric overload has assumed epidemic proportions. Obesity is frequently associated with metabolic dysfunctions, such as type 2 diabetes, non-alcoholic steatohepatitis (NASH), cardiovascular diseases, and cancer. Metabolic phenotyping is a set of techniques for studying metabolic dysfunction and behavior information including energy expenditure, body weight gain, glucose homeostasis, and lipid profile. Among different metabolic phenotyping methods, indirect calorimetry is an indispensable tool for quantifying the energy balance/imbalance in various mouse models, which enables researchers to probe the development of disease and to evaluate the therapeutic benefit from different interventions. In this chapter, we will describe the procedures of metabolic phenotyping using indirect calorimetry in db/db mouse, a metabolic disorder mouse model which develops NASH.
Collapse
Affiliation(s)
- Bin Ni
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA.
| | - Shanshan Chen
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, USA
| | - Jared S Farrar
- Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, USA
| | - Francesco S Celi
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
233
|
Human loss-of-function variants in the serotonin 2C receptor associated with obesity and maladaptive behavior. Nat Med 2022; 28:2537-2546. [PMID: 36536256 PMCID: PMC9800280 DOI: 10.1038/s41591-022-02106-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/25/2022] [Indexed: 12/24/2022]
Abstract
Serotonin reuptake inhibitors and receptor agonists are used to treat obesity, anxiety and depression. Here we studied the role of the serotonin 2C receptor (5-HT2CR) in weight regulation and behavior. Using exome sequencing of 2,548 people with severe obesity and 1,117 control individuals without obesity, we identified 13 rare variants in the gene encoding 5-HT2CR (HTR2C) in 19 unrelated people (3 males and 16 females). Eleven variants caused a loss of function in HEK293 cells. All people who carried variants had hyperphagia and some degree of maladaptive behavior. Knock-in male mice harboring a human loss-of-function HTR2C variant developed obesity and reduced social exploratory behavior; female mice heterozygous for the same variant showed similar deficits with reduced severity. Using the 5-HT2CR agonist lorcaserin, we found that depolarization of appetite-suppressing proopiomelanocortin neurons was impaired in knock-in mice. In conclusion, we demonstrate that 5-HT2CR is involved in the regulation of human appetite, weight and behavior. Our findings suggest that melanocortin receptor agonists might be effective in treating severe obesity in individuals carrying HTR2C variants. We suggest that HTR2C should be included in diagnostic gene panels for severe childhood-onset obesity.
Collapse
|
234
|
Ma QX, Zhu WY, Lu XC, Jiang D, Xu F, Li JT, Zhang L, Wu YL, Chen ZJ, Yin M, Huang HY, Lei QY. BCAA-BCKA axis regulates WAT browning through acetylation of PRDM16. Nat Metab 2022; 4:106-122. [PMID: 35075301 DOI: 10.1038/s42255-021-00520-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/09/2021] [Indexed: 12/11/2022]
Abstract
The link between branched-chain amino acids (BCAAs) and obesity has been known for decades but the functional role of BCAA metabolism in white adipose tissue (WAT) of obese individuals remains vague. Here, we show that mice with adipose tissue knockout of Bcat2, which converts BCAAs to branched-chain keto acids (BCKAs), are resistant to high-fat diet-induced obesity due to increased inguinal WAT browning and thermogenesis. Mechanistically, acetyl-CoA derived from BCKA suppresses WAT browning by acetylation of PR domain-containing protein 16 (PRDM16) at K915, disrupting the interaction between PRDM16 and peroxisome proliferator-activated receptor-γ (PPARγ) to maintain WAT characteristics. Depletion of BCKA-derived acetyl-CoA robustly prompts WAT browning and energy expenditure. In contrast, BCKA supplementation re-establishes high-fat diet-induced obesity in Bcat2 knockout mice. Moreover, telmisartan, an anti-hypertension drug, significantly represses Bcat2 activity via direct binding, resulting in enhanced WAT browning and reduced adiposity. Strikingly, BCKA supplementation reverses the lean phenotype conferred by telmisartan. Thus, we uncover the critical role of the BCAA-BCKA axis in WAT browning.
