1
|
Chen X, Mei XY, Ren ZM, Chen SS, Tong YL, Zhang CP, Chen J, Dai GH. Comprehensive insights into berberine's hypoglycemic mechanisms: A focus on ileocecal microbiome in db/db mice. Heliyon 2024; 10:e33704. [PMID: 39040376 PMCID: PMC11261017 DOI: 10.1016/j.heliyon.2024.e33704] [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: 12/13/2023] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/24/2024] Open
Abstract
The efficacy of berberine in managing diabetes through modulation of gut microbiome has been established through fecal sample analyses. However, relying solely on fecal materials constrains our comprehension of berberine's effects on diverse gastrointestinal locations. This study specifically explores the ileocecal region, a segment characterized by higher microbial diversity than fecal samples. Berberine exhibits a robust hypoglycemic impact by significantly reducing glucose levels in blood and urine. Beyond glycemic control, berberine ameliorates various diabetes-related symptoms in serum, including increased insulin and leptin, but decreased NEFA and MDA. Notably, berberine demonstrates liver-protective functions by alleviating oxidative stress and enhancing hepatic glycogen abundance. These outcomes prompted a high-throughput sequencing analysis of the ileocecal microbiome, revealing an augmentation of beneficial bacterial genera (four genera in the Lachnospiraceae family, Erysipelatoclostridium, and Escherichia-Shigella), along with a reduction in harmful bacterial genera (Romboutsia). Additionally, we predicted the impact of the ileocecal microbiome on clinically relevant factors associated with diabetes. These findings elucidate the multi-pathway mechanisms of berberine in treating T2D, underscoring its potential as a natural anti-diabetic agent or functional food, particularly through the modulation of the gut microbiota.
Collapse
Affiliation(s)
- Xuan Chen
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Xi-yu Mei
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Ze-ming Ren
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Si-si Chen
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Ye-ling Tong
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | | | - Jia Chen
- Hangzhou Linping Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Guan-hai Dai
- Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
2
|
Ishida E, Horiguchi K, Matsumoto S, Ozawa A, Sekiguchi S, Yamada E. Influence of diet and body weight in treatment-resistant acquired partial lipodystrophy after hematopoietic stem cell transplantation and its potential for metabolic improvement. Diabetol Int 2024; 15:290-296. [PMID: 38524924 PMCID: PMC10959909 DOI: 10.1007/s13340-023-00674-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/06/2023] [Indexed: 03/26/2024]
Abstract
Lipodystrophy is a rare disease characterized by various metabolic complications resulting from the complete or partial loss of adipose tissues and abnormal fat accumulation. Acquired lipodystrophy may occur due to certain drugs, autoimmunity or for unknown reasons. Recently, cases of acquired lipodystrophy after hematopoietic stem cell transplantation (HSCT) have been reported. Leptin administration, used recently to treat generalized lipodystrophy, effectively controlled metabolic complications; however, few reports demonstrated the effectiveness of leptin for acquired partial lipodystrophy. In this report, we present the case of a 17-year-old woman who developed insulin resistance, hypertriglyceridemia, and fatty liver after HSCT. Due to her thin gluteal fat and low blood adiponectin levels, her metabolic abnormalities were attributed to partial lipodystrophy. While both leptin and pemafibrate administration partially attenuated metabolic abnormalities, its effects were relatively limited, probably because the serum leptin levels were maintained, which is not likely in generalized lipodystrophy. Nevertheless, after she developed adjustment disorder and experienced weight loss, along with decreased food intake, her metabolic markers significantly improved. This case suggests the modest effect of leptin and permafibrate in partial lipodystrophy after HSCT, highlighting the importance of diet therapy in metreleptin treatment for acquired partial lipodystrophy.
Collapse
Affiliation(s)
- Emi Ishida
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| | - Kazuhiko Horiguchi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| | - Shunichi Matsumoto
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| | - Atsushi Ozawa
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| | - Sho Sekiguchi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| | - Eijiro Yamada
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Gunma University Graduate School of Medicine, 3-39-15 Showa-Machi, Maebashi, Gunma 371-8511 Japan
| |
Collapse
|
3
|
Chen X, Tong YL, Ren ZM, Chen SS, Mei XY, Zhou QY, Dai GH. Hypoglycemic mechanisms of Polygonatum sibiricum polysaccharide in db/db mice via regulation of glycolysis/gluconeogenesis pathway and alteration of gut microbiota. Heliyon 2023; 9:e15484. [PMID: 37128343 PMCID: PMC10147986 DOI: 10.1016/j.heliyon.2023.e15484] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/02/2023] [Accepted: 04/12/2023] [Indexed: 05/03/2023] Open
Abstract
Polygonatum rhizoma polysaccharide (PP) is a main ingredient of Polygonatum rhizoma , which is both food and traditional herbal medicine. In this study, we aimed to investigate the hypoglycemic effect of PP and the underlying mechanisms in db/db mice. Our finding showed that PP significantly ameliorates diabetic symptoms by reducing glucose levels in blood and urine and increasing insulin and leptin abundance in the serum. Histopathological examination revealed that PP improved the pathological state and increased hepatic glycogen storage in liver. Additionally, RT-qPCR results indicated that PP significantly down-regulated the expression of phosphoenolpyruvate carboxykinase 1. Furthermore, 16s rRNA sequencing results demonstrated that PP intervention resulted in an increase in beneficial bacteria genus and a reduction in harmful genus. Redundancy analysis revealed the correlation between intestinal flora and clinical factors. Taken together, these results suggest that PP has a significant hypoglycemic effect on type 2 diabetes (T2D) through up-regulating serum insulin and leptin, as well as hepatic glycogen storage, and down-regulating hepatic phosphoenolpyruvate carboxykinase 1 expression, as well as modulating gut microbiota composition. In conclusion, this study investigated the mechanisms of PP in the treatment of diabetes in db/db mice. To the best of our knowledge, this is the first study to explore the positive and negative correlations between gut microbiota and clinical factors, such as oxidative stress injury in liver and glucose related indicators in the blood.
Collapse
Affiliation(s)
- Xuan Chen
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Ye-ling Tong
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Ze-ming Ren
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Si-si Chen
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Xi-yu Mei
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
| | - Qing-yun Zhou
- Second Clinical Medical School, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, People's Republic of China
| | - Guan-hai Dai
- Institute of Basic Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, People's Republic of China
- Corresponding author.
| |
Collapse
|
4
|
Genetic Markers of Insulin Resistance and Atherosclerosis in Type 2 Diabetes Mellitus Patients with Coronary Artery Disease. Metabolites 2023; 13:metabo13030427. [PMID: 36984867 PMCID: PMC10054456 DOI: 10.3390/metabo13030427] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) is characterized by impaired insulin secretion on a background of insulin resistance (IR). IR and T2DM are associated with atherosclerotic coronary artery disease (CAD). The mechanisms of IR and atherosclerosis are known to share similar genetic and environmental roots. Endothelial dysfunction (ED) detected at the earliest stages of IR might be the origin of atherosclerosis progression. ED influences the secretion of pro-inflammatory cytokines and their encoding genes. The genes and their single nucleotide polymorphisms (SNPs) act as potential genetic markers of IR and atherosclerosis. This review focuses on the link between IR, T2DM, atherosclerosis, CAD, and the potential genetic markers CHI3L1, CD36, LEPR, RETN, IL-18, RBP-4, and RARRES2 genes.
