1
|
Amorim MR, Aung O, Mokhlesi B, Polotsky VY. Leptin-mediated neural targets in obesity hypoventilation syndrome. Sleep 2022; 45:zsac153. [PMID: 35778900 PMCID: PMC9453616 DOI: 10.1093/sleep/zsac153] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/20/2022] [Indexed: 07/30/2023] Open
Abstract
Obesity hypoventilation syndrome (OHS) is defined as daytime hypercapnia in obese individuals in the absence of other underlying causes. In the United States, OHS is present in 10%-20% of obese patients with obstructive sleep apnea and is linked to hypoventilation during sleep. OHS leads to high cardiorespiratory morbidity and mortality, and there is no effective pharmacotherapy. The depressed hypercapnic ventilatory response plays a key role in OHS. The pathogenesis of OHS has been linked to resistance to an adipocyte-produced hormone, leptin, a major regulator of metabolism and control of breathing. Mechanisms by which leptin modulates the control of breathing are potential targets for novel therapeutic strategies in OHS. Recent advances shed light on the molecular pathways related to the central chemoreceptor function in health and disease. Leptin signaling in the nucleus of the solitary tract, retrotrapezoid nucleus, hypoglossal nucleus, and dorsomedial hypothalamus, and anatomical projections from these nuclei to the respiratory control centers, may contribute to OHS. In this review, we describe current views on leptin-mediated mechanisms that regulate breathing and CO2 homeostasis with a focus on potential therapeutics for the treatment of OHS.
Collapse
Affiliation(s)
- Mateus R Amorim
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - O Aung
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Babak Mokhlesi
- Department of Internal Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Vsevolod Y Polotsky
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Abstract
Diabetes is a chronic metabolic disease affecting an increasing number of people. Although diabetes has negative health outcomes for diagnosed individuals, a population at particular risk are pregnant women, as diabetes impacts not only a pregnant woman's health but that of her child. In this review, we cover the current knowledge and unanswered questions on diabetes affecting an expectant mother, focusing on maternal and fetal outcomes.
Collapse
Affiliation(s)
- Cecilia González Corona
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA
| | - Ronald J. Parchem
- Center for Cell and Gene Therapy, Stem Cells and Regenerative Medicine Center, One Baylor Plaza, Houston, TX 77030, USA,Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
3
|
Xu S, Ye B, Li J, Dou Y, Yu Y, Feng Y, Wang L, Wan DCC, Rong X. Astragalus mongholicus powder, a traditional Chinese medicine formula ameliorate type 2 diabetes by regulating adipoinsular axis in diabetic mice. Front Pharmacol 2022; 13:973927. [PMID: 36046814 PMCID: PMC9420938 DOI: 10.3389/fphar.2022.973927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
The global morbidity of obesity and type 2 diabetes mellitus (T2DM) has dramatically increased. Insulin resistance is the most important pathogenesis and therapeutic target of T2DM. The traditional Chinese medicine formula Astragalus mongholicus powder (APF), consists of Astragalus mongholicus Bunge [Fabaceae], Pueraria montana (Lour.) Merr. [Fabaceae], and Morus alba L. [Moraceae] has a long history to be used to treat diabetes in ancient China. This work aims to investigate the effects of APF on diabetic mice and its underlying mechanism. Diabetic mice were induced by High-fat-diet (HFD) and streptozotocin (STZ). The body weight of mice and their plasma levels of glucose, insulin, leptin and lipids were examined. Reverse transcription-polymerase chain reaction, histology, and Western blot analysis were performed to validate the effects of APF on diabetic mice and investigate the underlying mechanism. APF reduced hyperglycemia, hyperinsulinemia, and hyerleptinemia and attenuate the progression of obesity and non-alcoholic fatty liver disease (NAFLD). However, these effects disappeared in leptin deficient ob/ob diabetic mice and STZ-induced insulin deficient type 1 diabetic mice. Destruction of either these hormones would abolish the therapeutic effects of APF. In addition, APF inhibited the protein expression of PTP1B suppressing insulin–leptin sensitivity, the gluconeogenic gene PEPCK, and the adipogenic gene FAS. Therefore, insulin–leptin sensitivity was normalized, and the gluconeogenic and adipogenic genes were suppressed. In conclusion, APF attenuated obesity, NAFLD, and T2DM by regulating the balance of adipoinsular axis in STZ + HFD induced T2DM mice.
Collapse
Affiliation(s)
- Siyuan Xu
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Bixian Ye
- Department of Nursing, Medical College of Jiaying University, Meizhou, China
| | - Jinlei Li
- School of Chinese Meteria Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yonghui Dou
- School of Chinese Meteria Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuying Yu
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yifan Feng
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lexun Wang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - David Chi-Cheong Wan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xianglu Rong
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education of China, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Xianglu Rong,
| |
Collapse
|
4
|
Angelidi AM, Belanger MJ, Kokkinos A, Koliaki CC, Mantzoros CS. Novel Noninvasive Approaches to the Treatment of Obesity: From Pharmacotherapy to Gene Therapy. Endocr Rev 2022; 43:507-557. [PMID: 35552683 PMCID: PMC9113190 DOI: 10.1210/endrev/bnab034] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Indexed: 02/08/2023]
Abstract
Recent insights into the pathophysiologic underlying mechanisms of obesity have led to the discovery of several promising drug targets and novel therapeutic strategies to address the global obesity epidemic and its comorbidities. Current pharmacologic options for obesity management are largely limited in number and of modest efficacy/safety profile. Therefore, the need for safe and more efficacious new agents is urgent. Drugs that are currently under investigation modulate targets across a broad range of systems and tissues, including the central nervous system, gastrointestinal hormones, adipose tissue, kidney, liver, and skeletal muscle. Beyond pharmacotherapeutics, other potential antiobesity strategies are being explored, including novel drug delivery systems, vaccines, modulation of the gut microbiome, and gene therapy. The present review summarizes the pathophysiology of energy homeostasis and highlights pathways being explored in the effort to develop novel antiobesity medications and interventions but does not cover devices and bariatric methods. Emerging pharmacologic agents and alternative approaches targeting these pathways and relevant research in both animals and humans are presented in detail. Special emphasis is given to treatment options at the end of the development pipeline and closer to the clinic (ie, compounds that have a higher chance to be added to our therapeutic armamentarium in the near future). Ultimately, advancements in our understanding of the pathophysiology and interindividual variation of obesity may lead to multimodal and personalized approaches to obesity treatment that will result in safe, effective, and sustainable weight loss until the root causes of the problem are identified and addressed.
Collapse
Affiliation(s)
- Angeliki M Angelidi
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew J Belanger
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alexander Kokkinos
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Chrysi C Koliaki
- First Department of Propaedeutic Medicine, Medical School, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Christos S Mantzoros
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA, USA
- Department of Medicine Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Kamstra K, Rizwan MZ, Grattan DR, Horsfield JA, Tups A. Leptin regulates glucose homeostasis via the canonical Wnt pathway in the zebrafish. FASEB J 2022; 36:e22207. [PMID: 35188286 DOI: 10.1096/fj.202101764r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 11/11/2022]
Abstract
Leptin is best known for its role in adipostasis, but it also regulates blood glucose levels. The molecular mechanism by which leptin controls glucose homeostasis remains largely unknown. Here, we use a zebrafish model to show that Wnt signaling mediates the glucoregulatory effects of leptin. Under normal feeding conditions, leptin regulates glucose homeostasis but not adipostasis in zebrafish. In times of nutrient excess, however, we found that leptin also regulates body weight and size. Using a Wnt signaling reporter fish, we show that leptin activates the canonical Wnt pathway in vivo. Utilizing two paradigms for hyperglycemia, it is revealed that leptin regulates glucose homeostasis via the Wnt pathway, as pharmacological inhibition of this pathway impairs the glucoregulatory actions of leptin. Our results may shed new light on the evolution of the physiological function of leptin.
Collapse
Affiliation(s)
- Kaj Kamstra
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Physiology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Mohammed Z Rizwan
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Anatomy, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - David R Grattan
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Anatomy, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Julia A Horsfield
- Department of Pathology, University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, Auckland, New Zealand
| | - Alexander Tups
- Centre for Neuroendocrinology and Brain Health Research Centre, University Otago, Dunedin, New Zealand.,Department of Physiology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| |
Collapse
|
6
|
Clark KA, Shin AC, Sirivelu MP, MohanKumar RC, Maddineni SR, Ramachandran R, MohanKumar PS, MohanKumar SMJ. Evaluation of the Central Effects of Systemic Lentiviral-Mediated Leptin Delivery in Streptozotocin-Induced Diabetic Rats. Int J Mol Sci 2021; 22:ijms222413197. [PMID: 34947993 PMCID: PMC8703968 DOI: 10.3390/ijms222413197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/23/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Type 1 diabetes (T1D) is characterized by hyperphagia, hyperglycemia and activation of the hypothalamic-pituitary-adrenal (HPA) axis. We have reported previously that daily leptin injections help to alleviate these symptoms. Therefore, we hypothesized that leptin gene therapy could help to normalize the neuroendocrine dysfunction seen in T1D. Adult male Sprague Dawley rats were injected i.v. with a lentiviral vector containing the leptin gene or green fluorescent protein. Ten days later, they were injected with the vehicle or streptozotocin (STZ). HPA function was assessed by measuring norepinephrine (NE) levels in the paraventricular nucleus (PVN) and serum corticosterone (CS). Treatment with the leptin lentiviral vector (Lepvv) increased leptin and insulin levels in non-diabetic rats, but not in diabetic animals. There was a significant reduction in blood glucose levels in diabetic rats due to Lepvv treatment. Both NE levels in the PVN and serum CS were reduced in diabetic rats treated with Lepvv. Results from this study provide evidence that leptin gene therapy in STZ-induced diabetic rats was able to partially normalize some of the neuroendocrine abnormalities, but studies with higher doses of the Lepvv are needed to develop this into a viable option for treating T1D.
Collapse
MESH Headings
- Animals
- Corticosterone/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/genetics
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/therapy
- Disease Models, Animal
- Genetic Therapy
- Genetic Vectors/administration & dosage
- Injections, Intravenous
- Lentivirus/genetics
- Leptin/genetics
- Male
- Norepinephrine/metabolism
- Paraventricular Hypothalamic Nucleus/metabolism
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Kimberly A. Clark
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
| | - Andrew C. Shin
- Neurobiology of Nutrition Laboratory, Department of Nutritional Sciences, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA;
| | - Madhu P. Sirivelu
- Pathobiology and Diagnostic Investigation, Michigan State University, E. Lansing, MI 48824, USA;
| | - Ramya C. MohanKumar
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
| | - Sreenivasa R. Maddineni
- Department of Poultry Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.R.M.); (R.R.)
| | - Ramesh Ramachandran
- Department of Poultry Science, College of Agricultural Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.R.M.); (R.R.)
| | - Puliyur S. MohanKumar
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
- Pathobiology and Diagnostic Investigation, Michigan State University, E. Lansing, MI 48824, USA;
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
- Department of Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
| | - Sheba M. J. MohanKumar
- Neuroscience Graduate Program, Michigan State University, E. Lansing, MI 48824, USA; (K.A.C.); (P.S.M.)