Collapse
Affiliation(s)
- Qi-Xiang Ma
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wen-Ying Zhu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiao-Chen Lu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Duo Jiang
- Key Laboratory of Metabolism and Molecular Medicine of Chinese Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Feng Xu
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Jin-Tao Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ying-Li Wu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng-Jun Chen
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Miao Yin
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Hai-Yan Huang
- Key Laboratory of Metabolism and Molecular Medicine of Chinese Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Qun-Ying Lei
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences; Cancer Institutes; Key Laboratory of Breast Cancer in Shanghai; Shanghai Key Laboratory of Radiation Oncology; The Shanghai Key Laboratory of Medical Epigenetics; Department of Oncology; State Key Laboratory of Medical Neurobiology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
235
|
Centanni SW, Conley SY, Luchsinger JR, Lantier L, Winder DG. The impact of intermittent exercise on mouse ethanol drinking and abstinence-associated affective behavior and physiology. Alcohol Clin Exp Res 2022; 46:114-128. [PMID: 34773282 PMCID: PMC9152923 DOI: 10.1111/acer.14742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/04/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Negative emotional states are associated with the initiation and maintenance of alcohol use and drive relapse to drinking during withdrawal and protracted abstinence. Physical exercise is correlated with decreased negative affective symptoms, although a direct relationship between drinking patterns and exercise level has not been fully elucidated. METHODS We incorporated intermittent running wheel access into a chronic continuous access, two-bottle choice alcohol drinking model in female C57BL/6J mice. Wheel access was granted intermittently once mice established a preference for alcohol over water. After 6 weeks, alcohol was removed (forced abstinence) and mice were given continuous access to unlocked or locked wheels. Negative affect-like behavior, home cage behavior, and metabolic activity were measured during protracted abstinence. RESULTS Wheel access shifted drinking patterns in the mice, increasing drinking when the wheel was locked, and decreasing drinking when unlocked. Moreover, alcohol preference and consumption were strongly negatively correlated with the amount of running. An assessment of negative affect-like behavior in abstinence via the novelty suppressed feeding and saccharin preference tests (SPT) showed that unlimited wheel access mitigated abstinence-induced latency increases. Mice in abstinence also spent more time sleeping during the active dark cycle than control mice, providing additional evidence for abstinence-induced anhedonia- and depression-like behavior. Furthermore, running wheel access in abstinence decreased dark cycle sleep to comparable alcohol- and wheel-naïve mice. Given the positive impact of exercise and the negative impact of alcohol on metabolic health, we compared metabolic phenotypes of alcohol-abstinent mice with and without wheel access. Wheel access increased energy expenditure, carbon dioxide production, and oxygen consumption, providing a potential metabolic mechanism through which wheel access improves affective state. CONCLUSIONS This study suggests that including exercise in AUD treatment regimens has the potential to reduce drinking, improve affective state during abstinence and could serve as a non-pharmacological approach to prevent the development of an AUD in high-risk individuals.
Collapse
Affiliation(s)
- Samuel W. Centanni
- Vanderbilt Center for Addiction Research
- Molecular Physiology & Biophysics, the
- Vanderbilt Brain Institute
- Vanderbilt J.F. Kennedy Center for Research on Human Development
| | | | - Joseph R. Luchsinger
- Vanderbilt Center for Addiction Research
- Molecular Physiology & Biophysics, the
- Vanderbilt Brain Institute
- Vanderbilt J.F. Kennedy Center for Research on Human Development
| | - Louise Lantier
- Molecular Physiology & Biophysics, the
- Vanderbilt J.F. Kennedy Center for Research on Human Development
- Department of Psychiatry and Behavioral Sciences
- Mouse Metabolic Phenotyping Center, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research
- Molecular Physiology & Biophysics, the
- Vanderbilt Brain Institute
- Vanderbilt J.F. Kennedy Center for Research on Human Development
- Department of Psychiatry and Behavioral Sciences
- Mouse Metabolic Phenotyping Center, Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
236
|
Liu X, Wang Z, Song X, Chang X, Zu E, Ma X, Sukegawa M, Liu D, Wang DO. Crocetin Alleviates Ovariectomy-Induced Metabolic Dysfunction through Regulating Estrogen Receptor β. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14824-14839. [PMID: 34851635 DOI: 10.1021/acs.jafc.1c04570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Metabolic dysfunction (MD) is a major health problem threatening the life quality of menopausal women. Saffron has been widely used in herb prescriptions for treating menopausal syndrome. However, the pharmacological effects and mechanisms of saffron are poorly understood. Here, we investigated the effect of crocin, the major ingredient of saffron and its active metabolite in blood, crocetin, on MD and lipid metabolism in ovariectomized (OVX) mice and 3T3-L1 adipocytes. The present study showed that intragastric treatment of crocin prevented weight gain, fat accumulation, and insulin resistance in OVX mice by increasing energy expenditure and fat oxidation. Mechanistically, crocin influenced adipose tissue homeostasis by regulating adipogenic and lipolytic factors, which was strongly associated with the restoration of the downregulated ERβ function in white adipose tissue (WAT). In vitro, crocetin facilitated lipid metabolism in an ERβ-dependent manner. Our results demonstrated the beneficial effects of crocetin/crocin-mediated intervention against metabolic dysfunction, revealing a prospective therapeutic application in menopausal women.
Collapse
Affiliation(s)
- Xiaoling Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ziqi Wang
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xintong Song
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinyu Chang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Er Zu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiaowei Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Momoe Sukegawa
- Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
- Graduate School of Biostudies, Kyoto University, Yoshida hon-machi, Kyoto 606-8501, Japan
| | - Dongchun Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Dan Ohtan Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
- Center for Biosystems Dynamics Research, RIKEN, 2-2-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
- Graduate School of Biostudies, Kyoto University, Yoshida hon-machi, Kyoto 606-8501, Japan
| |
Collapse
|
237
|
Rekhi UR, Omar M, Alexiou M, Delyea C, Immaraj L, Elahi S, Febbraio M. Endothelial Cell CD36 Reduces Atherosclerosis and Controls Systemic Metabolism. Front Cardiovasc Med 2021; 8:768481. [PMID: 34888367 PMCID: PMC8650007 DOI: 10.3389/fcvm.2021.768481] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/20/2021] [Indexed: 01/08/2023] Open
Abstract
High-fat Western diets contribute to tissue dysregulation of fatty acid and glucose intake, resulting in obesity and insulin resistance and their sequelae, including atherosclerosis. New therapies are desperately needed to interrupt this epidemic. The significant idea driving this research is that the understudied regulation of fatty acid entry into tissues at the endothelial cell (EC) interface can provide novel therapeutic targets that will greatly modify health outcomes and advance health-related knowledge. Dysfunctional endothelium, defined as activated, pro-inflammatory, and pro-thrombotic, is critical in atherosclerosis initiation, in modulating thrombotic events that could result in myocardial infarction and stroke, and is a hallmark of insulin resistance. Dyslipidemia from high-fat diets overwhelmingly contributes to the development of dysfunctional endothelium. CD36 acts as a receptor for pathological ligands generated by high-fat diets and in fatty acid uptake, and therefore, it may additionally contribute to EC dysfunction. We created EC CD36 knockout (CD36°) mice using cre-lox technology and a cre-promoter that does not eliminate CD36 in hematopoietic cells (Tie2e cre). These mice were studied on different diets, and crossed to the low density lipoprotein receptor (LDLR) knockout for atherosclerosis assessment. Our data show that EC CD36° and EC CD36°/LDLR° mice have metabolic changes suggestive of an uncompensated role for EC CD36 in fatty acid uptake. The mice lacking expression of EC CD36 had increased glucose clearance compared with controls when fed with multiple diets. EC CD36° male mice showed increased carbohydrate utilization and decreased energy expenditure by indirect calorimetry. Female EC CD36°/LDLR° mice have reduced atherosclerosis. Taken together, these data support a significant role for EC CD36 in systemic metabolism and reveal sex-specific impact on atherosclerosis and energy substrate use.