Collapse
|
5
|
Alzamil H, Aldokhi L. Triglycerides and leptin soluble receptor: Which one is the target to protect β-cells in patients with type 2 diabetes? Front Endocrinol (Lausanne) 2023; 14:1077678. [PMID: 36950695 PMCID: PMC10027012 DOI: 10.3389/fendo.2023.1077678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
OBJECTIVES to study the relationships of leptin and leptin SR with adiposity indices, and glycemic indices in patients with type 2 diabetes mellitus (T2DM) compared to healthy subjects. METHODS This cross-sectional study involved 65 patients with T2DM and 63 healthy controls. Fasting plasma levels of leptin, leptin SR, insulin and lipid profile were measured by enzyme linked immunosorbent essay, basal insulin resistance and beta-cell function were assessed using the homeostasis model assessment. RESULTS leptin SR level was significantly higher in T2DM patients than in controls (5.8 ± 1.6 and 4.8 ± 1.3 respectively; p= 0.001). In patients with T2DM, leptin SR was negatively correlated with homeostasis model of β-cell function and body fat mass while it has a significant positive correlation with glycosylated hemoglobin (HbA1c). The independent predictors for leptin SR in patients with T2DM were triglycerides (TG) and HbA1c. CONCLUSIONS elevated serum leptin SR level in patients with T2DM was positively correlated with TG and abnormal glucose metabolism which indicate that it plays a role in pathophysiology of T2DM. The association of elevated leptin SR level with high TG and deterioration of β-cell function indicate that in some individuals, particularly non-obese, dyslipidemia might be a cause rather than a complication of diabetes.
Collapse
|
6
|
Liu F, Cai Z, Yang Y, Plasko G, Zhao P, Wu X, Tang C, Li D, Li T, Hu S, Song L, Yu S, Xu R, Luo H, Fan L, Wang E, Xiao Z, Ji Y, Zeng R, Li R, Bai J, Zhou Z, Liu F, Zhang J. The adipocyte-enriched secretory protein tetranectin exacerbates type 2 diabetes by inhibiting insulin secretion from β cells. SCIENCE ADVANCES 2022; 8:eabq1799. [PMID: 36129988 PMCID: PMC9491725 DOI: 10.1126/sciadv.abq1799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/04/2022] [Indexed: 06/15/2023]
Abstract
Pancreatic β cell failure is a hallmark of diabetes. However, the causes of β cell failure remain incomplete. Here, we report the identification of tetranectin (TN), an adipose tissue-enriched secretory molecule, as a negative regulator of insulin secretion in β cells in diabetes. TN expression is stimulated by high glucose in adipocytes via the p38 MAPK/TXNIP/thioredoxin/OCT4 signaling pathway, and elevated serum TN levels are associated with diabetes. TN treatment greatly exacerbates hyperglycemia in mice and suppresses glucose-stimulated insulin secretion in islets. Conversely, knockout of TN or neutralization of TN function notably improves insulin secretion and glucose tolerance in high-fat diet-fed mice. Mechanistically, TN binds with high selectivity to β cells and inhibits insulin secretion by blocking L-type Ca2+ channels. Our study uncovers an adipocyte-β cell cross-talk that contributes to β cell dysfunction in diabetes and suggests that neutralization of TN levels may provide a new treatment strategy for type 2 diabetes.
Collapse
Affiliation(s)
- Fen Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zixin Cai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Yan Yang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - George Plasko
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX 78229, USA
| | - Piao Zhao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Xiangyue Wu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Cheng Tang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Dandan Li
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ting Li
- Department of Liver Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shanbiao Hu
- Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Lei Song
- Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Shaojie Yu
- Department of Urological Organ Transplantation, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ran Xu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Hairong Luo
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Libin Fan
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Ersong Wang
- Department of Neurosurgery, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Zhen Xiao
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, China
| | - Yujiao Ji
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Rong Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rongxia Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Juli Bai
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
- Department of Pharmacology, University of Texas Health at San Antonio, San Antonio, TX 78229, USA
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Feng Liu
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Jingjing Zhang
- National Clinical Research Center for Metabolic Diseases, Metabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| |
Collapse
|
7
|
Huang Q, Hao S, You J, Yao X, Li Z, Schilling J, Thyparambil S, Liao WL, Zhou X, Mo L, Ladella S, Davies-Balch SR, Zhao H, Fan D, Whitin JC, Cohen HJ, McElhinney DB, Wong RJ, Shaw GM, Stevenson DK, Sylvester KG, Ling XB. Early-pregnancy prediction of risk for pre-eclampsia using maternal blood leptin/ceramide ratio: discovery and confirmation. BMJ Open 2021; 11:e050963. [PMID: 34824115 PMCID: PMC8627403 DOI: 10.1136/bmjopen-2021-050963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE This study aimed to develop a blood test for the prediction of pre-eclampsia (PE) early in gestation. We hypothesised that the longitudinal measurements of circulating adipokines and sphingolipids in maternal serum over the course of pregnancy could identify novel prognostic biomarkers that are predictive of impending event of PE early in gestation. STUDY DESIGN Retrospective discovery and longitudinal confirmation. SETTING Maternity units from two US hospitals. PARTICIPANTS Six previously published studies of placental tissue (78 PE and 95 non-PE) were compiled for genomic discovery, maternal sera from 15 women (7 non-PE and 8 PE) enrolled at ProMedDx were used for sphingolipidomic discovery, and maternal sera from 40 women (20 non-PE and 20 PE) enrolled at Stanford University were used for longitudinal observation. OUTCOME MEASURES Biomarker candidates from discovery were longitudinally confirmed and compared in parallel to the ratio of placental growth factor (PlGF) and soluble fms-like tyrosine kinase (sFlt-1) using the same cohort. The datasets were generated by enzyme-linked immunosorbent and liquid chromatography-tandem mass spectrometric assays. RESULTS Our discovery integrating genomic and sphingolipidomic analysis identified leptin (Lep) and ceramide (Cer) (d18:1/25:0) as novel biomarkers for early gestational assessment of PE. Our longitudinal observation revealed a marked elevation of Lep/Cer (d18:1/25:0) ratio in maternal serum at a median of 23 weeks' gestation among women with impending PE as compared with women with uncomplicated pregnancy. The Lep/Cer (d18:1/25:0) ratio significantly outperformed the established sFlt-1/PlGF ratio in predicting impending event of PE with superior sensitivity (85% vs 20%) and area under curve (0.92 vs 0.52) from 5 to 25 weeks of gestation. CONCLUSIONS Our study demonstrated the longitudinal measurement of maternal Lep/Cer (d18:1/25:0) ratio allows the non-invasive assessment of PE to identify pregnancy at high risk in early gestation, outperforming the established sFlt-1/PlGF ratio test.