- Neuroendocrine Research Laboratory, University of Georgia, Athens, GA 30602, USA;
- Department of Biomedical Sciences, University of Georgia, Athens, GA 30602, USA
- Correspondence: ; Tel.: +1-706-542-1945
| |
Collapse
|
7
|
Ectopic Leptin Production by Intraocular Pancreatic Islet Organoids Ameliorates the Metabolic Phenotype of ob/ob Mice. Metabolites 2021; 11:metabo11060387. [PMID: 34198579 PMCID: PMC8231910 DOI: 10.3390/metabo11060387] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 06/11/2021] [Indexed: 11/17/2022] Open
Abstract
The pancreatic islets of Langerhans consist of endocrine cells that secrete peptide hormones into the blood circulation in response to metabolic stimuli. When transplanted into the anterior chamber of the eye (ACE), pancreatic islets engraft and maintain morphological features of native islets as well as islet-specific vascularization and innervation patterns. In sufficient amounts, intraocular islets are able to maintain glucose homeostasis in diabetic mice. Islet organoids (pseudo-islets), which are formed by self-reassembly of islet cells following disaggregation and genetic manipulation, behave similarly to native islets. Here, we tested the hypothesis that genetically engineered intraocular islet organoids can serve as production sites for leptin. To test this hypothesis, we chose the leptin-deficient ob/ob mouse as a model system, which becomes severely obese, hyperinsulinemic, hyperglycemic, and insulin resistant. We generated a Tet-OFF-based beta-cell-specific adenoviral expression construct for mouse leptin, which allowed efficient transduction of native beta-cells, optical monitoring of leptin expression by co-expressed fluorescent proteins, and the possibility to switch-off leptin expression by treatment with doxycycline. Intraocular transplantation of islet organoids formed from transduced islet cells, which lack functional leptin receptors, to ob/ob mice allowed optical monitoring of leptin expression and ameliorated their metabolic phenotype by improving bodyweight, glucose tolerance, serum insulin, and C-peptide levels.
Collapse
|
8
|
Romanelli SM, MacDougald OA. Viral and Nonviral Transfer of Genetic Materials to Adipose Tissues: Toward a Gold Standard Approach. Diabetes 2020; 69:2581-2588. [PMID: 33219099 PMCID: PMC7679771 DOI: 10.2337/dbi20-0036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/04/2020] [Indexed: 01/03/2023]
Abstract
Gene transfer using viral or nonviral vectors enables the ability to manipulate specific cells and tissues for gene silencing, protein overexpression, or genome modification. Despite the widespread application of viral- and non-viral-mediated gene transfer to liver, heart, skeletal muscle, and the central nervous system, its use in adipose tissue has been limited. This is largely because adipose tissue is distributed throughout the body in distinct depots and adipocytes make up a minority of the cells within the tissue, making transduction difficult. Currently, there is no consensus methodology for efficient gene transfer to adipose tissue and many studies report conflicting information with regard to transduction efficiency and vector biodistribution. In this review, we summarize the challenges associated with gene transfer to adipose tissue and report on innovations that improve efficacy. We describe how vector and route of administration are the two key factors that influence transduction efficiency and outline a "gold standard" approach and experimental workflow for validating gene transfer to adipose tissue. Lastly, we speculate on how CRISPR/Cas9 can be integrated to improve adipose tissue research.
Collapse
Affiliation(s)
- Steven M Romanelli
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
| | - Ormond A MacDougald
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
9
|
Roth CL, von Schnurbein J, Elfers C, Moss A, Wabitsch M. Changes in Satiety Hormones in Response to Leptin Treatment in a Patient with Leptin Deficiency. Horm Res Paediatr 2019; 90:424-430. [PMID: 29996141 DOI: 10.1159/000489884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/04/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND We tested whether leptin treatment affects secretion of satiety-related gut peptides and brain-derived neurotrophic factor (BDNF), which is a regulator of energy homeostasis downstream of hypothalamic leptin signaling. METHODS We report the case of a morbidly obese 14.7-year-old girl with a novel previously reported homozygous leptin gene mutation, in whom hormone secretion was evaluated in 30-min intervals for 10 h (07.30-17.30) to assess BDNF, insulin, glucagon-like peptide-1 (GLP-1), ghrelin, and peptide YY (PYY) secretion before as well as 11 and 46 weeks after start of metreleptin treatment. RESULTS Leptin substitution resulted in strong reductions of body fat and calorie intake. Insulin secretion increased by 58.9% after 11 weeks, but was reduced by -44.8% after 46 weeks compared to baseline. Similarly, GLP-1 increased after 11 weeks (+15.2%) and decreased after 46 weeks. PYY increased consistently (+5%/ +13.2%, after 11/46 weeks). Ghrelin decreased after 46 weeks (-11%). BDNF secretion was not affected by leptin treatment. CONCLUSION The strong increase in insulin and GLP-1 secretion after 11 weeks of metreleptin treatment cannot be explained by reduced adiposity and might contribute to improved central satiety. Observed changes of PYY can lead to increased satiety as well. However, leptin replacement does not seem to affect circulating BDNF levels.
Collapse
Affiliation(s)
- Christian L Roth
- Seattle Children's Research Institute, Center for Integrative Brain Research, University of Washington, Department of Pediatrics, Seattle, Washington, USA,
| | - Julia von Schnurbein
- Division of Pediatric Endocrinology, Diabetes and Obesity Unit, Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| | - Clinton Elfers
- Seattle Children's Research Institute, Center for Integrative Brain Research, University of Washington, Department of Pediatrics, Seattle, Washington, USA
| | - Anja Moss
- Division of Pediatric Endocrinology, Diabetes and Obesity Unit, Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology, Diabetes and Obesity Unit, Department of Pediatrics and Adolescent Medicine, Ulm University, Ulm, Germany
| |
Collapse
|
10
|
Toffoli B, Bernardi S, Winkler C, Carrascosa C, Gilardi F, Desvergne B. Renal mineralocorticoid receptor expression is reduced in lipoatrophy. FEBS Open Bio 2019; 9:328-334. [PMID: 30761257 PMCID: PMC6356154 DOI: 10.1002/2211-5463.12579] [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: 04/12/2018] [Revised: 11/29/2018] [Accepted: 12/12/2018] [Indexed: 11/29/2022] Open
Abstract
Obesity is a condition characterized by adipose tissue hypertrophy; it is estimated that the obesity epidemic accounted for 4 million deaths in 2015 and that 70% of these were due to cardiovascular disease (CVD). One of the mechanisms linking obesity to CVD is the ability of adipose tissue to secrete circulating factors. We hypothesized that adipose tissue and its secretory products may influence mineralocorticoid receptor (MR) expression. Here, we showed that expression of MR and its downstream targets (Cnksr3, Scnn1b, and Sgk1) were significantly reduced in the kidneys of peroxisome proliferator‐activated receptor‐γ null (PpargΔ/Δ) and A‐ZIP/F‐1 (AZIPtg/+) lipoatrophic mice with respect to their controls. Intriguingly, MR expression was also found to be significantly reduced in the kidneys of genetically obese ob/ob mice. Our data suggest that adipose tissue contributes to the regulation of MR expression. Given that leptin deficiency seems to be the major feature shared by PpargΔ/Δ, AZIPtg/+, and ob/ob mice, we speculate that adipose tissue modulates MR expression through the leptin system.
Collapse
Affiliation(s)
- Barbara Toffoli
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne Switzerland
| | | | - Carine Winkler
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne Switzerland
| | - Coralie Carrascosa
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne Switzerland
| | - Federica Gilardi
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne Switzerland
| | - Béatrice Desvergne
- Center for Integrative Genomics Faculty of Biology and Medicine University of Lausanne Switzerland
| |
Collapse
|
11
|
Pharmacodynamical effects of orally administered exenatide nanoparticles embedded in gastro-resistant microparticles. Eur J Pharm Biopharm 2018; 133:214-223. [DOI: 10.1016/j.ejpb.2018.10.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/17/2018] [Accepted: 10/17/2018] [Indexed: 01/22/2023]
|
12
|
Hayakawa J, Wang M, Wang C, Han RH, Jiang ZY, Han X. Lipidomic analysis reveals significant lipogenesis and accumulation of lipotoxic components in ob/ob mouse organs. Prostaglandins Leukot Essent Fatty Acids 2018; 136:161-169. [PMID: 28110829 PMCID: PMC6203299 DOI: 10.1016/j.plefa.2017.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/03/2016] [Accepted: 01/03/2017] [Indexed: 12/13/2022]
Abstract
To further understand the role of lipogenesis and lipotoxicity in the development of obesity and diabetes, lipidomes of various organs from ob/ob mice and their wild type controls were analyzed by shotgun lipidomics at 10, 12, and 16 weeks of age. We observed that the amounts of fatty acyl (FA) chains corresponding to those from de novo synthesis (e.g., 16:0, 16:1, and 18:1 FA) were substantially elevated in ob/ob mice, consistent with increased expression of genes and proteins involved in biosynthesis. Polyunsaturated fatty acid species were moderately increased in the examined tissues of ob/ob mice, since they can only be absorbed from diets or elongated from the ingested n-3 or n-6 FA. Different profiles of FA chains between ob/ob mouse liver and skeletal muscle reflect diverging lipogenesis pathways in these organs. Amounts of vaccenic acids (i.e., 18:1(n-7) FA) in 12- and 16-week ob/ob mouse liver were significantly increased compared to their controls, indicating enhanced de novo synthesis of this acid through 16:1(n-7) FA in the liver starting at 12 weeks of age. Coincidentally, synthesis of triacylglycerol from monoacylglycerol in the liver was also increased in ob/ob mice starting at 12 weeks of age, as revealed by simulation of triacylglycerol synthesis. Moreover, levels of lipotoxic lipid classes were significantly higher in ob/ob mice than their age-matched controls, supporting the notion that elevated lipotoxic components are tightly associated with insulin resistance in ob/ob mice. Taken together, the current study revealed that lipogenesis and lipotoxicity in ob/ob mice likely contribute to insulin resistance and provides great insights into the underlying mechanisms of diabetes and obesity.