Collapse
Affiliation(s)
- Umar R Rekhi
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mohamed Omar
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Maria Alexiou
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Cole Delyea
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada
| | - Linnet Immaraj
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shokrollah Elahi
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Maria Febbraio
- Department of Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
238
|
Wu D, Eeda V, Undi RB, Mann S, Stout M, Lim HY, Wang W. A novel peroxisome proliferator-activated receptor gamma ligand improves insulin sensitivity and promotes browning of white adipose tissue in obese mice. Mol Metab 2021; 54:101363. [PMID: 34710641 PMCID: PMC8627988 DOI: 10.1016/j.molmet.2021.101363] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/18/2021] [Accepted: 10/20/2021] [Indexed: 11/23/2022] Open
Abstract
OBJECTIVE Nuclear receptor Peroxisome Proliferator-Activated Receptor γ (PPARγ) is a promising target for the treatment of type 2 diabetes. The antidiabetic drug thiazolidinediones (TZDs) are potent insulin sensitizers as full agonists of PPARγ, but cause unwanted side effects. Recent discoveries have shown that TZDs improve insulin sensitivity by blocking PPARγ phosphorylation at S273, which decouples the full agonism-associated side effects. PPARγ ligands that act through the blockage of PPARγ phosphorylation but lack the full agonist activity would be expected to improve insulin sensitivity without TZD-associated side effects, however, chemicals that carry such traits and bind to PPARγ with high-affinity are lacking. Moreover, TZDs are known to promote white-to-brown adipocyte conversion and energy expenditure and appear to require their full agonism on PPARγ for this activity. It is unknown whether a partial or non-TZD agonist of PPARγ is capable of promoting browning effect. In this study, we developed a novel non-TZD partial agonist of PPARγ and investigated its function on insulin sensitivity and white-to-brown conversion and energy expenditure in diet-induced obese mice. METHODS A novel indole-based chemical WO95E was designed via medicinal chemistry and tested for PPARγ binding and activity and for the effect on PPARγ phosphorylation. Diet-induced obese mice were administered with WO95E for 4 weeks. Insulin sensitivity, glucose tolerance, body weight, fat tissue weight, adipocyte size, morphology, energy expenditure, and expression levels of genes involved in PPARγ activity, thermogenesis/browning, and TZD-related side effects were evaluated. RESULTS WO95E binds to PPARγ with high affinity and acts as a PPARγ partial agonist. WO95E inhibits PPARγ phosphorylation and regulates PPARγ phosphorylation-dependent genes. WO95E ameliorates insulin resistance and glucose tolerance in mice of diet-induced obesity, with minimal TZD use-associated side effects. We found that WO95E promotes white-to-brown adipocyte conversion and energy expenditure and hence protects against diet-induced obesity. WO95E decreases the size of adipocytes and suppresses adipose tissue inflammation. WO95E also suppresses obesity-associated liver steatosis. CONCLUSIONS WO95E improves insulin sensitivity and glucose homeostasis and promotes browning and energy expenditure by acting as a novel PPARγ phosphorylation inhibitor/partial agonist. Our findings suggest the potential of this compound or its derivative for the therapeutic treatment of insulin resistance and obesity.
Collapse
Affiliation(s)
- Dan Wu
- Department of Medicine, Division of Endocrinology, USA
| | | | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, OK 73104, USA
| | - Shivani Mann
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael Stout
- Aging & Metabolism Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Hui-Ying Lim
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, OK 73104, USA
| | - Weidong Wang
- Department of Medicine, Division of Endocrinology, USA.
| |
Collapse
|
239
|
Li F, Cui X, Jing J, Wang S, Shi H, Xue B, Shi H. Brown Fat Dnmt3b Deficiency Ameliorates Obesity in Female Mice. Life (Basel) 2021; 11:life11121325. [PMID: 34947856 PMCID: PMC8703316 DOI: 10.3390/life11121325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/26/2021] [Accepted: 11/26/2021] [Indexed: 11/16/2022] Open
Abstract
Obesity results from a chronic energy imbalance due to energy intake exceeding energy expenditure. Activation of brown fat thermogenesis has been shown to combat obesity. Epigenetic regulation, including DNA methylation, has emerged as a key regulator of brown fat thermogenic function. Here we aimed to study the role of Dnmt3b, a DNA methyltransferase involved in de novo DNA methylation, in the regulation of brown fat thermogenesis and obesity. We found that the specific deletion of Dnmt3b in brown fat promotes the thermogenic and mitochondrial program in brown fat, enhances energy expenditure, and decreases adiposity in female mice fed a regular chow diet. With a lean phenotype, the female knockout mice also exhibit increased insulin sensitivity. In addition, Dnmt3b deficiency in brown fat also prevents diet-induced obesity and insulin resistance in female mice. Interestingly, our RNA-seq analysis revealed an upregulation of the PI3K-Akt pathway in the brown fat of female Dnmt3b knockout mice. However, male Dnmt3b knockout mice have no change in their body weight, suggesting the existence of sexual dimorphism in the brown fat Dnmt3b knockout model. Our data demonstrate that Dnmt3b plays an important role in the regulation of brown fat function, energy metabolism and obesity in female mice.