Collapse
Affiliation(s)
| | - Shiying Hao
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Jin You
- Department of Bioengineering, University of California Riverside, Riverside, California, USA
| | | | - Zhen Li
- Department of Surgery, Stanford University, Stanford, California, USA
- Binhai Industrial Technology Research Institute, Zhejiang University, Tianjin, China
- School of Electrical Engineering, Southeast University, Nanjing, China
| | | | | | | | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, China
| | - Lihong Mo
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | - Subhashini Ladella
- Department of Obstetrics and Gynecology, University of California San Francisco, Fresno, California, USA
| | | | - Hangyi Zhao
- Department of Mathematics, Stanford University, Stanford, California, USA
| | - David Fan
- Department of Statistics and Applied Probability, University of California Santa Barbara, Santa Barbara, California, USA
| | - John C Whitin
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Harvey J Cohen
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Doff B McElhinney
- Department of Cardiothoracic Surgery, Stanford University, Stanford, California, USA
- Clinical and Translational Research Program, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Palo Alto, California, USA
| | - Ronald J Wong
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Gary M Shaw
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - David K Stevenson
- Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Karl G Sylvester
- Department of Surgery, Stanford University, Stanford, California, USA
| | - Xuefeng B Ling
- Department of Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
8
|
Abstract
Leptin is a pluripotent peptide hormone produced mainly by adipocytes, as well as by other tissues such as the stomach. Leptin primarily acts on the central nervous system, particularly the hypothalamus, where this hormone regulates energy homeostasis and neuroendocrine function. Owing to this, disruption of leptin signaling has been linked with numerous pathological conditions. Recent studies have also highlighted the diverse roles of leptin in the digestive system including immune regulation, cell proliferation, tissue healing, and glucose metabolism. Of note, leptin acts differently under physiological and pathological conditions. Here, we review the current knowledge on the functions of leptin and its downstream signaling in the gastrointestinal tract and accessory digestive organs, with an emphasis on its physiological and pathological implications. We also discuss the current therapeutic uses of recombinant leptin, as well as its limitations.
Collapse
Affiliation(s)
- Min-Hyun Kim
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Hyeyoung Kim
- Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, Korea
| |
Collapse
|
9
|
Zammouri J, Vatier C, Capel E, Auclair M, Storey-London C, Bismuth E, Mosbah H, Donadille B, Janmaat S, Fève B, Jéru I, Vigouroux C. Molecular and Cellular Bases of Lipodystrophy Syndromes. Front Endocrinol (Lausanne) 2021; 12:803189. [PMID: 35046902 PMCID: PMC8763341 DOI: 10.3389/fendo.2021.803189] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/09/2021] [Indexed: 12/14/2022] Open
Abstract
Lipodystrophy syndromes are rare diseases originating from a generalized or partial loss of adipose tissue. Adipose tissue dysfunction results from heterogeneous genetic or acquired causes, but leads to similar metabolic complications with insulin resistance, diabetes, hypertriglyceridemia, nonalcoholic fatty liver disease, dysfunctions of the gonadotropic axis and endocrine defects of adipose tissue with leptin and adiponectin deficiency. Diagnosis, based on clinical and metabolic investigations, and on genetic analyses, is of major importance to adapt medical care and genetic counseling. Molecular and cellular bases of these syndromes involve, among others, altered adipocyte differentiation, structure and/or regulation of the adipocyte lipid droplet, and/or premature cellular senescence. Lipodystrophy syndromes frequently present as systemic diseases with multi-tissue involvement. After an update on the main molecular bases and clinical forms of lipodystrophy, we will focus on topics that have recently emerged in the field. We will discuss the links between lipodystrophy and premature ageing and/or immuno-inflammatory aggressions of adipose tissue, as well as the relationships between lipomatosis and lipodystrophy. Finally, the indications of substitutive therapy with metreleptin, an analog of leptin, which is approved in Europe and USA, will be discussed.
Collapse
Affiliation(s)
- Jamila Zammouri
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
| | - Camille Vatier
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
- Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, National Reference Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
| | - Emilie Capel
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
| | - Martine Auclair
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
| | - Caroline Storey-London
- Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Pediatric Endocrinology Department, National Competence Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
| | - Elise Bismuth
- Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Pediatric Endocrinology Department, National Competence Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
| | - Héléna Mosbah
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
- Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, National Reference Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
| | - Bruno Donadille
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
- Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, National Reference Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
| | - Sonja Janmaat
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
- Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, National Reference Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
| | - Bruno Fève
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
- Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, National Reference Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
| | - Isabelle Jéru
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
- Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, National Reference Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
- Genetics Department, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Paris, France
| | - Corinne Vigouroux
- Sorbonne University, Inserm UMR_S 938, Saint-Antoine Research Centre, Cardiometabolism and Nutrition University Hospital Institute (ICAN), Paris, France
- Endocrinology Department, Assistance Publique-Hôpitaux de Paris, Saint-Antoine Hospital, National Reference Centre for Rare Diseases of Insulin Secretion and Insulin Sensitivity (PRISIS), Paris, France
- Genetics Department, Assistance Publique-Hôpitaux de Paris, La Pitié-Salpêtrière Hospital, Paris, France
| |
Collapse
|
10
|
Santos JL, Cortés VA. Eating behaviour in contrasting adiposity phenotypes: Monogenic obesity and congenital generalized lipodystrophy. Obes Rev 2021; 22:e13114. [PMID: 33030294 DOI: 10.1111/obr.13114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
Most known types of nonsyndromic monogenic obesity are caused by rare mutations in genes of the leptin-melanocortin pathway controlling appetite and adiposity. In contrast, congenital generalized lipodystrophy represents the most extreme form of leanness in humans caused by recessive mutations in four genes involved in phospholipid/triglyceride synthesis and lipid droplet/caveolae structure. In this disease, the inability to store triglyceride in adipocytes results in hypoleptinemia and ectopic hepatic and muscle fat accumulation leading to fatty liver, hypertriglyceridemia and severe insulin resistance. As a result of hypoleptinemia, patients with lipodystrophy show alterations in eating behaviour characterized by constant increased energy intake. As it occurs in obesity caused by genetic leptin deficiency, exogenous leptin rapidly reduces hunger scores in patients with congenital generalized lipodystrophy, with additional beneficial effects on glucose homeostasis and metabolic profile normalization. The melanocortin-4 receptor agonist setmelanotide has been used in the treatment of monogenic obesities. There is only one report on the effect of setmelanotide in a patient with partial lipodystrophy resulting in mild reductions in hunger scores, with no improvements in metabolic status. The assessment of contrasting phenotypes of obesity/leanness represents an adequate strategy to understand the pathophysiology and altered eating behaviour associated with adipose tissue excessive accumulation/paucity.