Collapse
Affiliation(s)
- Jun Hayakawa
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Miao Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Chunyan Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Rowland H Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA
| | - Zhen Y Jiang
- Department of Pharmacology & Experimental Therapeutics, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL 32827, USA.
| |
Collapse
|
13
|
Ferguson D, Blenden M, Hutson I, Du Y, Harris CA. Mouse Embryonic Fibroblasts Protect ob/ob Mice From Obesity and Metabolic Complications. Endocrinology 2018; 159:3275-3286. [PMID: 30085057 PMCID: PMC6109302 DOI: 10.1210/en.2018-00561] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 07/25/2018] [Indexed: 12/22/2022]
Abstract
The global obesity epidemic is fueling alarming rates of diabetes, associated with increased risk of cardiovascular disease and cancer. Leptin is a hormone secreted by adipose tissue that is a key regulator of body weight (BW) and energy expenditure. Leptin-deficient humans and mice are obese, diabetic, and infertile and have hepatic steatosis. Although leptin replacement therapy can alleviate the pathologies seen in leptin-deficient patients and mouse models, treatment is costly and requires daily injections. Because adipocytes are the source of leptin secretion, we investigated whether mouse embryonic fibroblasts (MEFs), capable of forming adipocytes, could be injected into ob/ob mice and prevent the metabolic phenotype seen in these leptin-deficient mice. We performed a single subcutaneous injection of MEFs into leptin-deficient ob/ob mice. The MEF injection formed a single fat pad that is histologically similar to white adipose tissue. The ob/ob mice receiving MEFs (obRs) had significantly lower BW compared with nontreated ob/ob mice, primarily because of decreased adipose tissue mass. Additionally, obR mice had significantly less liver steatosis and greater glucose tolerance and insulin sensitivity. obR mice also manifested lower food intake and greater energy expenditure than ob/ob mice, providing a mechanism underlying their metabolic improvement. Furthermore, obRs have sustained metabolic protection and restoration of fertility. Collectively, our studies show the importance of functional adipocytes in preventing metabolic abnormalities seen in leptin deficiency and point to the possibility of cell-based therapies for the treatment of leptin-deficient states.
Collapse
Affiliation(s)
- Daniel Ferguson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Mitchell Blenden
- Department of Medical Education, College of Medicine, University of Central Florida, Orlando, Florida
| | - Irina Hutson
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Yingqiu Du
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
| | - Charles A Harris
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Veterans Affairs St. Louis Healthcare System, John Cochran Division, St. Louis, Missouri
| |
Collapse
|
14
|
Borck PC, Vettorazzi JF, Branco RCS, Batista TM, Santos-Silva JC, Nakanishi VY, Boschero AC, Ribeiro RA, Carneiro EM. Taurine supplementation induces long-term beneficial effects on glucose homeostasis in ob/ob mice. Amino Acids 2018; 50:765-774. [DOI: 10.1007/s00726-018-2553-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 03/11/2018] [Indexed: 10/17/2022]
|
15
|
Endotoxemia-mediated activation of acetyltransferase P300 impairs insulin signaling in obesity. Nat Commun 2017; 8:131. [PMID: 28743992 PMCID: PMC5526866 DOI: 10.1038/s41467-017-00163-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 06/01/2017] [Indexed: 12/16/2022] Open
Abstract
Diabetes and obesity are characterized by insulin resistance and chronic low-grade inflammation. An elevated plasma concentration of lipopolysaccharide (LPS) caused by increased intestinal permeability during diet-induced obesity promotes insulin resistance in mice. Here, we show that LPS induces endoplasmic reticulum (ER) stress and protein levels of P300, an acetyltransferase involved in glucose production. In high-fat diet fed and genetically obese ob/ob mice, P300 translocates from the nucleus into the cytoplasm of hepatocytes. We also demonstrate that LPS activates the transcription factor XBP1 via the ER stress sensor IRE1, resulting in the induction of P300 which, in turn, acetylates IRS1/2, inhibits its association with the insulin receptor, and disrupts insulin signaling. Pharmacological inhibition of P300 acetyltransferase activity by a specific inhibitor improves insulin sensitivity and decreases hyperglycemia in obese mice. We suggest that P300 acetyltransferase activity may be a promising therapeutic target for the treatment of obese patients.Elevated plasma LPS levels have been associated with insulin resistance. Here Cao et al. show that LPS induces ER stress and P300 activity via the XBP1/IRE1 pathway. P300 acetylates IRS1/2 and inhibits its binding with the insulin receptor. The consequent impairment of insulin signaling can be rescued by pharmacological inhibition of P300.
Collapse
|
16
|
Takanashi M, Taira Y, Okazaki S, Takase S, Kimura T, Li CC, Xu PF, Noda A, Sakata I, Kumagai H, Ikeda Y, Iizuka Y, Yahagi N, Shimano H, Osuga JI, Ishibashi S, Kadowaki T, Okazaki H. Role of Hormone-sensitive Lipase in Leptin-Promoted Fat Loss and Glucose Lowering. J Atheroscler Thromb 2017; 24:1105-1116. [PMID: 28413180 PMCID: PMC5684476 DOI: 10.5551/jat.39552] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: Myriad biological effects of leptin may lead to broad therapeutic applications for various metabolic diseases, including diabetes and its complications; however, in contrast to its anorexic effect, the molecular mechanisms underlying adipopenic and glucose-lowering effects of leptin have not been fully understood. Here we aim to clarify the role of hormone-sensitive lipase (HSL) in leptin's action. Methods: Wild-type (WT) and HSL-deficient (HSLKO) mice were made hyperleptinemic by two commonly-used methods: adenovirus-mediated overexpression of leptin and continuous subcutaneous infusion of leptin by osmotic pumps. The amount of food intake, body weights, organ weights, and parameters of glucose and lipid metabolism were measured. Results: Hyperleptinemia equally suppressed the food intake in WT and HSLKO mice. On the other hand, leptin-mediated fat loss and glucose-lowering were significantly blunted in the absence of HSL when leptin was overexpressed by recombinant adenovirus carrying leptin. By osmotic pumps, the fat-losing and glucose-lowering effects of leptin were milder due to lower levels of hyperleptinemia; although the difference between WT and HSLKO mice did not reach statistical significance, HSLKO mice had a tendency to retain more fat than WT mice in the face of hyperleptinemia. Conclusions: We clarify for the first time the role of HSL in leptin's effect using a genetic model: leptin-promoted fat loss and glucose-lowering are at least in part mediated via HSL-mediated lipolysis. Further studies to define the pathophysiological role of adipocyte lipases in leptin action may lead to a new therapeutic approach to circumvent leptin resistance.
Collapse
Affiliation(s)
- Mikio Takanashi
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Yoshino Taira
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Sachiko Okazaki
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Satoru Takase
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Takeshi Kimura
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Cheng Cheng Li
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Peng Fei Xu
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Akari Noda
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University
| | - Hidetoshi Kumagai
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yuichi Ikeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Yoko Iizuka
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Naoya Yahagi
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Hitoshi Shimano
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Jun-Ichi Osuga
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University
| | - Shun Ishibashi
- Division of Endocrinology and Metabolism, Department of Medicine, Jichi Medical University
| | - Takashi Kadowaki
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| | - Hiroaki Okazaki
- Departments of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
17
|
Tups A, Benzler J, Sergi D, Ladyman SR, Williams LM. Central Regulation of Glucose Homeostasis. Compr Physiol 2017; 7:741-764. [PMID: 28333388 DOI: 10.1002/cphy.c160015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
18
|
Chiang HC, Wang CH, Yeh SC, Lin YH, Kuo YT, Liao CW, Tsai FY, Lin WY, Chuang WH, Tsou TC. Comparative microarray analyses of mono(2-ethylhexyl)phthalate impacts on fat cell bioenergetics and adipokine network. Cell Biol Toxicol 2017; 33:511-526. [DOI: 10.1007/s10565-016-9380-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/29/2016] [Indexed: 10/20/2022]
|
19
|
Odle B, Dennison N, Al-Nakkash L, Broderick TL, Plochocki JH. Genistein treatment improves fracture resistance in obese diabetic mice. BMC Endocr Disord 2017; 17:1. [PMID: 28183304 PMCID: PMC5299772 DOI: 10.1186/s12902-016-0144-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 10/27/2016] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Obese, type two diabetics are at an increased risk for fracturing their limb bones in comparison to the general population. Phytoestrogens like as the soy isoflavone genistein have been shown to protect against bone loss. In this study, we tested the effects of genistein treatment on femurs of ob/ob mice, a model for obesity and type two diabetes mellitus. METHODS Twenty six-week-old female mice were divided into obese (ob/ob) control, obese genistein-treated, lean (ob/+) control, and lean genistein-treated groups (n = 5 each). Treatment with genistein consisted of 600 mg genistein/kg diet. Control mice were given standard rodent chow. At the end of a four-week treatment period, bone histomorphometric and three-point bending properties were compared among groups. RESULTS Obese mice had larger bone areas (B.Ar.; P < 0.05) and total areas (Tt.Ar.; P < 0.05), but similar bone volume (B.Ar./Tt.Ar.; P > 0.05) of the proximal femoral epiphysis in comparison to lean mice. Treatment with genistein decreased Tt.Ar. and femur length, and increased ultimate force required to fracture the femur and the maximum deformation to failure (P < 0.05). CONCLUSIONS Genistein improves resistance to fracture from bending loads.
Collapse
Affiliation(s)
- Britton Odle
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| | - Nathan Dennison
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| | - Layla Al-Nakkash
- Department of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| | - Tom L. Broderick
- Department of Physiology, Laboratory of Diabetes and Exercise Metabolism, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| | - Jeffrey H. Plochocki
- Department of Anatomy, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ 85308 USA
| |
Collapse
|
20
|
Genetic Manipulation with Viral Vectors to Assess Metabolism and Adipose Tissue Function. Methods Mol Biol 2017; 1566:109-124. [PMID: 28244045 DOI: 10.1007/978-1-4939-6820-6_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Viral vectors have become widely used tools for genetic manipulation of adipose tissues to understand the biology and function of adipocytes in metabolism. There are a number of different viral vectors commonly used: retrovirus, lentivirus, adenovirus, and adeno-associated virus (AAV). Here, we review examples from the literature and describe methods to transduce adipocytes and adipose tissues using retrovirus, lentivirus, adenovirus, and AAV to ascertain gene function in adipose biology.
Collapse
|
21
|
Talton OO, Pennington KA, Pollock KE, Bates K, Ma L, Ellersieck MR, Schulz LC. Maternal Hyperleptinemia Improves Offspring Insulin Sensitivity in Mice. Endocrinology 2016; 157:2636-48. [PMID: 27145007 DOI: 10.1210/en.2016-1039] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Maternal obesity and gestational diabetes are prevalent worldwide. Offspring of mothers with these conditions weigh more and are predisposed to metabolic syndrome. A hallmark of both conditions is maternal hyperleptinemia, but the role of elevated leptin levels during pregnancy on developmental programming is largely unknown. We previously found that offspring of hyperleptinemic mothers weighed less and had increased activity. The goal of this study was to determine whether maternal leptin affects offspring insulin sensitivity by investigating offspring glucose metabolism and lipid accumulation. Offspring from two maternal hyperleptinemic models were compared. The first model of hyperleptinemia is the Lepr(db/+) mouse, which has a mutation in one copy of the gene that encodes the leptin receptor, resulting in a truncated long form of the receptor, and hyperleptinemia. Wild-type females served as the control for the Lepr(db/+) females. For the second hyperleptinemic model, wild-type females were implanted with miniosmotic pumps, which released leptin (350 ng/h) or saline (as the control) just prior to mating and throughout gestation. In the offspring of these dams, we measured glucose tolerance; serum leptin, insulin, and triglyceride levels; liver triglycerides; pancreatic α- and β-cell numbers; body composition; incidence of nonalcoholic fatty liver disease; and the expression of key metabolic genes in the liver and adipose tissue. We found that the offspring of hyperleptinemic dams exhibited improved glucose tolerance, reduced insulin and leptin concentrations, reduced liver triglycerides, and a lower incidence of nonalcoholic fatty liver disease. Overall, maternal hyperleptinemia was beneficial for offspring glucose and lipid metabolism.