Collapse
Affiliation(s)
- Fenfen Li
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Xin Cui
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Jia Jing
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Shirong Wang
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
- Correspondence: (B.X.); (H.S.)
| | - Hang Shi
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA; (F.L.); (X.C.); (J.J.); (S.W.)
- Correspondence: (B.X.); (H.S.)
| |
Collapse
|
240
|
Boland BB, Laker RC, O'Brien S, Sitaula S, Sermadiras I, Nielsen JC, Barkholt P, Roostalu U, Hecksher-Sørensen J, Sejthen SR, Thorbek DD, Suckow A, Burmeister N, Oldham S, Will S, Howard VG, Gill BM, Newton P, Naylor J, Hornigold DC, Austin J, Lantier L, McGuinness OP, Trevaskis JL, Grimsby JS, Rhodes CJ. Peptide-YY 3-36/glucagon-like peptide-1 combination treatment of obese diabetic mice improves insulin sensitivity associated with recovered pancreatic β-cell function and synergistic activation of discrete hypothalamic and brainstem neuronal circuitries. Mol Metab 2021; 55:101392. [PMID: 34781035 PMCID: PMC8717237 DOI: 10.1016/j.molmet.2021.101392] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/22/2021] [Accepted: 11/04/2021] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE Obesity-linked type 2 diabetes (T2D) is a worldwide health concern and many novel approaches are being considered for its treatment and subsequent prevention of serious comorbidities. Co-administration of glucagon like peptide 1 (Fc-GLP-1) and peptide YY3-36 (Fc-PYY3-36) renders a synergistic decrease in energy intake in obese men. However, mechanistic details of the synergy between these peptide agonists and their effects on metabolic homeostasis remain relatively scarce. METHODS In this study, we utilized long-acting analogues of GLP-1 and PYY3-36 (via Fc-peptide conjugation) to better characterize the synergistic pharmacological benefits of their co-administration on body weight and glycaemic regulation in obese and diabetic mouse models. Hyperinsulinemic-euglycemic clamps were used to measure weight-independent effects of Fc-PYY3-36 + Fc-GLP-1 on insulin action. Fluorescent light sheet microscopy analysis of whole brain was performed to assess activation of brain regions. RESULTS Co-administration of long-acting Fc-IgG/peptide conjugates of Fc-GLP-1 and Fc-PYY3-36 (specific for PYY receptor-2 (Y2R)) resulted in profound weight loss, restored glucose homeostasis, and recovered endogenous β-cell function in two mouse models of obese T2D. Hyperinsulinemic-euglycemic clamps in C57BLKS/J db/db and diet-induced obese Y2R-deficient (Y2RKO) mice indicated Y2R is required for a weight-independent improvement in peripheral insulin sensitivity and enhanced hepatic glycogenesis. Brain cFos staining demonstrated distinct temporal activation of regions of the hypothalamus and hindbrain following Fc-PYY3-36 + Fc-GLP-1R agonist administration. CONCLUSIONS These results reveal a therapeutic approach for obesity/T2D that improved insulin sensitivity and restored endogenous β-cell function. These data also highlight the potential association between the gut-brain axis in control of metabolic homeostasis.
Collapse
Affiliation(s)
- Brandon B Boland
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Gubra ApS, Horsholm, Denmark; PRECISIONscientia, Yardley, PA, USA
| | - Rhianna C Laker
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Siobhan O'Brien
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Sadichha Sitaula
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Isabelle Sermadiras
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | | | | | | | | | | | - Arthur Suckow
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; DTX Pharma, San Diego, CA, USA
| | - Nicole Burmeister
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Roche, Penzberg, Germany
| | - Stephanie Oldham
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Sarah Will
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Victor G Howard
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Benji M Gill
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Philip Newton
- Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Antibody and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jacqueline Naylor
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - David C Hornigold
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jotham Austin
- University of Chicago Advanced Electron Microscopy Core Facility, Chicago, IL, USA
| | - Louise Lantier
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Owen P McGuinness
- Vanderbilt University Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - James L Trevaskis
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK; Gilead Sciences, Foster City, CA, USA
| | - Joseph S Grimsby
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Christopher J Rhodes
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
241
|
Ahrends T, Aydin B, Matheis F, Classon CH, Marchildon F, Furtado GC, Lira SA, Mucida D. Enteric pathogens induce tissue tolerance and prevent neuronal loss from subsequent infections. Cell 2021; 184:5715-5727.e12. [PMID: 34717799 PMCID: PMC8595755 DOI: 10.1016/j.cell.2021.10.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/17/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023]
Abstract
The enteric nervous system (ENS) controls several intestinal functions including motility and nutrient handling, which can be disrupted by infection-induced neuropathies or neuronal cell death. We investigated possible tolerance mechanisms preventing neuronal loss and disruption in gut motility after pathogen exposure. We found that following enteric infections, muscularis macrophages (MMs) acquire a tissue-protective phenotype that prevents neuronal loss, dysmotility, and maintains energy balance during subsequent challenge with unrelated pathogens. Bacteria-induced neuroprotection relied on activation of gut-projecting sympathetic neurons and signaling via β2-adrenergic receptors (β2AR) on MMs. In contrast, helminth-mediated neuroprotection was dependent on T cells and systemic production of interleukin (IL)-4 and IL-13 by eosinophils, which induced arginase-expressing MMs that prevented neuronal loss from an unrelated infection located in a different intestinal region. Collectively, these data suggest that distinct enteric pathogens trigger a state of disease or tissue tolerance that preserves ENS number and functionality.