Collapse
Affiliation(s)
- José L Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Víctor A Cortés
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
11
|
|
12
|
|
13
|
Yoshino S, Iwasaki Y, Matsumoto S, Satoh T, Ozawa A, Yamada E, Kakizaki S, Trejo JAO, Uchiyama Y, Yamada M, Mori M. Administration of small-molecule guanabenz acetate attenuates fatty liver and hyperglycemia associated with obesity. Sci Rep 2020; 10:13671. [PMID: 32792584 PMCID: PMC7426972 DOI: 10.1038/s41598-020-70689-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by excessive accumulation of hepatic triglycerides (TG) and hyperglycemia arising due to persistent insulin resistance, and is profoundly linked to obesity. However, there is currently no established treatment for NAFLD in obese human subjects. We previously isolated Helz2, the expression of which was upregulated in human and mouse NAFLD, and its deletion activated the hepatic expression of functional leptin receptor long form (Leprb) and suppressed NAFLD development and body weight (BW) gain in obese mice. A high-throughput assay of small-molecule drugs revealed that guanabenz acetate (Ga), originally used to treat hypertension, possesses a high affinity constant against HELZ2, and its administration activates LEPRB expression in HepG2 cells in vitro. The chronic oral administration of Ga shows the selective leptin sensitization in the liver via upregulation of hepatic Leprb expression, which affects expression of genes involved in lipogenesis and fatty acid β-oxidation and diminishes hepatocyte hypertrophy with droplets enriched in TG in high-fat diet-induced obese mice. This activity significantly improves insulin resistance to decrease hyperglycemia and hepatocyte and adipocyte weights, resulting in BW reduction without reducing food intake. Regarding drug repositioning, Ga has the potential to effectively treat NAFLD and hyperglycemia in obese patients.
Collapse
Affiliation(s)
- Satoshi Yoshino
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Yusaku Iwasaki
- Laboratory of Animal Science, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, 606-8522, Japan
| | - Shunichi Matsumoto
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Tetsurou Satoh
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Atsushi Ozawa
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Eijiro Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Satoru Kakizaki
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Juan Alejandro Oliva Trejo
- Department of Cellular and Molecular Neuropathology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Yasuo Uchiyama
- Department of Cellular and Molecular Neuropathology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Masanobu Yamada
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Masatomo Mori
- Department of Medicine and Molecular Science, Gunma University Graduate School of Medicine, Maebashi, 371-8511, Japan.
- Metabolic and Obese Research Institute, Maebashi, 371-0048, Japan.
| |
Collapse
|
14
|
Lee H, Fenske RJ, Akcan T, Domask E, Davis DB, Kimple ME, Engin F. Differential Expression of Ormdl Genes in the Islets of Mice and Humans with Obesity. iScience 2020; 23:101324. [PMID: 32659722 PMCID: PMC7358727 DOI: 10.1016/j.isci.2020.101324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 05/05/2020] [Accepted: 06/25/2020] [Indexed: 12/28/2022] Open
Abstract
The orosomucoid-like (Ormdl) proteins play a critical role in sphingolipid homeostasis, inflammation, and ER stress, all of which are associated with obesity and βcell dysfunction. However, their roles in β cells and obesity remain unknown. Here, we show that islets from overweight/obese human donors displayed marginally reduced ORMDL1-2 expression, whereas ORMDL3 expression was significantly downregulated compared with islets from lean donors. In contrast, Ormdl3 was substantially upregulated in the islets of leptin-deficient obese (ob/ob) mice compared with lean mice. Treatment of ob/ob mice and their islets with leptin markedly reduced islet Ormld3 expression. Ormdl3 knockdown in a β cell line induced expression of pro-apoptotic markers, which was rescued by ceramide synthase inhibitor fumonisin B1. Our results reveal differential expression of Ormdl3 in the islets of a mouse model and humans with obesity, highlight the potential effect of leptin in this differential regulation, and suggest a role for Ormdl3 in β cell apoptosis. Islets of overweight/obese human donors display markedly reduced ORMDL3 expression Ormdl3 expression was significantly upregulated in the islets of ob/ob mice Leptin treatment markedly reduced Ormld3 expression in the islets of ob/ob mice Fumonisin B1 restores increased apoptotic marker levels induced by Ormdl3 silencing
Collapse
Affiliation(s)
- Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53706, USA
| | - Rachel J Fenske
- Interdepartmental Graduate Program in Nutritional Sciences, Madison, WI 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Tugce Akcan
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53706, USA
| | - Elliot Domask
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53706, USA
| | - Dawn B Davis
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Michelle E Kimple
- Interdepartmental Graduate Program in Nutritional Sciences, Madison, WI 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53705, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA; Department of Cell and Regenerative Biology, Madison, WI 53705, USA; Department of Academic Affairs, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, WI 53705, USA.
| |
Collapse
|
15
|
Gilloteaux J, Subramanian K, Solomon N, Nicaise C. The leptin receptor mutation of the obese Zucker rat causes sciatic nerve demyelination with a centripetal pattern defect. Ultrastruct Pathol 2018; 42:377-408. [PMID: 30339059 DOI: 10.1080/01913123.2018.1522405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Young male Zucker rats with a leptin receptor mutation are obese, have a non-insulin-dependent diabetes mellitus (NIDDM), and other endocrinopathies. Tibial branches of the sciatic nerve reveal a progressive demyelination that progresses out of the Schwann cells (SCs) where electron-contrast deposits are accumulated while the minor lines or intermembranous SC contacts display exaggerated spacings. Cajal bands contain diversely contrasted vesicles adjacent to the abaxonal myelin layer with blemishes; they appear dispatched centripetally out of many narrow electron densities, regularly spaced around the myelin annulus. These anomalies widen and yield into sectors across the stacked myelin layers. Throughout the worse degradations, the adaxonal membrane remains along the axonal neuroplasm. This peripheral neuropathy with irresponsive leptin cannot modulate hypothalamic-pituitary-adrenal axis and SC neurosteroids, thus exacerbates NIDDM condition. Additionally, the ultrastructure of the progressive myelin alterations may have unraveled a peculiar, centripetal mode of trafficking maintenance of the peripheral nervous system myelin, while some adhesive glycoproteins remain between myelin layers, somewhat hindering the axon mutilation. Heading title: Peripheral neuropathy and myelin.