Collapse
Affiliation(s)
- Omonseigho O Talton
- Departments of Obstetrics, Gynecology, and Women's Health (O.O.T., K.A.P., K.E.P., K.B., L.C.S.) and Radiology (L.M.) and Divisions of Biological Sciences (O.O.T., K.B., L.C.S.) and Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65212; and Biomolecular Imaging Center (L.M.), Harry S. Truman Veterans Affairs Hospital, Columbia, Missouri 65201
| | - Kathleen A Pennington
- Departments of Obstetrics, Gynecology, and Women's Health (O.O.T., K.A.P., K.E.P., K.B., L.C.S.) and Radiology (L.M.) and Divisions of Biological Sciences (O.O.T., K.B., L.C.S.) and Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65212; and Biomolecular Imaging Center (L.M.), Harry S. Truman Veterans Affairs Hospital, Columbia, Missouri 65201
| | - Kelly E Pollock
- Departments of Obstetrics, Gynecology, and Women's Health (O.O.T., K.A.P., K.E.P., K.B., L.C.S.) and Radiology (L.M.) and Divisions of Biological Sciences (O.O.T., K.B., L.C.S.) and Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65212; and Biomolecular Imaging Center (L.M.), Harry S. Truman Veterans Affairs Hospital, Columbia, Missouri 65201
| | - Keenan Bates
- Departments of Obstetrics, Gynecology, and Women's Health (O.O.T., K.A.P., K.E.P., K.B., L.C.S.) and Radiology (L.M.) and Divisions of Biological Sciences (O.O.T., K.B., L.C.S.) and Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65212; and Biomolecular Imaging Center (L.M.), Harry S. Truman Veterans Affairs Hospital, Columbia, Missouri 65201
| | - Lixin Ma
- Departments of Obstetrics, Gynecology, and Women's Health (O.O.T., K.A.P., K.E.P., K.B., L.C.S.) and Radiology (L.M.) and Divisions of Biological Sciences (O.O.T., K.B., L.C.S.) and Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65212; and Biomolecular Imaging Center (L.M.), Harry S. Truman Veterans Affairs Hospital, Columbia, Missouri 65201
| | - Mark R Ellersieck
- Departments of Obstetrics, Gynecology, and Women's Health (O.O.T., K.A.P., K.E.P., K.B., L.C.S.) and Radiology (L.M.) and Divisions of Biological Sciences (O.O.T., K.B., L.C.S.) and Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65212; and Biomolecular Imaging Center (L.M.), Harry S. Truman Veterans Affairs Hospital, Columbia, Missouri 65201
| | - Laura C Schulz
- Departments of Obstetrics, Gynecology, and Women's Health (O.O.T., K.A.P., K.E.P., K.B., L.C.S.) and Radiology (L.M.) and Divisions of Biological Sciences (O.O.T., K.B., L.C.S.) and Animal Sciences (K.E.P., M.R.E., L.C.S.), University of Missouri, Columbia, Missouri 65212; and Biomolecular Imaging Center (L.M.), Harry S. Truman Veterans Affairs Hospital, Columbia, Missouri 65201
| |
Collapse
|
22
|
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
|
23
|
Michelin RM, Al-Nakkash L, Broderick TL, Plochocki JH. Genistein treatment increases bone mass in obese, hyperglycemic mice. Diabetes Metab Syndr Obes 2016; 9:63-70. [PMID: 27042131 PMCID: PMC4801201 DOI: 10.2147/dmso.s97600] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Obesity and type 2 diabetes mellitus are associated with elevated risk of limb bone fracture. Incidences of these conditions are on the rise worldwide. Genistein, a phytoestrogen, has been shown by several studies to demonstrate bone-protective properties and may improve bone health in obese type 2 diabetics. METHODS In this study, we test the effects of genistein treatment on limb bone and growth plate cartilage histomorphometry in obese, hyperglycemic ob/ob mice. Six-week-old ob/ob mice were divided into control and genistein-treated groups. Genistein-treated mice were fed a diet containing 600 mg genistein/kg for a period of 4 weeks. Cross-sectional geometric and histomorphometric analyses were conducted on tibias. RESULTS Genistein-treated mice remained obese and hyperglycemic. However, histomorphometric comparisons show that genistein-treated mice have greater tibial midshaft diameters and ratios of cortical bone to total tissue area than the controls. Genistein-treated mice also exhibit decreased growth plate thickness of the proximal tibia. CONCLUSION Our results indicate that genistein treatment affects bone of the tibial midshaft in the ob/ob mouse, independent of improvements in the hyperglycemic state and body weight.
Collapse
Affiliation(s)
- Richard M Michelin
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| | - Layla Al-Nakkash
- Department of Physiology, Midwestern University, Glendale, AZ, USA
| | - Tom L Broderick
- Laboratory of Diabetes and Exercise Metabolism, Department of Physiology, Midwestern University, Glendale, AZ, USA
| | - Jeffrey H Plochocki
- Department of Anatomy, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ, USA
| |
Collapse
|
24
|
Wang M, Wang C, Han RH, Han X. Novel advances in shotgun lipidomics for biology and medicine. Prog Lipid Res 2016; 61:83-108. [PMID: 26703190 PMCID: PMC4733395 DOI: 10.1016/j.plipres.2015.12.002] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 12/14/2022]
Abstract
The field of lipidomics, as coined in 2003, has made profound advances and been rapidly expanded. The mass spectrometry-based strategies of this analytical methodology-oriented research discipline for lipid analysis are largely fallen into three categories: direct infusion-based shotgun lipidomics, liquid chromatography-mass spectrometry-based platforms, and matrix-assisted laser desorption/ionization mass spectrometry-based approaches (particularly in imagining lipid distribution in tissues or cells). This review focuses on shotgun lipidomics. After briefly introducing its fundamentals, the major materials of this article cover its recent advances. These include the novel methods of lipid extraction, novel shotgun lipidomics strategies for identification and quantification of previously hardly accessible lipid classes and molecular species including isomers, and novel tools for processing and interpretation of lipidomics data. Representative applications of advanced shotgun lipidomics for biological and biomedical research are also presented in this review. We believe that with these novel advances in shotgun lipidomics, this approach for lipid analysis should become more comprehensive and high throughput, thereby greatly accelerating the lipidomics field to substantiate the aberrant lipid metabolism, signaling, trafficking, and homeostasis under pathological conditions and their underpinning biochemical mechanisms.
Collapse
Affiliation(s)
- Miao Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Chunyan Wang
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Rowland H Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA
| | - Xianlin Han
- Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute; Orlando, FL 32827, USA; College of Basic Medical Sciences, Zhejiang Chinese Medical University, 548 Bingwen Road, Hangzhou, Zhejiang 310053, China.
| |
Collapse
|
25
|
Papaetis GS, Papakyriakou P, Panagiotou TN. Central obesity, type 2 diabetes and insulin: exploring a pathway full of thorns. Arch Med Sci 2015; 11:463-82. [PMID: 26170839 PMCID: PMC4495144 DOI: 10.5114/aoms.2015.52350] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2013] [Revised: 06/20/2013] [Accepted: 07/04/2013] [Indexed: 12/19/2022] Open
Abstract
The prevalence of type 2 diabetes (T2D) is rapidly increasing. This is strongly related to the contemporary lifestyle changes that have resulted in increased rates of overweight individuals and obesity. Central (intra-abdominal) obesity is observed in the majority of patients with T2D. It is associated with insulin resistance, mainly at the level of skeletal muscle, adipose tissue and liver. The discovery of macrophage infiltration in the abdominal adipose tissue and the unbalanced production of adipocyte cytokines (adipokines) was an essential step towards novel research perspectives for a better understanding of the molecular mechanisms governing the development of insulin resistance. Furthermore, in an obese state, the increased cellular uptake of non-esterified fatty acids is exacerbated without any subsequent β-oxidation. This in turn contributes to the accumulation of intermediate lipid metabolites that cause defects in the insulin signaling pathway. This paper examines the possible cellular mechanisms that connect central obesity with defects in the insulin pathway. It discusses the discrepancies observed from studies organized in cell cultures, animal models and humans. Finally, it emphasizes the need for therapeutic strategies in order to achieve weight reduction in overweight and obese patients with T2D.
Collapse
Affiliation(s)
- Georgios S. Papaetis
- Diabetes Clinic, Paphos, Cyprus
- Diabetes Clinic, 3 Department of Medicine, University of Athens Medical School, ‘Sotiria’ General Hospital, Athens, Greece
| | | | - Themistoklis N. Panagiotou
- Diabetes Clinic, 3 Department of Medicine, University of Athens Medical School, ‘Sotiria’ General Hospital, Athens, Greece
| |
Collapse
|
26
|
Cooley J, Broderick TL, Al-Nakkash L, Plochocki JH. Effects of resveratrol treatment on bone and cartilage in obese diabetic mice. J Diabetes Metab Disord 2015; 14:10. [PMID: 25789256 PMCID: PMC4363196 DOI: 10.1186/s40200-015-0141-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 02/23/2015] [Indexed: 12/23/2022]
Abstract
Background Resveratrol is a polyphenolic phytoalexin that has been shown to exhibit osteoprotective and chondroprotective properties. We examine the effects of resveratrol treatment on bone and cartilage tissue of obese, diabetic ob/ob mice. Methods Eight-week-old ob/ob and lean control mice were given trans-resveratrol at an oral dose of 25 mg/kg for 3 weeks. Histomorphometric and cross-sectional-geometric variables were analyzed. Results Ob/ob mice in our study exhibit significantly reduced femoral length, resistance to loading, and tibial growth plate total area and calcified area than lean controls (P < 0.05). Resveratrol treatment significantly increased cortical area in both ob/ob and control mice, but did not improve cross-sectional indicators of resistance to bending. Resveratrol treatment also reduced tibial length and calcified growth plate cartilage area in comparison to untreated mice (P < 0.05). Conclusion Resveratrol treatment of ob/ob mice had mixed effects on bone histomorphometry at the femoral midshaft. Treatment increased cortical area but decreased bone length.