Collapse
Affiliation(s)
- Tomasz Ahrends
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA.
| | - Begüm Aydin
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - Fanny Matheis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - Cajsa H Classon
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Gláucia C Furtado
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sérgio A Lira
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
242
|
Correia JC, Kelahmetoglu Y, Jannig PR, Schweingruber C, Shvaikovskaya D, Zhengye L, Cervenka I, Khan N, Stec M, Oliveira M, Nijssen J, Martínez-Redondo V, Ducommun S, Azzolini M, Lanner JT, Kleiner S, Hedlund E, Ruas JL. Muscle-secreted neurturin couples myofiber oxidative metabolism and slow motor neuron identity. Cell Metab 2021; 33:2215-2230.e8. [PMID: 34592133 DOI: 10.1016/j.cmet.2021.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 05/28/2021] [Accepted: 09/07/2021] [Indexed: 01/04/2023]
Abstract
Endurance exercise promotes skeletal muscle vascularization, oxidative metabolism, fiber-type switching, and neuromuscular junction integrity. Importantly, the metabolic and contractile properties of the muscle fiber must be coupled to the identity of the innervating motor neuron (MN). Here, we show that muscle-derived neurturin (NRTN) acts on muscle fibers and MNs to couple their characteristics. Using a muscle-specific NRTN transgenic mouse (HSA-NRTN) and RNA sequencing of MN somas, we observed that retrograde NRTN signaling promotes a shift toward a slow MN identity. In muscle, NRTN increased capillary density and oxidative capacity and induced a transcriptional reprograming favoring fatty acid metabolism over glycolysis. This combination of effects on muscle and MNs makes HSA-NRTN mice lean with remarkable exercise performance and motor coordination. Interestingly, HSA-NRTN mice largely recapitulate the phenotype of mice with muscle-specific expression of its upstream regulator PGC-1ɑ1. This work identifies NRTN as a myokine that couples muscle oxidative capacity to slow MN identity.
Collapse
Affiliation(s)
- Jorge C Correia
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Yildiz Kelahmetoglu
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Paulo R Jannig
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Christoph Schweingruber
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Biochemistry and Biophysics, Stockholm University, 10691 Stockholm, Sweden
| | - Dasha Shvaikovskaya
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Liu Zhengye
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Igor Cervenka
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Naveen Khan
- Regeneron Pharmaceuticals, Tarrytown, NY 10 591, USA
| | - Michael Stec
- Regeneron Pharmaceuticals, Tarrytown, NY 10 591, USA
| | - Mariana Oliveira
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Jik Nijssen
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Vicente Martínez-Redondo
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Serge Ducommun
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Michele Azzolini
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | - Johanna T Lanner
- Molecular Muscle Physiology and Pathophysiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden
| | | | - Eva Hedlund
- Department of Neuroscience, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden; Department of Biochemistry and Biophysics, Stockholm University, 10691 Stockholm, Sweden
| | - Jorge L Ruas
- Molecular and Cellular Exercise Physiology, Department of Physiology and Pharmacology, Biomedicum, Karolinska Institutet, 17165 Stockholm, Sweden.
| |
Collapse
|
243
|
Lin J, Jiang X, Dong M, Liu X, Shen Q, Huang Y, Zhang H, Ye R, Zhou H, Yan C, Yuan S, Wu X, Chen L, Wang Y, He M, Tao Y, Zhang Z, Jin W. Hepatokine Pregnancy Zone Protein Governs the Diet-Induced Thermogenesis Through Activating Brown Adipose Tissue. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101991. [PMID: 34514733 PMCID: PMC8564441 DOI: 10.1002/advs.202101991] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/23/2021] [Indexed: 05/06/2023]
Abstract
Intermittent fasting (IF), as a dietary intervention for weight loss, takes effects primarily through increasing energy expenditure. However, whether inter-organ systems play a key role in IF remains unclear. Here, a novel hepatokine, pregnancy zone protein (PZP) is identified, which has significant induction during the refeeding stage of IF. Further, loss of function studies and protein therapeutic experiment in mice revealed that PZP promotes diet-induced thermogenesis through activating brown adipose tissue (BAT). Mechanistically, circulating PZP can bind to cell surface glucose-regulated protein of 78 kDa (GRP78) to promote uncoupling protein 1 (UCP1) expression via a p38 MAPK-ATF2 signaling pathway in BAT. These studies illuminate a systemic regulation in which the IF promotes BAT thermogenesis through the endocrinal system and provide a novel potential target for treating obesity and related disorders.