Collapse
Affiliation(s)
- Jacques Gilloteaux
- a Department of Anatomical Sciences , St George's University School of Medicine, K.B. Taylor Global Scholar's Program at Northumbria University , Newcastle upon Tyne , UK.,b Unité de Recherche en Physiologie Moléculaire (URPhyM), Laboratoire de Neurodégénérescence et Régénération, Département de Médecine , Université de Namur , Namur , Belgium
| | - Kritika Subramanian
- a Department of Anatomical Sciences , St George's University School of Medicine, K.B. Taylor Global Scholar's Program at Northumbria University , Newcastle upon Tyne , UK.,c Department of Clinical and Epidemiological Virology , Rega Institute of Medical Research, Katholiele Universiteit Leuven , Leuven , Belgium
| | - Nadia Solomon
- a Department of Anatomical Sciences , St George's University School of Medicine, K.B. Taylor Global Scholar's Program at Northumbria University , Newcastle upon Tyne , UK
| | - Charles Nicaise
- b Unité de Recherche en Physiologie Moléculaire (URPhyM), Laboratoire de Neurodégénérescence et Régénération, Département de Médecine , Université de Namur , Namur , Belgium
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW We review the underlying mechanisms and potential benefits of intermittent fasting (IF) from animal models and recent clinical trials. RECENT FINDINGS Numerous variations of IF exist, and study protocols vary greatly in their interpretations of this weight loss trend. Most human IF studies result in minimal weight loss and marginal improvements in metabolic biomarkers, though outcomes vary. Some animal models have found that IF reduces oxidative stress, improves cognition, and delays aging. Additionally, IF has anti-inflammatory effects, promotes autophagy, and benefits the gut microbiome. The benefit-to-harm ratio varies by model, IF protocol, age at initiation, and duration. We provide an integrated perspective on potential benefits of IF as well as key areas for future investigation. In clinical trials, caloric restriction and IF result in similar degrees of weight loss and improvement in insulin sensitivity. Although these data suggest that IF may be a promising weight loss method, IF trials have been of moderate sample size and limited duration. More rigorous research is needed.
Collapse
Affiliation(s)
- Mary-Catherine Stockman
- Section of Endocrinology, Diabetes and Nutrition, Boston Medical Center, 720 Harrison Avenue, 8th Floor, Boston, MA, 02118, USA.
| | - Dylan Thomas
- Section of Endocrinology, Diabetes and Nutrition, Boston Medical Center, 720 Harrison Avenue, 8th Floor, Boston, MA, 02118, USA
| | - Jacquelyn Burke
- College of Health and Rehabilitation Sciences, Boston University Sargent College, 635 Commonwealth Avenue, Boston, MA, 02215, USA
| | - Caroline M Apovian
- Section of Endocrinology, Diabetes and Nutrition, Boston Medical Center, 720 Harrison Avenue, 8th Floor, Boston, MA, 02118, USA
| |
Collapse
|
17
|
Yu J, Yang J, Luo Y, Mengxue Y, Li W, Yang Y, He L, Xu J. The adverse effects of chronic low-dose exposure to nonylphenol on type 2 diabetes mellitus in high sucrose-high fat diet-treated rats. Islets 2018; 10:1-9. [PMID: 29215945 PMCID: PMC5800374 DOI: 10.1080/19382014.2017.1404211] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES Although it has been shown that exposure to environmental endocrine disruptors (EDCs) has been implicated as a potential risk factor for metabolic disease, information on adverse effect of chronic low-dose exposure to nonylphenol (NP), on the development and progress of type 2 diabetes mellitus (T2DM) is scarce. NP, as an EDC, is a ubiquitous degradation product of nonylphenol polyethoxylate (NPE) that is primarily used in cleaning and industrial processes. METHOD Eighty Sprague-Dawley rats were assigned into 8 groups (n = 10 per group): rats fed a normal-diet (ND) as the control (C-ND); rats fed a normal diet and were gavaged with NP at a dose level of 0.02 μg/kg/day (NP-L-ND), 0.2 μg/kg/day (NP-M-ND) or 2 μg/kg/day (NP-H-ND), respectively; rats fed a high-sucrose/high-fat diet (HSHFD) as the HSHFD control (C-HSHFD); rats fed a HSHFD and were gavaged with NP at a dose level of 0.02 μg/kg/day (NP-L-HSHFD), 0.2 μg/kg/day (NP-M-HSHFD) or 2 μg/kg/day (NP-H-HSHFD), respectively. RESULT On day 180, the rats in the groups treated with NP-M-HSHFD and NP-H-HSHFD showed significant increases in body weight (p < 0.05) in comparison with the C-ND group. Fast blood glucose (FBG) level in the NP-M-HSHFD and NP-H-HSHFD groups was higher than that in the C-ND group (F = 96.17, p < 0.001). The fast serum insulin (FINS) level of rats was lower in both the NP-M-HSHFD and NP-H-HSHFD groups compared with the C-ND group (F = 145.56, p < 0.001). Serum leptin (LEP) level in both the NP-M-HSHFD and NP-H-HSHFD groups was lower when compared with the C-ND group (F = 34.62, p < 0.001). The effect of NP at the dose level of 0.2 μg/kg/day on FBG, serum FINS and LEP levels in rats was greatest among the treatment groups (p < 0.05). Oral glucose tolerance test showed increased area under the curve (AUC) in treatment groups at week 12 (p < 0.05). A decrease of pancreatic islet numbers and size was exhibited in the pancreatic tissue of NP-M-HSHFD and NP-H-HSHFD treated rats compared with C-ND treated rats. Co-exposure to NP and HSHFD causes inflammatory changes histologically. CONCLUSION Chronic low-dose exposure to NP might induce impaired glucose tolerance, which further lead to insulin resistance, and pancreatic β cell insulin secretion deficiency, ultimately increase the risk of T2DM. Moreover, additive toxic effects of NP and HSHFD on pancreatic beta-cell function and glucose metabolism have been identified in rats as well.
Collapse
Affiliation(s)
- Jie Yu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jin Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Ya Luo
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yang Mengxue
- Department of Endocrinology, The First Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Wenmei Li
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yu Yang
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Liting He
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jie Xu
- School of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
- CONTACT Jie Xu School of Public Health, Zunyi Medical College, Zunyi, Guizhou 563000, China
| |
Collapse
|
18
|
Hjuler ST, Gydesen S, Andreassen KV, Karsdal MA, Henriksen K. The Dual Amylin- and Calcitonin-Receptor Agonist KBP-042 Works as Adjunct to Metformin on Fasting Hyperglycemia and HbA1c in a Rat Model of Type 2 Diabetes. J Pharmacol Exp Ther 2017; 362:24-30. [PMID: 28438778 DOI: 10.1124/jpet.117.241281] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 04/19/2017] [Indexed: 01/06/2023] Open
Abstract
KBP-042 is a dual amylin and calcitonin receptor agonist that increases glucose tolerance and insulin action and reduces body weight in rat models of obesity and prediabetes. The objective of the present study was to 1) evaluate KBP-042 as a treatment of late-stage type 2 diabetes in a rat model and 2) assess the value of adding KBP-042 to the standard of care, metformin, to consider KBP-042 as a relevant drug for treating patients with type 2 diabetes. Two studies were included: an intervention study and a prevention study. In the intervention study, treatment with 5 µg/kg KBP-042 was initiated in 11-week-old Zucker diabetic fatty (ZDF) rats, in which glucose tolerance, fasting glycemia, and glycated hemoglobin were assessed after 4 weeks. In the prevention study, either metformin (400 mg/kg), KBP-042 (5 µg/kg), or a combination of both were administered to ZDF rats for a total of 9 weeks. Glycemia, glucose tolerance, and insulin tolerance were tested. Furthermore, fasting plasma insulin and glucagon levels were evaluated. Finally, pancreatic content of insulin was assessed as a surrogate marker of beta-cell mass. It was found that KBP-042 was efficient in lowering fasting plasma glucose as well as improving glucose tolerance, both as prevention and intervention of disease progression. Furthermore, KBP-042 was efficient in combination with metformin and had additional effects compared with either therapy alone. In conclusion, KBP-042 is a highly relevant therapeutic candidate against type 2 diabetes, effective both as an add-on therapy to metformin and as a stand-alone therapy.