Collapse
Affiliation(s)
- Joseph Cooley
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| | - Tom L Broderick
- Departent of Physiology, Laboratory of Diabetes and Exercise Metabolism, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| | - Layla Al-Nakkash
- Departent of Physiology, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| | - Jeffrey H Plochocki
- Department of Anatomy, Arizona College of Osteopathic Medicine, Midwestern University, Glendale, AZ USA
| |
Collapse
|
27
|
O'Neill SM, Hinkle C, Chen SJ, Sandhu A, Hovhannisyan R, Stephan S, Lagor WR, Ahima RS, Johnston JC, Reilly MP. Targeting adipose tissue via systemic gene therapy. Gene Ther 2014; 21:653-61. [PMID: 24830434 PMCID: PMC4342115 DOI: 10.1038/gt.2014.38] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 02/18/2014] [Accepted: 03/18/2014] [Indexed: 02/06/2023]
Abstract
Adipose tissue has a critical role in energy and metabolic homeostasis, but it is challenging to adapt techniques to modulate adipose function in vivo. Here we develop an in vivo, systemic method of gene transfer specifically targeting adipose tissue using adeno-associated virus (AAV) vectors. We constructed AAV vectors containing cytomegalovirus promoter-regulated reporter genes, intravenously injected adult mice with vectors using multiple AAV serotypes, and determined that AAV2/8 best targeted adipose tissue. Altering vectors to contain adiponectin promoter/enhancer elements and liver-specific microRNA-122 target sites restricted reporter gene expression to adipose tissue. As proof of efficacy, the leptin gene was incorporated into the adipose-targeted expression vector, package into AAV2/8 and administered intravenously to 9- to 10-week-old ob/ob mice. Phenotypic changes were measured over an 8-week period. Leptin mRNA and protein were expressed in adipose and leptin protein was secreted into plasma. Mice responded with reversal of weight gain, decreased hyperinsulinemia and improved glucose tolerance. AAV2/8-mediated systemic delivery of an adipose-targeted expression vector can replace a gene lacking in adipose tissue and correct a mouse model of human disease, demonstrating experimental application and therapeutic potential in disorders of adipose.
Collapse
Affiliation(s)
- Sean M. O'Neill
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Christine Hinkle
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shu-Jen Chen
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arbansjit Sandhu
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ruben Hovhannisyan
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Stephen Stephan
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William R. Lagor
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rexford S. Ahima
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Julie C. Johnston
- Gene Therapy Program, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Muredach P. Reilly
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Correspondence should be addressed to Muredach P. Reilly Cardiovascular Institute Translational Research Center 3400 Civic Center Blvd, Bldg 421 11th floor, Room 11-136 Philadelphia, PA 19104 Tel: (215) 573-1214 Fax: (215) 746-7415
| |
Collapse
|
28
|
Neumann UH, Chen S, Tam YYC, Baker RK, Covey SD, Cullis PR, Kieffer TJ. IGFBP2 is neither sufficient nor necessary for the physiological actions of leptin on glucose homeostasis in male ob/ob mice. Endocrinology 2014; 155:716-25. [PMID: 24424049 DOI: 10.1210/en.2013-1622] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The ability of leptin to improve metabolic abnormalities in models of leptin deficiency, lipodystrophy, and even type 1 diabetes is of significant interest. However, the mechanism by which leptin mediates these effects remains ill-defined. Leptin was recently reported to regulate insulin-like growth factor-binding protein-2 (IGFBP2), and adenoviral overexpression of pharmacological levels of IGFBP2 ameliorates diabetic symptoms in many models of diabetes. We sought to determine the role of physiological levels of IGFBP2 in the glucoregulatory action of leptin. To investigate whether physiological levels of IGFBP2 are sufficient to mimic the action of leptin, we treated male ob/ob mice with low-dose IGFBP2 adenovirus (Ad-IGFBP2) or low-dose leptin. Despite similar levels of circulating IGFBP2, leptin but not Ad-IGFBP2 lowered body weight and plasma insulin and improved glucose and insulin tolerance. To elucidate the role of IGFBP2 in normal glucose homeostasis, we knocked down IGFBP2 in male C57BL/6 mice using small interfering RNA to determine whether this would recapitulate any aspect of the ob/ob phenotype. Despite successful IGFBP2 knockdown, body weight, blood glucose, and plasma insulin were unchanged. Finally, to determine whether IGFBP2 is required for the glucoregulatory actions of leptin, we prevented leptin-mediated increases in IGFBP2 in male ob/ob mice using RNA interference. Even though increases in IGFBP2 were blocked, the ability of leptin to decrease body weight, blood glucose, and plasma insulin levels were unaltered. In conclusion, physiological levels of IGFBP2 are neither sufficient to mimic nor required for the physiological action of leptin.
Collapse
Affiliation(s)
- Ursula H Neumann
- Departments of Cellular and Physiological Sciences (U.H.N., R.K.B., T.J.K.), Biochemistry and Molecular Biology (S.C., Y.Y.C.T., S.D.C., P.R.C.), and Surgery (T.J.K.), Life Sciences Institute, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | | | | | | | | | | | | |
Collapse
|
29
|
Koch CE, Lowe C, Pretz D, Steger J, Williams LM, Tups A. High-fat diet induces leptin resistance in leptin-deficient mice. J Neuroendocrinol 2014; 26:58-67. [PMID: 24382295 DOI: 10.1111/jne.12131] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 11/27/2013] [Accepted: 12/23/2013] [Indexed: 12/22/2022]
Abstract
The occurrence of type II diabetes is highly correlated with obesity, although the mechanisms linking the two conditions are incompletely understood. Leptin is a potent insulin sensitiser and, in leptin-deficient, insulin insensitive, Lep(ob/ob) mice, leptin improves glucose tolerance, indicating that leptin resistance may link obesity to insulin insensitivity. Leptin resistance occurs in response to a high-fat diet (HFD) and both hyperleptinaemia and inflammation have been proposed as causative mechanisms. Scrutinising the role of hyperleptinaemia in this process, central hyperleptinaemia in Lep(ob/ob) mice was induced by chronic i.c.v. infusion of leptin (4.2 μg/day) over 10 days. This treatment led to a dramatic decline in body weight and food intake, as well as an improvement in glucose tolerance. Transfer to HFD for 4 days markedly arrested the beneficial effects of leptin on these parameters. Because Lep(ob/ob) mice are exquisitely sensitive to leptin, the possibility that leptin could reverse HFD-induced glucose intolerance in these animals was investigated. HFD led to increased body weight and glucose intolerance compared to a low-fat diet (LFD). Older and heavier Lep(ob/ob) mice were used as body weight-matched controls. Mice in each group received either i.p. leptin (1.25 mg/kg) or vehicle, and glucose tolerance, food intake and the number of phosphorylated signal transducer and activator of transcription (pSTAT)3 immunoreactive cells in the arcuate nucleus (ARC) and ventromedial hypothalamus (VMH) were analysed. Leptin improved glucose tolerance (P = 0. 019) and reduced food intake in Lep(ob/ob) mice on LFD (P ≤ 0.001) but was ineffective in mice on HFD. Furthermore, when leptin was administered centrally, the glucose tolerance of Lep(ob/ob) mice on HFD was significantly impaired (P = 0.007). Although leptin induced the number of pSTAT3 immunoreactive cells in the ARC and VMH of Lep(ob/ob) mice on LFD, HFD was associated with elevated pSTAT3 immunoreactivity in vehicle-treated Lep(ob/ob) mice that was unaffected by leptin treatment, suggesting central leptin resistance. Negating central inflammation by co-administering a c-Jun n-terminal kinase (JNK) inhibitor reinstated the glucose-lowering effects of leptin. These findings demonstrate that Lep(ob/ob) mice develop leptin resistance on a HFD independent of hyperleptinaemia and also indicate that the JNK inflammatory pathway plays a key role in the induction of diet-induced glucose intolerance.
Collapse
Affiliation(s)
- C E Koch
- Department of Animal Physiology, Faculty of Biology, Philipps University Marburg, Marburg, Germany
| | | | | | | | | | | |
Collapse
|
30
|
von Schnurbein J, Heni M, Moss A, Nagel SA, Machann J, Muehleder H, Debatin KM, Farooqi S, Wabitsch M. Rapid improvement of hepatic steatosis after initiation of leptin substitution in a leptin-deficient girl. Horm Res Paediatr 2014; 79:310-7. [PMID: 23651953 DOI: 10.1159/000348541] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/01/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Leptin deficiency is associated with severe obesity and metabolic disturbances. Increased liver fat content has been reported in only one case beforehand, even though hepatic steatosis is a typical comorbidity of common obesity. It is also frequent in patients with lipodystrophy where it resolves under leptin therapy. SUBJECT AND METHODS In 2010, we reported a leptin-deficient patient with a novel homozygous mutation in the leptin gene and severe hepatic steatosis. We have now studied serum changes and changes in liver fat content during the substitution with recombinant methionyl human leptin. RESULTS After 23 weeks of leptin substitution, elevated transaminases, total cholesterol and low-density lipoprotein levels normalized. After 62 weeks, homeostasis model assessment of insulin resistance improved from 10.7 to 6.0 and body fat mass dropped from 50.2 to 37.8%. Liver fat content was drastically reduced from 49.7 to 9.4%. The first changes in liver fat content were detectable after 3 days of therapy. CONCLUSION Our patient showed a remarkable reduction of liver fat content during the treatment with recombinant methionyl human leptin. These changes occurred rapidly after initiation of the substitution, which implies that leptin has a direct effect on hepatic lipid metabolism in humans as it is seen in rodents.
Collapse
Affiliation(s)
- J von Schnurbein
- Division of Pediatric Endocrinology and Diabetes, University Medical Center Ulm, Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kappel V, van Noort B, Ritschel F, Seidel M, Ehrlich S. [Anorexia nervosa - from a neuroscience perspective]. ZEITSCHRIFT FUR KINDER-UND JUGENDPSYCHIATRIE UND PSYCHOTHERAPIE 2013; 42:39-48; quiz 49-50. [PMID: 24365962 DOI: 10.1024/1422-4917/a000268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Anorexia nervosa is a frequent disorder especially among adolescent girls and young women, with high morbidity, mortality, and relapse rates. To date, no single therapeutic approach has proved to be superior to others (Herpertz et al., 2011). It remains unclear how its etiology and pathology are encoded within cognitive, neural, and endocrinological processes that modulate important mechanisms in appetitive processing and weight regulation. Yet, several trait characteristics have been identified in AN which might reflect predisposing factors. Further, altered levels of neuropeptides and hormones that regulate appetite and feeding behavior have been found during both the acute and the recovered state, pointing to dysfunctional mechanisms in AN that persist even after malnutrition has ceased. Researchers are also hoping that brain imaging techniques will allow for a more detailed investigation of the neural basis of reward and punishment sensitivity that appears to be altered in AN. The integration and extension of recent findings in these areas will hopefully provide a more comprehensive understanding of the disorder and hence enable the development of more effective treatments.