Collapse
Affiliation(s)
- Jun Lin
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Xiaoxiao Jiang
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Meng Dong
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Xiaomeng Liu
- Institute of Neuroscience and Translational MedicineCollege of Life Science and AgronomyZhoukou Normal UniversityZhoukou466000China
| | - Qiwei Shen
- Department of General SurgeryHuashan HospitalFudan UniversityShanghaiChina
| | - Yuanyuan Huang
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hanlin Zhang
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Rongcai Ye
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Huiqiao Zhou
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Chunlong Yan
- College of AgricultureYanbian UniversityYanji133000China
| | - Shouli Yuan
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Xiangnan Wu
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Li Chen
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yanfang Wang
- State Key Laboratory of Animal NutritionInstitute of Animal ScienceChinese Academy of Agricultural SciencesBeijing100193China
| | - Min He
- Division of Endocrinology and MetabolismHuashan HospitalFudan UniversityShanghaiChina
| | - Yi Tao
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
| | - Zhaoyun Zhang
- Division of Endocrinology and MetabolismHuashan HospitalFudan UniversityShanghaiChina
| | - Wanzhu Jin
- Key Laboratory of Animal Ecology and Conservation BiologyInstitute of ZoologyChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
244
|
Ortuno MJ, Schneeberger M, Ilanges A, Marchildon F, Pellegrino K, Friedman JM, Ducy P. Melanocortin 4 receptor stimulation prevents anti-depressant-associated weight gain in mice caused by long-term fluoxetine exposure. J Clin Invest 2021; 131:151976. [PMID: 34673574 DOI: 10.1172/jci151976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 10/19/2021] [Indexed: 11/17/2022] Open
Abstract
Contrasting with the predicted anorexigenic effect of increasing brain serotonin signaling, long-term use of selective serotonin reuptake inhibitors (SSRIs) antidepressants correlates with body weight gain. This adverse outcome increases the risk of transitioning to obesity and interferes with treatment compliance. Here we show that orally administered fluoxetine (Flx), a widely prescribed SSRI, increased body weight by enhancing food intake in healthy mice at two different time points and through two distinct mechanisms. Within hours, Flx decreased the activity of a subset of brainstem serotonergic neurons by triggering autoinhibitory signaling through the Htr1a receptor. Upon longer treatment Flx blunted Htr2c expression/signaling, decreased the phosphorylation of Creb and Stat3 and dampened the production of POMC/α-MSH in hypothalamic neurons, thereby increasing food intake. Accordingly, exogenous stimulation of the melanocortin 4 receptor (MC4R) by co-treating mice with Flx and lipocalin-2, an anorexigenic hormone signaling through this receptor, normalized feeding and body weight. Flx and other SSRIs also inhibit CREB/STAT3 phosphorylation in a human neuronal cell line suggesting that these non-canonical effects could also occur in long-term users of SSRIs. By defining the molecular basis of the long-term SSRIs-associated weight gain this study proposes a therapeutic strategy to counter it.
Collapse
Affiliation(s)
- Maria Jose Ortuno
- Department of Genetics and Development, Columbia University, New York, United States of America
| | - Marc Schneeberger
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - Anoj Ilanges
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, United States of America
| | - Kyle Pellegrino
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - Jeffrey M Friedman
- Laboratory of Molecular Genetics, The Rockefeller University, New York, United States of America
| | - Patricia Ducy
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, United States of America
| |
Collapse
|
245
|
Weiner J, Roth L, Kranz M, Brust P, Boelen A, Klöting N, Heiker JT, Blüher M, Tönjes A, Pfluger PT, Stumvoll M, Mittag J, Krause K. Leptin counteracts hypothermia in hypothyroidism through its pyrexic effects and by stabilizing serum thyroid hormone levels. Mol Metab 2021; 54:101348. [PMID: 34610354 PMCID: PMC8556519 DOI: 10.1016/j.molmet.2021.101348] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 11/30/2022] Open
Abstract
Objective Thyroid hormones (TH) are essential for the homeostatic control of energy metabolism and the regulation of body temperature. The hypothalamic–pituitary–thyroid (HPT) axis is regulated by negative feedback mechanisms, ensuring that TH levels are maintained at a constant level. However, the feedback mechanisms underlying the resetting of the HPT axis regulation in the control of body temperature are still not fully understood. Here, we aimed to determine the thermoregulatory response in hypothyroid mice to different environmental temperatures and the underlying mechanisms. Methods Distinct thermogenic challenges were induced in hypothyroid female C57BL/6N and leptin-deficient ob/ob mice through housing at either room temperature or thermoneutrality. The thermogenic and metabolic effects were analyzed through metabolic chambers, 18F-FDG-PET/MRI, infrared thermography, metabolic profiling, histology, gene expression and Western blot analysis. Results In hypothyroid mice maintained at room temperature, high leptin serum levels induce a pyrexic effect leading to the stabilization of body temperature through brown adipose tissue thermogenesis and white adipose tissue browning. Housing at thermoneutrality leads to the normalization of leptin levels and a reduction of the central temperature set point, resulting in decreased thermogenesis in brown and white adipose tissue and skeletal muscle and a significant decline in body temperature. Furthermore, anapyrexia in hypothyroid leptin-deficient ob/ob mice indicates that besides its pyrexic actions, leptin exerts a stimulatory effect on the HPT axis to stabilize the remaining TH serum levels in hypothyroid mice. Conclusion This study led to the identification of a previously unknown endocrine loop in which leptin acts in concert with the HPT axis to stabilize body temperature in hypothyroid mice. Thyroid hormones are essential for the regulation of body temperature. Thyroid hormone-deficient (hypothyroid) mice show distinct leptin serum concentrations in response to changes in ambient housing temperature. High leptin serum levels confer a stimulatory effect on the hypothalamic-pituitary-thyroid axis. High leptin serum level prevents fall in body temperature in hypothyroid mice at room temperature through its pyrexic effects.