Collapse
Affiliation(s)
- Sara T Hjuler
- Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Sofie Gydesen
- Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Kim V Andreassen
- Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Morten A Karsdal
- Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Kim Henriksen
- Department of Musculoskeletal Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| |
Collapse
|
19
|
Abstract
Pancreatic islet β cells secrete insulin in response to nutrient secretagogues, like glucose, dependent on calcium influx and nutrient metabolism. One of the most intriguing qualities of β cells is their ability to use metabolism to amplify the amount of secreted insulin independent of further alterations in intracellular calcium. Many years studying this amplifying process have shaped our current understanding of β cell stimulus-secretion coupling; yet, the exact mechanisms of amplification have been elusive. Recent studies utilizing metabolomics, computational modeling, and animal models have progressed our understanding of the metabolic amplifying pathway of insulin secretion from the β cell. New approaches will be discussed which offer in-roads to a more complete model of β cell function. The development of β cell therapeutics may be aided by such a model, facilitating the targeting of aspects of the metabolic amplifying pathway which are unique to the β cell.
Collapse
Affiliation(s)
- Michael A Kalwat
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States.
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
20
|
Contreras C, Nogueiras R, Diéguez C, Medina-Gómez G, López M. Hypothalamus and thermogenesis: Heating the BAT, browning the WAT. Mol Cell Endocrinol 2016; 438:107-115. [PMID: 27498420 DOI: 10.1016/j.mce.2016.08.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 12/20/2022]
Abstract
Brown adipose tissue (BAT) has been also considered as the main thermogenic organ responsible of maintenance body temperature through heat production. However, a new type of thermogenic fat has been characterized during the last years, the beige or brite fat, that is developed from white adipose tissue (WAT) in response to different stimuli by a process known as browning. The activities of brown and beige adipocytes ameliorate metabolic disease, including obesity in mice and correlate with leanness in humans. Many genes and pathways that regulate brown and beige adipocyte biology have now been identified, providing a variety of promising therapeutic targets for metabolic disease. The hypothalamus is the main central place orchestrating the outflow signals that drive the sympathetic nerve activity to BAT and WAT, controlling heat production and energy homeostasis. Recent data have revealed new hypothalamic molecular mechanisms, such as hypothalamic AMP-activated protein kinase (AMPK), that control both thermogenesis and browning. This review provides an overview of the factors influencing BAT and WAT thermogenesis, with special focus on the integration of peripheral information on hypothalamic circuits controlling thermoregulation.
Collapse
Affiliation(s)
- Cristina Contreras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Carlos Diéguez
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain
| | - Gema Medina-Gómez
- Área de Bioquímica y Biología Molecular, Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, 28922, Alcorcón, Madrid, Spain
| | - Miguel López
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, 15706, Spain.
| |
Collapse
|
21
|
Vatier C, Fetita S, Boudou P, Tchankou C, Deville L, Riveline J, Young J, Mathivon L, Travert F, Morin D, Cahen J, Lascols O, Andreelli F, Reznik Y, Mongeois E, Madelaine I, Vantyghem M, Gautier J, Vigouroux C. One-year metreleptin improves insulin secretion in patients with diabetes linked to genetic lipodystrophic syndromes. Diabetes Obes Metab 2016; 18:693-7. [PMID: 26584826 DOI: 10.1111/dom.12606] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 11/16/2015] [Accepted: 11/16/2015] [Indexed: 12/21/2022]
Abstract
Recombinant methionyl human leptin (metreleptin) therapy was shown to improve hyperglycaemia, dyslipidaemia and insulin sensitivity in patients with lipodystrophic syndromes, but its effects on insulin secretion remain controversial. We used dynamic intravenous (i.v.) clamp procedures to measure insulin secretion, adjusted to insulin sensitivity, at baseline and after 1 year of metreleptin therapy, in 16 consecutive patients with lipodystrophy, diabetes and leptin deficiency. Patients, with a mean [± standard error of the mean (s.e.m.)] age of 39.2 (±4) years, presented with familial partial lipodystrophy (n = 11, 10 women) or congenital generalized lipodystrophy (n = 5, four women). Their mean (± s.e.m.) BMI (23.9 ± 0.7 kg/m(2) ), glycated haemoglobin levels (8.5 ± 0.4%) and serum triglycerides levels (4.6 ± 0.9 mmol/l) significantly decreased within 1 month of metreleptin therapy, then remained stable. Insulin sensitivity (from hyperglycaemic or euglycaemic-hyperinsulinaemic clamps, n = 4 and n = 12, respectively), insulin secretion during graded glucose infusion (n = 12), and acute insulin response to i.v. glucose adjusted to insulin sensitivity (disposition index, n = 12), significantly increased after 1 year of metreleptin therapy. The increase in disposition index was related to a decrease in percentage of total and trunk body fat. Metreleptin therapy improves not only insulin sensitivity, but also insulin secretion in patients with diabetes attributable to genetic lipodystrophies.