Collapse
Affiliation(s)
- Viola Kappel
- Charité-Universitätsmedizin Berlin, Klinik für Psychiatrie, Psychosomatik und Psychotherapie des Kindes- und Jugendalters
| | - Betteke van Noort
- Charité-Universitätsmedizin Berlin, Klinik für Psychiatrie, Psychosomatik und Psychotherapie des Kindes- und Jugendalters
| | - Franziska Ritschel
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus Dresden, Klinik und Poliklinik für Kinder- und Jugendpsychiatrie und -psychotherapie
| | - Maria Seidel
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus Dresden, Klinik und Poliklinik für Kinder- und Jugendpsychiatrie und -psychotherapie
| | - Stefan Ehrlich
- Technische Universität Dresden, Universitätsklinikum Carl Gustav Carus Dresden, Klinik und Poliklinik für Kinder- und Jugendpsychiatrie und -psychotherapie Massachusetts General Hospital, Harvard Medical School, Athinoula A. Martinos Center for Biomedical Imaging, Psychiatric Neuroimaging Research Program
| |
Collapse
|
32
|
Amitani M, Asakawa A, Amitani H, Inui A. The role of leptin in the control of insulin-glucose axis. Front Neurosci 2013; 7:51. [PMID: 23579596 PMCID: PMC3619125 DOI: 10.3389/fnins.2013.00051] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Accepted: 03/18/2013] [Indexed: 12/21/2022] Open
Abstract
Obesity and diabetes mellitus are great public health concerns throughout the world because of their increasing incidence and prevalence. Leptin, the adipocyte hormone, is well known for its role in the regulation of food intake and energy expenditure. In addition to the regulation of appetite and satiety that recently has attracted much attentions, insight has also been gained into the critical role of leptin in the control of the insulin-glucose axis, peripheral glucose and insulin responsiveness. Since the discovery of leptin, leptin has been taken for its therapeutic potential to obesity and diabetes. Recently, the therapeutic effects of central leptin gene therapy have been reported in insulin-deficient diabetes in obesity animal models such as ob/ob mise, diet-induced obese mice, and insulin-deficient type 1 diabetes mice, and also in patients with inactivating mutations in the leptin gene. Herein, we review the role of leptin in regulating feeding behavior and glucose metabolism and also the therapeutic potential of leptin in obesity and diabetes mellitus.
Collapse
Affiliation(s)
- Marie Amitani
- Department of Psychosomatic Internal Medicine, Kagoshima University Graduate School of Medical and Dental Sciences Kagoshima, Japan
| | | | | | | |
Collapse
|
33
|
Paz-Filho G, Mastronardi C, Wong ML, Licinio J. Leptin therapy, insulin sensitivity, and glucose homeostasis. Indian J Endocrinol Metab 2012; 16:S549-S555. [PMID: 23565489 PMCID: PMC3602983 DOI: 10.4103/2230-8210.105571] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Glucose homeostasis is closely regulated not only by insulin, but also by leptin. Both hormones act centrally, regulating food intake and adiposity in humans. Leptin has several effects on the glucose-insulin homeostasis, some of which are independent of body weight and adiposity. Those effects of leptin are determined centrally in the hypothalamus and peripherally in the pancreas, muscles and liver. Leptin has beneficial effects on the glucose-insulin metabolism, by decreasing glycemia, insulinemia and insulin resistance. The understanding of the effects of leptin on the glucose-insulin homeostasis will lead to the development of leptin-based therapies against diabetes and other insulin resistance syndromes. In these review, we summarize the interactions between leptin and insulin, and their effects on the glucose metabolism.
Collapse
Affiliation(s)
- Gilberto Paz-Filho
- Department of Translational Medicine, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Claudio Mastronardi
- Department of Translational Medicine, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Ma-Li Wong
- Department of Translational Medicine, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| | - Julio Licinio
- Department of Translational Medicine, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia
| |
Collapse
|
34
|
Sáinz N, Rodríguez A, Catalán V, Becerril S, Ramírez B, Lancha A, Burgos-Ramos E, Gómez-Ambrosi J, Frühbeck G. Leptin reduces the expression and increases the phosphorylation of the negative regulators of GLUT4 traffic TBC1D1 and TBC1D4 in muscle of ob/ob mice. PLoS One 2012; 7:e29389. [PMID: 22253718 PMCID: PMC3253781 DOI: 10.1371/journal.pone.0029389] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 11/28/2011] [Indexed: 02/01/2023] Open
Abstract
Leptin improves insulin sensitivity in skeletal muscle. Our goal was to determine whether proteins controlling GLUT4 traffic are altered by leptin deficiency and in vivo leptin administration in skeletal muscle of wild type and ob/ob mice. Leptin-deficient ob/ob mice were divided in three groups: control, leptin-treated (1 mg/kg/d) and leptin pair-fed ob/ob mice. Microarray analysis revealed that 1,546 and 1,127 genes were regulated by leptin deficiency and leptin treatment, respectively. Among these, we identified 24 genes involved in intracellular vesicle-mediated transport in ob/ob mice. TBC1 domain family, member 1 (Tbc1d1), a negative regulator of GLUT4 translocation, was up-regulated (P = 0.001) in ob/ob mice as compared to wild types. Importantly, leptin treatment reduced the transcript levels of Tbc1d1 (P<0.001) and Tbc1d4 (P = 0.004) in the leptin-treated ob/ob as compared to pair-fed ob/ob animals. In addition, phosphorylation levels of TBC1D1 and TBC1D4 were enhanced in leptin-treated ob/ob as compared to control ob/ob (P = 0.015 and P = 0.023, respectively) and pair-fed ob/ob (P = 0.036 and P = 0.034, respectively) mice. Despite similar GLUT4 protein expression in wild type and ob/ob groups a different immunolocalization of this protein was evidenced in muscle sections. Leptin treatment increased GLUT4 immunoreactivity in gastrocnemius and extensor digitorum longus sections of leptin-treated ob/ob mice. Moreover, GLUT4 protein detected in immunoprecipitates from TBC1D4 was reduced by leptin replacement compared to control ob/ob (P = 0.013) and pair-fed ob/ob (P = 0.037) mice. Our findings suggest that leptin enhances the intracellular GLUT4 transport in skeletal muscle of ob/ob animals by reducing the expression and activity of the negative regulators of GLUT4 traffic TBC1D1 and TBC1D4.
Collapse
Affiliation(s)
- Neira Sáinz
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Andoni Lancha
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Emma Burgos-Ramos
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Universidad de Navarra, Pamplona, Spain
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Pamplona, Spain
- * E-mail:
| |
Collapse
|
35
|
Albarran-Zeckler RG, Sun Y, Smith RG. Physiological roles revealed by ghrelin and ghrelin receptor deficient mice. Peptides 2011; 32:2229-35. [PMID: 21781995 PMCID: PMC3221867 DOI: 10.1016/j.peptides.2011.07.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2011] [Revised: 05/13/2011] [Accepted: 07/05/2011] [Indexed: 02/04/2023]
Abstract
Ghrelin is a hormone made in the stomach and known primarily for its growth hormone releasing and orexigenic properties. Nevertheless, ghrelin through its receptor, the GHS-R1a, has been shown to exert many roles including regulation of glucose homeostasis, memory & learning, food addiction and neuroprotection. Furthermore, ghrelin could promote overall health and longevity by acting directly in the immune system and promoting an extended antigen repertoire. The development of mice lacking either ghrelin (ghrelin-/-) or its receptor (ghsr-/-) have provided a valuable tool for determining the relevance of ghrelin and its receptor in these multiple and diverse roles. In this review, we summarize the most important findings and lessons learned from the ghrelin-/- and ghsr-/- mice.
Collapse
Affiliation(s)
- Rosie G Albarran-Zeckler
- Department of Metabolism and Aging, Scripps Research Institute Florida, 130 Scripps Way B3B, Jupiter, FL 33458, United States.
| | | | | |
Collapse
|
36
|
Wang Y, Asakawa A, Inui A, Kosai KI. Leptin gene therapy in the fight against diabetes. Expert Opin Biol Ther 2011; 10:1405-14. [PMID: 20690892 DOI: 10.1517/14712598.2010.512286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
IMPORTANCE OF THE FIELD The incidence of diabetes is increasing worldwide, yet current treatments are not always effective for all patient or disease types. AREAS COVERED IN THIS REVIEW Here, we summarize the biologic and clinical roles of leptin in diabetes, and discuss candidate viral vectors that may be employed in the clinical use of central leptin gene therapy for diabetes. WHAT THE READER WILL GAIN We discuss how studies on leptin, a regulator of the insulin-glucose axis, have significantly advanced our understanding of the roles of energy homeostasis and insulin resistance in the pathogeneses of metabolic syndrome and diabetes. Recent studies have demonstrated the long-term therapeutic effects of central leptin gene therapy in obesity and diabetes via decreased insulin resistance and increased glucose metabolism. Many of these studies have employed viral vectors, which afford high in vivo gene transduction efficiencies compared with non-viral vectors. TAKE HOME MESSAGE Adeno-associated viral vectors are particularly well suited for central leptin gene therapy owing to their low toxicity and ability to drive transgene expression for extended periods.
Collapse
Affiliation(s)
- Yuqing Wang
- Kagoshima University Graduate School of Medical and Dental Sciences, Department of Gene Therapy and Regenerative Medicine, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | | | | | | |
Collapse
|
37
|
Leptin rapidly improves glucose homeostasis in obese mice by increasing hypothalamic insulin sensitivity. J Neurosci 2011; 30:16180-7. [PMID: 21123564 DOI: 10.1523/jneurosci.3202-10.2010] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Obesity is associated with resistance to the actions of both leptin and insulin via mechanisms that remain incompletely understood. To investigate whether leptin resistance per se contributes to insulin resistance and impaired glucose homeostasis, we investigated the effect of acute leptin administration on glucose homeostasis in normal as well as leptin- or leptin receptor-deficient mice. In hyperglycemic, leptin-deficient Lep(ob/ob) mice, leptin acutely and potently improved glucose metabolism, before any change of body fat mass, via a mechanism involving the p110α and β isoforms of phosphatidylinositol-3-kinase (PI3K). Unlike insulin, however, the anti-diabetic effect of leptin occurred independently of phospho-AKT, a major downstream target of PI3K, and instead involved enhanced sensitivity of the hypothalamus to insulin action upstream of PI3K, through modulation of IRS1 (insulin receptor substrate 1) phosphorylation. These data suggest that leptin resistance, as occurs in obesity, reduces the hypothalamic response to insulin and thereby impairs peripheral glucose homeostasis, contributing to the development of type 2 diabetes.
Collapse
|
38
|
Zheng C, Shinomiya T, Goldsmith CM, Di Pasquale G, Baum BJ. Convenient and reproducible in vivo gene transfer to mouse parotid glands. Oral Dis 2011; 17:77-82. [PMID: 20646229 PMCID: PMC3010376 DOI: 10.1111/j.1601-0825.2010.01707.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVES Published studies of gene transfer to mouse salivary glands have not employed the parotid glands. Parotid glands are the likely target tissue for most clinical applications of salivary gene transfer. The purpose of the present study was to develop a convenient and reproducible method of retroductal gene transfer to mouse parotid glands. METHODS The volume for vector delivery was assessed by infusion of Toluidine Blue into Stensen's ducts of Balb/c mice after direct intraoral cannulation. Recombinant, serotype 5 adenoviral vectors, encoding either firefly luciferase or human erythropoietin (hEpo), were constructed and then administered to parotid glands (10(7) vector particles/gland). Transgene expression in vivo was measured by enzyme activity (luciferase) or an enzyme-linked immunosorbent assay (hEpo). Vector biodistribution was measured by real-time quantitative (Q) PCR. RESULTS The chosen volume for mouse parotid vector delivery was 20μL. Little vector was detected outside of the targeted glands, with both QPCR and luciferase assays. Transgene expression was readily detected in glands (luciferase, hEpo), and serum and saliva (hEpo). Most secreted hEpo was detected in saliva. CONCLUSION These studies show that mouse parotid glands can be conveniently and reproducibly targeted for gene transfer, and should be useful for pre-clinical studies with many murine disease models.