Collapse
Affiliation(s)
- Juliane Weiner
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Lisa Roth
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Mathias Kranz
- University Hospital of North Norway, Tromsø, Norway; Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Anita Boelen
- Endocrine Laboratory, Department of Clinical Chemistry, Amsterdam Gastroenterology Endocrinology and Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Nora Klöting
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - John T Heiker
- Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Anke Tönjes
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Paul T Pfluger
- Helmholtz Zentrum München, Research Unit NeuroBiology of Diabetes, Neuherberg, Germany; Technical University of Munich (TUM), TUM School of Medicine, NeuroBiology of Diabetes, Munich, Germany
| | - Michael Stumvoll
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany; Helmholtz Zentrum München, Helmholtz Institute for Metabolic, Obesity and Vascular Research, Leipzig, Germany
| | - Jens Mittag
- Institute for Endocrinology & Diabetes/CBBM, University of Lübeck, Lübeck, Germany
| | - Kerstin Krause
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
246
|
Cartwright DM, Oakey LA, Fletcher RS, Doig CL, Heising S, Larner DP, Nasteska D, Berry CE, Heaselgrave SR, Ludwig C, Hodson DJ, Lavery GG, Garten A. Nicotinamide riboside has minimal impact on energy metabolism in mouse models of mild obesity. J Endocrinol 2021; 251:111-123. [PMID: 34370682 PMCID: PMC8494379 DOI: 10.1530/joe-21-0123] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
Supplementation with precursors of NAD has been shown to prevent and reverse insulin resistance, mitochondrial dysfunction, and liver damage in mouse models of diet-induced obesity. We asked whether the beneficial effects of supplementation with the NAD precursor nicotinamide riboside (NR) are dependent on mouse strain. We compared the effects of NR supplementation on whole-body energy metabolism and mitochondrial function in mildly obese C57BL/6N and C57BL/6J mice, two commonly used strains to investigate metabolism. Male C57BL/6N and C57BL/6J mice were fed a high-fat diet (HFD) or standard chow with or without NR supplementation for 8 weeks. Body and organ weights, glucose tolerance, and metabolic parameters as well as mitochondrial O2 flux in liver and muscle fibers were assessed. We found that NR supplementation had no influence on body or organ weight, glucose metabolism or hepatic lipid accumulation, energy expenditure, or metabolic flexibility but increased mitochondrial respiration in soleus muscle in both mouse strains. Strain-dependent differences were detected for body and fat depot weight, fasting blood glucose, hepatic lipid accumulation, and energy expenditure. We conclude that, in mild obesity, NR supplementation does not alter metabolic phenotype in two commonly used laboratory mouse strains.
Collapse
Affiliation(s)
- David M Cartwright
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lucy A Oakey
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rachel S Fletcher
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Craig L Doig
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Silke Heising
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Dean P Larner
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Daniela Nasteska
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Caitlin E Berry
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sam R Heaselgrave
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Christian Ludwig
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - David J Hodson
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gareth G Lavery
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Antje Garten
- Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Pediatric Research Center, Hospital for Child and Adolescent Medicine, Leipzig University, Leipzig, Germany
| |
Collapse
|
247
|
Agarwal N, Iyer D, Saha P, Cox AR, Xia Y, Utay NS, Somasundaram A, Schubert U, Lake JE, Hartig SM, Balasubramanyam A. HIV-1 Viral Protein R Couples Metabolic Inflexibility With White Adipose Tissue Thermogenesis. Diabetes 2021; 70:2014-2025. [PMID: 34233931 PMCID: PMC8576429 DOI: 10.2337/db20-0888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 06/29/2021] [Indexed: 11/13/2022]
Abstract
Persons living with HIV (PLWH) manifest chronic disorders of brown and white adipose tissues that lead to diabetes and metabolic syndrome. The mechanisms that link viral factors to defective adipose tissue function and abnormal energy balance in PLWH remain incompletely understood. Here, we explored how the HIV accessory protein viral protein R (Vpr) contributes to adaptive thermogenesis in two mouse models and human adipose tissues. Uncoupling protein 1 (UCP1) gene expression was strongly increased in subcutaneous white adipose tissue (WAT) biopsy specimens from PLWH and in subcutaneous WAT of the Vpr mice, with nearly equivalent mRNA copy number. Histology and functional studies confirmed beige transformation in subcutaneous but not visceral WAT in the Vpr mice. Measurements of energy balance indicated Vpr mice displayed metabolic inflexibility and could not shift efficiently from carbohydrate to fat metabolism during day-night cycles. Furthermore, Vpr mice showed a marked inability to defend body temperature when exposed to 4°C. Importantly, Vpr couples higher tissue catecholamine levels with UCP1 expression independent of β-adrenergic receptors. Our data reveal surprising deficits of adaptive thermogenesis that drive metabolic inefficiency in HIV-1 Vpr mouse models, providing an expanded role for viral factors in the pathogenesis of metabolic disorders in PLWH.