Collapse
Affiliation(s)
- C Vatier
- Sorbonne Universités, UPMC, Univ Paris 06, Paris, France
- Centre de Recherche Saint-Antoine, INSERM, UMR_S938, Paris, France
- ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
| | - S Fetita
- Service de Diabétologie et Endocrinologie, AP-HP, Groupe Hospitalier Lariboisière-Saint-Louis, Paris, France
| | - P Boudou
- Service de Biochimie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - C Tchankou
- Service de Diabétologie et Endocrinologie, AP-HP, Groupe Hospitalier Lariboisière-Saint-Louis, Paris, France
| | - L Deville
- Département de Pharmacie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jp Riveline
- Service de Diabétologie et Endocrinologie, AP-HP, Groupe Hospitalier Lariboisière-Saint-Louis, Paris, France
- Centre de Recherche des Cordeliers, INSERM, UMR_S1138, Paris, France
| | - J Young
- Service d'Endocrinologie et des Maladies de la Reproduction, AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
| | - L Mathivon
- Service de Pédiatrie, Centre Hospitalier de Meaux, Meaux, France
| | - F Travert
- Service d'Endocrinologie, Diabétologie, Nutrition, AP-HP, Hôpital Bichat, Paris, France
| | - D Morin
- Service de Pédiatrie, CHRU Montpellier, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - J Cahen
- Service d'Endocrinologie et Métabolismes, Centre Hospitalier, Argenteuil, France
| | - O Lascols
- Sorbonne Universités, UPMC, Univ Paris 06, Paris, France
- Centre de Recherche Saint-Antoine, INSERM, UMR_S938, Paris, France
- ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
- Laboratoire Commun de Biologie et Génétique Moléculaires, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - F Andreelli
- ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
- Service de Diabétologie, AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | - Y Reznik
- Service d'Endocrinologie, Centre Hospitalier Universitaire Côte-de-Nacre, Caen, France
| | - E Mongeois
- Service d'Endocrinologie, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - I Madelaine
- Département de Pharmacie, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Mc Vantyghem
- Service d'Endocrinologie et Métabolisme, Centre Hospitalier Régional Universitaire de Lille, Lille, France
| | - Jf Gautier
- Service de Diabétologie et Endocrinologie, AP-HP, Groupe Hospitalier Lariboisière-Saint-Louis, Paris, France
- Centre de Recherche des Cordeliers, INSERM, UMR_S1138, Paris, France
- University Paris-Diderot Paris-7, Paris, France
| | - C Vigouroux
- Sorbonne Universités, UPMC, Univ Paris 06, Paris, France
- Centre de Recherche Saint-Antoine, INSERM, UMR_S938, Paris, France
- ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
- Laboratoire Commun de Biologie et Génétique Moléculaires, AP-HP, Hôpital Saint-Antoine, Paris, France
| |
Collapse
|
22
|
Kusminski CM, Bickel PE, Scherer PE. Targeting adipose tissue in the treatment of obesity-associated diabetes. Nat Rev Drug Discov 2016; 15:639-660. [PMID: 27256476 DOI: 10.1038/nrd.2016.75] [Citation(s) in RCA: 518] [Impact Index Per Article: 57.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adipose tissue regulates numerous physiological processes, and its dysfunction in obese humans is associated with disrupted metabolic homeostasis, insulin resistance and type 2 diabetes mellitus (T2DM). Although several US-approved treatments for obesity and T2DM exist, these are limited by adverse effects and a lack of effective long-term glucose control. In this Review, we provide an overview of the role of adipose tissue in metabolic homeostasis and assess emerging novel therapeutic strategies targeting adipose tissue, including adipokine-based strategies, promotion of white adipose tissue beiging as well as reduction of inflammation and fibrosis.
Collapse
Affiliation(s)
- Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| | - Perry E Bickel
- Division of Endocrinology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, The University of Texas Southwestern Medical Center
| |
Collapse
|
23
|
Fontes-Villalba M, Lindeberg S, Granfeldt Y, Knop FK, Memon AA, Carrera-Bastos P, Picazo Ó, Chanrai M, Sunquist J, Sundquist K, Jönsson T. Palaeolithic diet decreases fasting plasma leptin concentrations more than a diabetes diet in patients with type 2 diabetes: a randomised cross-over trial. Cardiovasc Diabetol 2016; 15:80. [PMID: 27216013 PMCID: PMC4877952 DOI: 10.1186/s12933-016-0398-1] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/13/2016] [Indexed: 12/13/2022] Open
Abstract
Background We have previously shown that a Palaeolithic diet consisting of the typical food groups that our ancestors ate during the Palaeolithic era, improves cardiovascular disease risk factors and glucose control compared to the currently recommended diabetes diet in patients with type 2 diabetes. To elucidate the mechanisms behind these effects, we evaluated fasting plasma concentrations of glucagon, insulin, incretins, ghrelin, C-peptide and adipokines from the same study. Methods In a randomised, open-label, cross-over study, 13 patients with type 2 diabetes were randomly assigned to eat a Palaeolithic diet based on lean meat, fish, fruits, vegetables, root vegetables, eggs and nuts, or a diabetes diet designed in accordance with current diabetes dietary guidelines during two consecutive 3-month periods. The patients were recruited from primary health-care units and included three women and 10 men [age (mean ± SD) 64 ± 6 years; BMI 30 ± 7 kg/m2; diabetes duration 8 ± 5 years; glycated haemoglobin 6.6 ± 0.6 % (57.3 ± 6 mmol/mol)] with unaltered diabetes treatment and stable body weight for 3 months prior to the start of the study. Outcome variables included fasting plasma concentrations of leptin, adiponectin, adipsin, visfatin, resistin, glucagon, insulin, C-peptide, glucose-dependent insulinotropic polypeptide, glucagon-like peptide-1 and ghrelin. Dietary intake was evaluated by use of 4-day weighed food records. Results Seven participants started with the Palaeolithic diet and six with the diabetes diet. The Palaeolithic diet resulted in a large effect size (Cohen’s d = −1.26) at lowering fasting plasma leptin levels compared to the diabetes diet [mean difference (95 % CI), −2.3 (−5.1 to 0.4) ng/ml, p = 0.023]. No statistically significant differences between the diets for the other variables, analysed in this study, were observed. Conclusions Over a 3-month study period, a Palaeolithic diet resulted in reduced fasting plasma leptin levels, but did not change fasting levels of insulin, C-peptide, glucagon, incretins, ghrelin and adipokines compared to the currently recommended diabetes diet. Trial registration: ClinicalTrials.gov NCT00435240 Electronic supplementary material The online version of this article (doi:10.1186/s12933-016-0398-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maelán Fontes-Villalba
- Clinical Research Centre, Faculty of Medicine, Center for Primary Health Care Research, Lund University, Malmö, Sweden. .,, Calle José Betancort, 15, 35530, Teguise-Lanzarote, Spain.