Collapse
Affiliation(s)
- C Zheng
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892-1190, USA
| | | | | | | | | |
Collapse
|
39
|
Leptin administration downregulates the increased expression levels of genes related to oxidative stress and inflammation in the skeletal muscle of ob/ob mice. Mediators Inflamm 2010; 2010:784343. [PMID: 20671928 PMCID: PMC2910527 DOI: 10.1155/2010/784343] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2010] [Revised: 03/31/2010] [Accepted: 04/24/2010] [Indexed: 11/17/2022] Open
Abstract
Obese leptin-deficient ob/ob mice exhibit a low-grade chronic inflammation together with a low muscle mass. Our aim was to analyze the changes in muscle expression levels of genes related to oxidative stress and inflammatory responses in leptin deficiency and to identify the effect of in vivo leptin administration. Ob/ob mice were divided in three groups as follows: control ob/ob, leptin-treated ob/ob (1 mg/kg/d) and leptin pair-fed ob/ob mice. Gastrocnemius weight was lower in control ob/ob than in wild type mice (P < .01) exhibiting an increase after leptin treatment compared to control and pair-fed (P < .01) ob/ob animals. Thiobarbituric acid reactive substances, markers of oxidative stress, were higher in serum (P < .01) and gastrocnemius (P = .05) of control ob/ob than in wild type mice and were significantly decreased (P < .01) by leptin treatment. Leptin deficiency altered the expression of 1,546 genes, while leptin treatment modified the regulation of 1,127 genes with 86 of them being involved in oxidative stress, immune defense and inflammatory response. Leptin administration decreased the high expression of Crybb1, Hspb3, Hspb7, Mt4, Cat, Rbm9, Serpinc1 and Serpinb1a observed in control ob/ob mice, indicating that it improves inflammation and muscle loss.
Collapse
|
40
|
Transcription factor AP-2beta inhibits expression and secretion of leptin, an insulin-sensitizing hormone, in 3T3-L1 adipocytes. Int J Obes (Lond) 2010; 34:670-8. [PMID: 20065963 DOI: 10.1038/ijo.2009.295] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND We have previously reported an association between the activator protein-2beta (AP-2beta) transcription factor gene and type 2 diabetes. This gene is preferentially expressed in adipose tissue, and subjects with a disease-susceptible allele of AP-2beta showed stronger AP-2beta expression in adipose tissue than those without the susceptible allele. Furthermore, overexpression of AP-2beta led to lipid accumulation and induced insulin resistance in 3T3-L1 adipocytes. RESULT We found that overexpression of AP-2beta in 3T3-L1 adipocytes decreased the promoter activity of leptin, and subsequently decreased both messenger RNA (mRNA) and protein expression and secretion. Furthermore, knockdown of endogenous AP-2beta by RNA-interference increased mRNA and protein expression of leptin. Electrophoretic mobility shift and chromatin immunoprecipitation assays revealed specific binding of AP-2beta to leptin promoter regions in vitro and in vivo. In addition, site-directed mutagenesis of the AP-2-binding site located between position +34 and +42 relative to the transcription start site abolished the inhibitory effect of AP-2beta. Our results clearly showed that AP-2beta directly inhibited insulin-sensitizing hormone leptin expression by binding to its promoter. CONCLUSION AP-2beta modulated the expression of leptin through direct interaction with its promoter region.
Collapse
|
41
|
Kim IS, Lee J, Lee JS, Shin DY, Kim MJ, Lee MK. Effect of Fermented Yacon (Smallanthus Sonchifolius) Leaves Tea on Blood Glucose Levels and Glucose Metabolism in High-Fat Diet and Streptozotocin-Induced Type 2 Diabetic Mice. ACTA ACUST UNITED AC 2010. [DOI: 10.4163/kjn.2010.43.4.333] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- In-Sook Kim
- Department of Nutrition Education, Graduate School of Education, Sunchon National University, Suncheon 540-742, Korea
| | - Jin Lee
- Department of Food and Nutrition, Sunchon National University, Suncheon 540-742, Korea
| | - Jeom-Sook Lee
- Department of Food and Nutrition, Sunchon National University, Suncheon 540-742, Korea
| | - Dong-Young Shin
- Department of Development in Resource Plants, Sunchon National University, Suncheon 540-742, Korea
| | - Myung-Joo Kim
- Faculty of Hotel Cuisine, Daegu Polytechnic College, Daegu 706-022, Korea
| | - Mi-Kyung Lee
- Department of Food and Nutrition, Sunchon National University, Suncheon 540-742, Korea
| |
Collapse
|
42
|
Hedbacker K, Birsoy K, Wysocki RW, Asilmaz E, Ahima RS, Farooqi IS, Friedman JM. Antidiabetic effects of IGFBP2, a leptin-regulated gene. Cell Metab 2010; 11:11-22. [PMID: 20074524 DOI: 10.1016/j.cmet.2009.11.007] [Citation(s) in RCA: 221] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 07/02/2009] [Accepted: 11/30/2009] [Indexed: 12/16/2022]
Abstract
We tested whether leptin can ameliorate diabetes independent of weight loss by defining the lowest dose at which leptin treatment of ob/ob mice reduces plasma glucose and insulin concentration. We found that a leptin dose of 12.5 ng/hr significantly lowers blood glucose and that 25 ng/hr of leptin normalizes plasma glucose and insulin without significantly reducing body weight, establishing that leptin exerts its most potent effects on glucose metabolism. To find possible mediators of this effect, we profiled liver mRNA using microarrays and identified IGF Binding Protein 2 (IGFBP2) as being regulated by leptin with a similarly high potency. Overexpression of IGFBP2 by an adenovirus reversed diabetes in insulin-resistant ob/ob, Ay/a, and diet-induced obese mice, as well as insulin-deficient streptozotocin-treated mice. Hyperinsulinemic clamp studies showed a 3-fold improvement in hepatic insulin sensitivity following IGFBP2 treatment of ob/ob mice. These results show that IGFBP2 can regulate glucose metabolism, a finding with potential implications for the pathogenesis and treatment of diabetes.
Collapse
|
43
|
Kobayashi S, Fukuhara A, Taguchi T, Matsuda M, Tochino Y, Otsuki M, Shimomura I. Identification of a new secretory factor, CCDC3/Favine, in adipocytes and endothelial cells. Biochem Biophys Res Commun 2009; 392:29-35. [PMID: 20043878 DOI: 10.1016/j.bbrc.2009.12.142] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 12/23/2009] [Indexed: 01/15/2023]
Abstract
The vascular system secretes many bioactive factors. In a gene chip database, we searched for novel genes with signal sequences that are specifically expressed in murine aorta, and focused on one gene previously named CCDC3 (NCBI nucleotide entry NM_028804), and we designated as Favine (fat/vessel-derived secretory protein). Northern blot analysis revealed that CCDC3 was expressed abundantly in the aorta and adipose tissues. The mRNA levels of CCDC3 were higher in adipose tissues of obese db/db mice than control mice, and induced during differentiation of rat primary adipocytes. In differentiated adipocytes, CCDC3 mRNA expression was enhanced by insulin and pioglitazone, a PPARgamma agonist, and suppressed by TNF-alpha, isoproterenol and norepinephrine. Transient expression experiments followed by N-terminal amino acid sequence analysis revealed secretion of CCDC3 protein into the culture medium, which was dose-dependently reduced by brefeldin A, an inhibitor of Golgi-mediated secretory pathway. When expressed in COS-7 cells, CCDC3 protein was post-transcriptionally modified with N-glycosylation, and formed a dimer complex. These results indicate that CCDC3 is a protein secreted by adipocytes and endothelial cells, and that its level is regulated both hormonally and nutritionally.
Collapse
Affiliation(s)
- Sachiko Kobayashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
44
|
Kalra SP. Central leptin gene therapy ameliorates diabetes type 1 and 2 through two independent hypothalamic relays; a benefit beyond weight and appetite regulation. Peptides 2009; 30:1957-63. [PMID: 19647774 PMCID: PMC2755606 DOI: 10.1016/j.peptides.2009.07.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 07/22/2009] [Accepted: 07/23/2009] [Indexed: 01/12/2023]
Abstract
Although its role in energy homeostasis is firmly established, the evidence accumulated over a decade linking the adipocyte leptin-hypothalamus axis in the pathogenesis of diabetes mellitus has received little attention in the contemporary thinking. In this context various lines of evidence are collated here to show that (1) under the direction of leptin two independent relays emanating from the hypothalamus restrain insulin secretion from the pancreas and mobilize peripheral organs--liver, skeletal muscle and brown adipose tissue--to upregulate glucose disposal, and (2), leptin insufficiency in the hypothalamus produced by either leptinopenia or restriction of leptin transport across the blood brain barrier due to hyperleptinemia of obesity and aging, initiate antecedent pathophysiological sequalae of diabetes type 1 and 2. Further, we document here the efficacy of leptin replenishment in vivo, especially by supplying it to the hypothalamus with the aid of gene therapy, in preventing the antecedent pathophysiological sequalae--hyperinsulinemia, insulin resistance and hyperglycemia--in various animal models and clinical paradigms of diabetes type 1 and 2 with or without attendant obesity. Overall, the new insights on the long-lasting antidiabetic potential of two independent hypothalamic relays engendered by central leptin gene therapy and the preclinical safety indicators in rodents warrant further validation in subhuman primates and humans.
Collapse
Affiliation(s)
- Satya P Kalra
- Department of Neuroscience, McKnight Brain Institute, College of Medicine, University of Florida, PO Box 100244, Gainesville, FL 32610-0244, United States.
| |
Collapse
|
45
|
Sáinz N, Rodríguez A, Catalán V, Becerril S, Ramírez B, Gómez-Ambrosi J, Frühbeck G. Leptin administration favors muscle mass accretion by decreasing FoxO3a and increasing PGC-1alpha in ob/ob mice. PLoS One 2009; 4:e6808. [PMID: 19730740 PMCID: PMC2733298 DOI: 10.1371/journal.pone.0006808] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Accepted: 07/31/2009] [Indexed: 12/26/2022] Open
Abstract
Absence of leptin has been associated with reduced skeletal muscle mass in leptin-deficient ob/ob mice. The aim of our study was to examine the effect of leptin on the catabolic and anabolic pathways regulating muscle mass. Gastrocnemius, extensor digitorum longus and soleus muscle mass as well as fiber size were significantly lower in ob/ob mice compared to wild type littermates, being significantly increased by leptin administration (P<0.001). This effect was associated with an inactivation of the muscle atrophy-related transcription factor forkhead box class O3 (FoxO3a) (P<0.05), and with a decrease in the protein expression levels of the E3 ubiquitin-ligases muscle atrophy F-box (MAFbx) (P<0.05) and muscle RING finger 1 (MuRF1) (P<0.05). Moreover, leptin increased (P<0.01) protein expression levels of peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), a regulator of muscle fiber type, and decreased (P<0.05) myostatin protein, a negative regulator of muscle growth. Leptin administration also activated (P<0.01) the regulators of cell cycle progression proliferating cell nuclear antigen (PCNA) and cyclin D1, and increased (P<0.01) myofibrillar protein troponin T. The present study provides evidence that leptin treatment may increase muscle mass of ob/ob mice by inhibiting myofibrillar protein degradation as well as enhancing muscle cell proliferation.