Collapse
Affiliation(s)
- Neeti Agarwal
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX
| | - Dinakar Iyer
- Department of Biology and Biochemistry, University of Houston, Houston, TX
| | - Pradip Saha
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX
| | - Aaron R Cox
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX
| | - Yan Xia
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Netanya S Utay
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX
| | - Anoma Somasundaram
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX
| | | | - Jordan E Lake
- Division of Infectious Diseases, Department of Internal Medicine, McGovern Medical School at the University of Texas Health Science Center, Houston, TX
| | - Sean M Hartig
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX
| | - Ashok Balasubramanyam
- Division of Diabetes, Endocrinology, and Metabolism, Baylor College of Medicine, Houston, TX
| |
Collapse
|
248
|
Müller TD, Klingenspor M, Tschöp MH. Revisiting energy expenditure: how to correct mouse metabolic rate for body mass. Nat Metab 2021; 3:1134-1136. [PMID: 34489606 DOI: 10.1038/s42255-021-00451-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research, Neuherberg, Germany.
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Matthias H Tschöp
- German Center for Diabetes Research, Neuherberg, Germany
- Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität, Munich, Germany
| |
Collapse
|
249
|
Oteng AB, Higuchi S, Banks AS, Haeusler RA. Cyp2c-deficiency depletes muricholic acids and protects against high-fat diet-induced obesity in male mice but promotes liver damage. Mol Metab 2021; 53:101326. [PMID: 34438105 PMCID: PMC8449133 DOI: 10.1016/j.molmet.2021.101326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/08/2021] [Accepted: 08/18/2021] [Indexed: 01/07/2023] Open
Abstract
Objective Murine-specific muricholic acids (MCAs) are reported to protect against obesity and associated metabolic disorders. However, the response of mice with genetic depletion of MCA to an obesogenic diet has not been evaluated. We used Cyp2c-deficient (Cyp2c−/−) mice, which lack MCAs and thus have a human-like bile acid (BA) profile, to directly investigate the potential role of MCAs in diet-induced obesity. Methods Male and female Cyp2c−/− mice and wild-type (WT) littermate controls were fed a standard chow diet or a high-fat diet (HFD) for 18 weeks. We measured BA composition from a pool of liver, gallbladder, and intestine, as well as weekly body weight, food intake, lean and fat mass, systemic glucose homeostasis, energy expenditure, intestinal lipid absorption, fecal lipid, and energy content. Results Cyp2c-deficiency depleted MCAs and caused other changes in BA composition, namely a decrease in the ratio of 12α-hydroxylated (12α-OH) BAs to non-12α-OH BAs, without altering the total BA levels. While WT male mice became obese after HFD feeding, Cyp2c−/− male mice were protected from obesity and associated metabolic dysfunctions. Cyp2c−/− male mice also showed reduced intestinal lipid absorption and increased lipid excretion, which was reversed by oral gavage with the 12α-OH BA and taurocholic acid (TCA). Cyp2c−/− mice also showed increased liver damage, which appeared stronger in females. Conclusions MCA does not protect against diet-induced obesity but may protect against liver injury. Reduced lipid absorption in Cyp2c-deficient male mice is potentially due to a reduced ratio of 12α-OH/non-12α-OH BAs. Presence of MCA does not necessarily protect against diet-induced obesity. Cyp2c deficiency promotes resistance to diet-induced obesity in males. Cyp2c-knockout mice have decreased the ratio of 12α-OH/non-12α-OH BAs that promotes decreased intestinal lipid absorption. Cyp2c-knockout mice have improved glucose homeostasis. Cyp2c-deficiency promotes mild and severe liver injury in male and female mice, respectively.
Collapse
Affiliation(s)
- Antwi-Boasiako Oteng
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Sei Higuchi
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Alexander S Banks
- Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA; Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
250
|
Al Rijjal D, Liu Y, Lai M, Song Y, Danaei Z, Wu A, Mohan H, Wei L, Schopfer FJ, Dai FF, Wheeler MB. Vascepa protects against high-fat diet-induced glucose intolerance, insulin resistance, and impaired β-cell function. iScience 2021; 24:102909. [PMID: 34458694 PMCID: PMC8379293 DOI: 10.1016/j.isci.2021.102909] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/28/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Omega-3 fatty acid prescription drugs, Vascepa (≥96% eicosapentaenoic acid [EPA] ethyl ester) and Lovaza (46.5% EPA and 37.5% docosahexaenoic acid ethyl ester) are known therapeutic regimens to treat hypertriglyceridemia. However, their impact on glucose homeostasis, progression to type 2 diabetes, and pancreatic beta cell function are not well understood. In the present study, mice were treated with Vascepa or Lovaza for one week prior to six weeks of high-fat diet feeding. Vascepa but not Lovaza led to reduced insulin resistance, reduced fasting insulin and glucose, and improved glucose intolerance. Vascepa improved beta cell function, reduced liver triglycerides with enhanced expression of hepatic fatty acid oxidation genes, and altered microbiota composition. Vascepa has protective effects on diet-induced insulin resistance and glucose intolerance in mice.
Collapse
Affiliation(s)
- Dana Al Rijjal
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Ying Liu
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
- Division of Advanced Diagnostics, Metabolism, Toronto General Research Institute, ON, Canada
| | - Mi Lai
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
- Division of Advanced Diagnostics, Metabolism, Toronto General Research Institute, ON, Canada
| | - Youchen Song
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Zahra Danaei
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Anne Wu
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Haneesha Mohan
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Li Wei
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Francisco J. Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA; Pittsburgh Liver Research Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Feihan F. Dai
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
| | - Michael B. Wheeler
- Department of Physiology, University of Toronto, 1 King's College Circle, Medical Science Building Rm#3352, Toronto, ON, M5S 1A8, Canada
- Division of Advanced Diagnostics, Metabolism, Toronto General Research Institute, ON, Canada
| |
Collapse
|