| | - Staffan Lindeberg
- Clinical Research Centre, Faculty of Medicine, Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Yvonne Granfeldt
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Filip K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Ashfaque A Memon
- Clinical Research Centre, Faculty of Medicine, Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Pedro Carrera-Bastos
- Clinical Research Centre, Faculty of Medicine, Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Óscar Picazo
- NutriScience-Education and Consulting, Lda, Lisbon, Portugal
| | | | - Jan Sunquist
- Clinical Research Centre, Faculty of Medicine, Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Kristina Sundquist
- Clinical Research Centre, Faculty of Medicine, Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Tommy Jönsson
- Clinical Research Centre, Faculty of Medicine, Center for Primary Health Care Research, Lund University, Malmö, Sweden
| |
Collapse
|
24
|
Gao S, Stevens JR, Butler AA. Adropin - a circulating factor in metabolic control or a drop in the ocean? Expert Rev Endocrinol Metab 2016; 11:239-241. [PMID: 30058929 DOI: 10.1080/17446651.2016.1175938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Su Gao
- a Cardiovascular Research Center , University of Alberta , Edmonton , Canada
| | - Joseph R Stevens
- b Department of Pharmacology & Physiology , Saint Louis University School of Medicine , St. Louis , MO , USA
| | - Andrew A Butler
- b Department of Pharmacology & Physiology , Saint Louis University School of Medicine , St. Louis , MO , USA
| |
Collapse
|
25
|
DiSilvestro DJ, Melgar-Bermudez E, Yasmeen R, Fadda P, Lee LJ, Kalyanasundaram A, Gilor CL, Ziouzenkova O. Leptin Production by Encapsulated Adipocytes Increases Brown Fat, Decreases Resistin, and Improves Glucose Intolerance in Obese Mice. PLoS One 2016; 11:e0153198. [PMID: 27055280 PMCID: PMC4824514 DOI: 10.1371/journal.pone.0153198] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 03/24/2016] [Indexed: 12/03/2022] Open
Abstract
The neuroendocrine effects of leptin on metabolism hold promise to be translated into a complementary therapy to traditional insulin therapy for diabetes and obesity. However, injections of leptin can provoke inflammation. We tested the effects of leptin, produced in the physiological adipocyte location, on metabolism in mouse models of genetic and dietary obesity. We generated 3T3-L1 adipocytes constitutively secreting leptin and encapsulated them in a poly-L-lysine membrane, which protects the cells from immune rejection. Ob/ob mice (OB) were injected with capsules containing no cells (empty, OB[Emp]), adipocytes (OB[3T3]), or adipocytes overexpressing leptin (OB[Lep]) into both visceral fat depots. Leptin was found in the plasma of OB[Lep], but not OB[Emp] and OB[3T3] mice at the end of treatment (72 days). The OB[Lep] and OB[3T3] mice have transiently suppressed appetite and weight loss compared to OB[Emp]. Only OB[Lep] mice have greater brown fat mass, metabolic rate, and reduced resistin plasma levels compared to OB[Emp]. Glucose tolerance was markedly better in OB[Lep]vs. OB[Emp] and OB[3T3] mice as well as in wild type mice with high-fat diet-induced obesity and insulin resistance treated with encapsulated leptin-producing adipocytes. Our proof-of-principle study provides evidence of long-term improvement of glucose tolerance with encapsulated adipocytes producing leptin.
Collapse
Affiliation(s)
- David J. DiSilvestro
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Emiliano Melgar-Bermudez
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Rumana Yasmeen
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Paolo Fadda
- Genomics Shared Resource, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - L. James Lee
- NSF Nanoscale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices, The Ohio State University, Columbus, Ohio, United States of America
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Chen L. Gilor
- Veterinary Clinical Sciences, The Ohio State University, Columbus, Ohio, 43210, United States of America
| | - Ouliana Ziouzenkova
- Department of Human Sciences, The Ohio State University, Columbus, Ohio, 43210, United States of America
- * E-mail:
| |
Collapse
|
26
|
Goldstein I, Hager GL. Transcriptional and Chromatin Regulation during Fasting - The Genomic Era. Trends Endocrinol Metab 2015; 26:699-710. [PMID: 26520657 PMCID: PMC4673016 DOI: 10.1016/j.tem.2015.09.005] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/10/2015] [Accepted: 09/12/2015] [Indexed: 12/21/2022]
Abstract
An elaborate metabolic response to fasting is orchestrated by the liver and is heavily reliant on transcriptional regulation. In response to hormones (glucagon, glucocorticoids) many transcription factors (TFs) are activated and regulate various genes involved in metabolic pathways aimed at restoring homeostasis: gluconeogenesis, fatty acid oxidation, ketogenesis, and amino acid shuttling. We summarize recent discoveries regarding fasting-related TFs with an emphasis on genome-wide binding patterns. Collectively, the findings we discuss reveal a large degree of cooperation between TFs during fasting that occurs at motif-rich DNA sites bound by a combination of TFs. These new findings implicate transcriptional and chromatin regulation as major determinants of the response to fasting and unravels the complex, multi-TF nature of this response.
Collapse
Affiliation(s)
- Ido Goldstein
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National institutes of Health, Bethesda, MD, 20892, USA.
| | - Gordon L Hager
- Laboratory of Receptor Biology and Gene Expression, The National Cancer Institute, The National institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
27
|
Angptl4 links α-cell proliferation following glucagon receptor inhibition with adipose tissue triglyceride metabolism. Proc Natl Acad Sci U S A 2015; 112:15498-503. [PMID: 26621734 DOI: 10.1073/pnas.1513872112] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Type 2 diabetes is characterized by a reduction in insulin function and an increase in glucagon activity that together result in hyperglycemia. Glucagon receptor antagonists have been developed as drugs for diabetes; however, they often increase glucagon plasma levels and induce the proliferation of glucagon-secreting α-cells. We find that the secreted protein Angiopoietin-like 4 (Angptl4) is up-regulated via Pparγ activation in white adipose tissue and plasma following an acute treatment with a glucagon receptor antagonist. Induction of adipose angptl4 and Angptl4 supplementation promote α-cell proliferation specifically. Finally, glucagon receptor antagonist improves glycemia in diet-induced obese angptl4 knockout mice without increasing glucagon levels or α-cell proliferation, underscoring the importance of this protein. Overall, we demonstrate that triglyceride metabolism in adipose tissue regulates α-cells in the endocrine pancreas.
Collapse
|
28
|
Glucagon receptor antibody completely suppresses type 1 diabetes phenotype without insulin by disrupting a novel diabetogenic pathway. Proc Natl Acad Sci U S A 2015; 112:2503-8. [PMID: 25675519 DOI: 10.1073/pnas.1424934112] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Insulin monotherapy can neither maintain normoglycemia in type 1 diabetes (T1D) nor prevent the long-term damage indicated by elevated glycation products in blood, such as glycated hemoglobin (HbA1c). Here we find that hyperglycemia, when unaccompanied by an acute increase in insulin, enhances itself by paradoxically stimulating hyperglucagonemia. Raising glucose from 5 to 25 mM without insulin enhanced glucagon secretion ∼two- to fivefold in InR1-G9 α cells and ∼18-fold in perfused pancreata from insulin-deficient rats with T1D. Mice with T1D receiving insulin treatment paradoxically exhibited threefold higher plasma glucagon during hyperglycemic surges than during normoglycemic intervals. Blockade of glucagon action with mAb Ac, a glucagon receptor (GCGR) antagonizing antibody, maintained glucose below 100 mg/dL and HbA1c levels below 4% in insulin-deficient mice with T1D. In rodents with T1D, hyperglycemia stimulates glucagon secretion, up-regulating phosphoenolpyruvate carboxykinase and enhancing hyperglycemia. GCGR antagonism in mice with T1D normalizes glucose and HbA1c, even without insulin.
Collapse
|