Collapse
Affiliation(s)
- Neira Sáinz
- Metabolic Research Laboratory, University of Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, University of Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, University of Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, University of Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, University of Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, University of Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, University of Navarra, Pamplona, Spain
- Department of Endocrinology, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
46
|
Kusakabe T, Tanioka H, Ebihara K, Hirata M, Miyamoto L, Miyanaga F, Hige H, Aotani D, Fujisawa T, Masuzaki H, Hosoda K, Nakao K. Beneficial effects of leptin on glycaemic and lipid control in a mouse model of type 2 diabetes with increased adiposity induced by streptozotocin and a high-fat diet. Diabetologia 2009; 52:675-83. [PMID: 19169663 DOI: 10.1007/s00125-009-1258-2] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Accepted: 12/18/2008] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS We have previously demonstrated the therapeutic usefulness of leptin in lipoatrophic diabetes and insulin-deficient diabetes in mouse models and could also demonstrate its dramatic effects on lipoatrophic diabetes in humans. The aim of the present study was to explore the therapeutic usefulness of leptin in a mouse model of type 2 diabetes with increased adiposity. METHODS To generate a mouse model mimicking human type 2 diabetes with increased adiposity, we used a combination of low-dose streptozotocin (STZ, 120 microg/g body weight) and high-fat diet (HFD, 45% of energy as fat). Recombinant mouse leptin was infused chronically (20 ng [g body weight](-1) h(-1)) for 14 days using a mini-osmotic pump. The effects of leptin on food intake, body weight, metabolic variables, tissue triacylglycerol content and AMP-activated protein kinase (AMPK) activity were examined. RESULTS Low-dose STZ injection led to a substantial reduction of plasma insulin levels and hyperglycaemia. Subsequent HFD feeding increased adiposity and induced insulin resistance and further augmentation of hyperglycaemia. In this model mouse mimicking human type 2 diabetes (STZ/HFD), continuous leptin infusion reduced food intake and body weight and improved glucose and lipid metabolism with enhancement of insulin sensitivity. Leptin also decreased liver and skeletal muscle triacylglycerol content accompanied by an increase of alpha2 AMPK activity in skeletal muscle. Pair-feeding experiments demonstrated that leptin improved glucose and lipid metabolism independently of the food intake reduction. CONCLUSIONS/INTERPRETATION This study demonstrates the beneficial effects of leptin on glycaemic and lipid control in a mouse model of type 2 diabetes with increased adiposity, indicating the possible clinical usefulness of leptin as a new glucose-lowering drug in humans.
Collapse
Affiliation(s)
- T Kusakabe
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lehrke M, Broedl UC, Biller-Friedmann IM, Vogeser M, Henschel V, Nassau K, Göke B, Kilger E, Parhofer KG. Serum concentrations of cortisol, interleukin 6, leptin and adiponectin predict stress induced insulin resistance in acute inflammatory reactions. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2008; 12:R157. [PMID: 19087258 PMCID: PMC2646322 DOI: 10.1186/cc7152] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 11/08/2008] [Accepted: 12/17/2008] [Indexed: 01/04/2023]
Abstract
Introduction Inflammatory stimuli are causative for insulin resistance in obesity as well as in acute inflammatory reactions. Ongoing research has identified a variety of secreted proteins that are released from immune cells and adipocytes as mediators of insulin resistance; however, knowledge about their relevance for acute inflammatory insulin resistance remains limited. In this study we aimed for a clarification of the relevance of different insulin resistance mediating factors in an acute inflammatory situation. Methods Insulin resistance was measured in a cohort of 37 non-diabetic patients undergoing cardiac surgery by assessment of insulin requirement to maintain euglycaemia and repeated measurements of an insulin glycaemic index. The kinetics of cortisol, interleukin 6 (IL6), tumour necrosis factor α (TNFα), resistin, leptin and adiponectin were assessed by repeated measurements in a period of 48 h. Results Insulin resistance increased during the observation period and peaked 22 h after the beginning of the operation. IL6 and TNFα displayed an early increase with peak concentrations at the 4-h time point. Serum levels of cortisol, resistin and leptin increased more slowly and peaked at the 22-h time point, while adiponectin declined, reaching a base at the 22-h time point. Model assessment identified cortisol as the best predictor of insulin resistance, followed by IL6, leptin and adiponectin. No additional information was gained by modelling for TNFα, resistin, catecholamine infusion rate, sex, age, body mass index (BMI), operation time or medication. Conclusions Serum cortisol levels are the best predictor for inflammatory insulin resistance followed by IL6, leptin and adiponectin. TNFα, and resistin have minor relevance as predictors of stress dependent insulin resistance.
Collapse
Affiliation(s)
- Michael Lehrke
- Department of Internal Medicine II, University of Munich, Grosshadern Campus, Marchioninistr 15, 81377 Munich, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Paz-Filho G, Esposito K, Hurwitz B, Sharma A, Dong C, Andreev V, Delibasi T, Erol H, Ayala A, Wong ML, Licinio J. Changes in insulin sensitivity during leptin replacement therapy in leptin-deficient patients. Am J Physiol Endocrinol Metab 2008; 295:E1401-8. [PMID: 18854428 PMCID: PMC2652497 DOI: 10.1152/ajpendo.90450.2008] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Leptin replacement rescues the phenotype of morbid obesity and hypogonadism in leptin-deficient adults. However, leptin's effects on insulin resistance are not well understood. Our objective was to evaluate the effects of leptin on insulin resistance. Three leptin-deficient adults (male, 32 yr old, BMI 23.5 kg/m(2); female, 42 yr old, BMI 25.1 kg/m(2); female, 46 yr old, BMI 31.7 kg/m(2)) with a missense mutation of the leptin gene were evaluated during treatment with recombinant methionyl human leptin (r-metHuLeptin). Insulin resistance was determined by euglycemic hyperinsulinemic clamps and by oral glucose tolerance tests (OGTTs), whereas patients were on r-metHuLeptin and after treatment was interrupted for 2-4 wk in the 4th, 5th, and 6th years of treatment. At baseline, all patients had normal insulin levels, C-peptide, and homeostatic model assessment of insulin resistance index, except for one female diagnosed with type 2 diabetes. The glucose infusion rate was significantly lower with r-metHuLeptin (12.03 +/- 3.27 vs. 8.16 +/- 2.77 mg.kg(-1).min(-1), P = 0.0016) but did not differ in the 4th, 5th, and 6th years of treatment when all results were analyzed by a mixed model [F(1,4) = 0.57 and P = 0.5951]. The female patient with type 2 diabetes became euglycemic after treatment with r-metHuLeptin and subsequent weight loss. The OGTT suggested that two patients showed decreased insulin resistance while off treatment. During an off-leptin OGTT, one of the patients developed a moderate hypoglycemic reaction attributed to increased posthepatic insulin delivery and sensitivity. We conclude that, in leptin-deficient adults, the interruption of r-metHuLeptin decreases insulin resistance in the context of rapid weight gain. Our results suggest that hyperleptinemia may contribute to mediate the increased insulin resistance of obesity.
Collapse
Affiliation(s)
- Gilberto Paz-Filho
- Dept. of Psychiatry & Behavioral Sciences, Univ. of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Dube JJ, Bhatt BA, Dedousis N, Bonen A, O'Doherty RM. Leptin, skeletal muscle lipids, and lipid-induced insulin resistance. Am J Physiol Regul Integr Comp Physiol 2007; 293:R642-50. [PMID: 17491114 DOI: 10.1152/ajpregu.00133.2007] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Leptin-induced increases in insulin sensitivity are well established and may be related to the effects of leptin on lipid metabolism. However, the effects of leptin on the levels of lipid metabolites implicated in pathogenesis of insulin resistance and the effects of leptin on lipid-induced insulin resistance are unknown. The current study addressed in rats the effects of hyperleptinemia (HL) on insulin action and markers of skeletal muscle (SkM) lipid metabolism in the absence or presence of acute hyperlipidemia induced by an infusion of a lipid emulsion. Compared with controls (CONT), HL increased insulin sensitivity, as assessed by hyperinsulinemic-euglycemic clamp ( approximately 15%), and increased SkM Akt ( approximately 30%) and glycogen synthase kinase 3 alpha ( approximately 52%) phosphorylation. These improvements in insulin action were associated with decreased SkM triglycerides (TG; approximately 61%), elevated ceramides ( approximately 50%), and similar diacylglycerol (DAG) levels in HL compared with CONT. Acute hyperlipidemia in CONT decreased insulin sensitivity ( approximately 25%) and increased SkM DAG ( approximately 33%) and ceramide ( approximately 60%) levels. However, hyperlipidemia did not induce insulin resistance or SkM DAG and ceramide accumulation in HL. SkM total fatty acid transporter CD36, plasma membrane fatty acid binding protein, acetyl Co-A carboxylase phosphorylation, and fatty acid oxidation were similar in HL compared with CONT. However, HL decreased SkM protein kinase C theta (PKC theta), a kinase implicated in mediating the detrimental effects of lipids on insulin action. We conclude that increases in insulin sensitivity induced by HL are associated with decreased levels of SkM TG and PKC theta and increased SkM insulin signaling, but not with decreases in other lipid metabolites implicated in altering SkM insulin sensitivity (DAG and ceramide). Furthermore, insulin resistance induced by an acute lipid infusion is prevented by HL.
Collapse
Affiliation(s)
- John J Dube
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
50
|
Sell H, Dietze-Schroeder D, Eckel J. The adipocyte-myocyte axis in insulin resistance. Trends Endocrinol Metab 2006; 17:416-22. [PMID: 17084639 DOI: 10.1016/j.tem.2006.10.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2006] [Revised: 10/13/2006] [Accepted: 10/24/2006] [Indexed: 12/16/2022]
Abstract
Insulin resistance in skeletal muscle is linked to an elevated adipose tissue mass, as is found in obesity, but can also be observed in lipodystrophy, in which adipose tissue is greatly reduced. Adipose tissue releases endocrine and metabolic mediators and is actively involved in crosstalk with skeletal muscle, a process that precedes and underlies the development of insulin resistance in muscles. Adipokines including tumor necrosis factor alpha, interleukin-6, leptin and adiponectin influence insulin signaling in skeletal muscle. Free fatty acids, their metabolites and ectopic fat in muscle also contribute to insulin resistance. Recent research indicates inflammation, endoplasmic reticulum stress and oxidative stress could be underlying mechanisms at the center of the development of insulin resistance. Insights into the role of macrophages in adipose tissue add to the complicated interplay between adipose tissue and skeletal muscle.
Collapse
Affiliation(s)
- Henrike Sell
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center, Düsseldorf, Germany
| | | | | |
Collapse
|