201
|
A Leptin Analog Locally Produced in the Brain Acts via a Conserved Neural Circuit to Modulate Obesity-Linked Behaviors in Drosophila. Cell Metab 2017; 25:208-217. [PMID: 28076762 PMCID: PMC5235317 DOI: 10.1016/j.cmet.2016.12.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/30/2016] [Accepted: 12/21/2016] [Indexed: 11/21/2022]
Abstract
Leptin, a typically adipose-derived "satiety hormone," has a well-established role in weight regulation. Here we describe a functionally conserved model of genetically induced obesity in Drosophila by manipulating the fly leptin analog unpaired 1 (upd1). Unexpectedly, cell-type-specific knockdown reveals upd1 in the brain, not the adipose tissue, mediates obesity-related traits. Disrupting brain-derived upd1 in flies leads to all the hallmarks of mammalian obesity: increased attraction to food cues, increased food intake, and increased weight. These effects are mediated by domeless receptors on neurons expressing Drosophila neuropeptide F, the orexigenic mammalian neuropeptide Y homolog. In vivo two-photon imaging reveals upd1 and domeless inhibit this hedonic signal in fed animals. Manipulations along this central circuit also create hypersensitivity to obesogenic conditions, emphasizing the critical interplay between biological predisposition and environment in overweight and obesity prevalence. We propose adipose- and brain-derived upd/leptin may control differing features of weight regulation through distinct neural circuits.
Collapse
|
202
|
Comparative Study of Reproductive Development in Wild and Captive-Reared Greater Amberjack Seriola dumerili (Risso, 1810). PLoS One 2017; 12:e0169645. [PMID: 28056063 PMCID: PMC5215828 DOI: 10.1371/journal.pone.0169645] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/20/2016] [Indexed: 01/06/2023] Open
Abstract
The greater amberjack Seriola dumerili is a large teleost fish with rapid growth and excellent flesh quality, whose domestication represents an ambitious challenge for aquaculture. The occurrence of reproductive dysfunctions in greater amberjack reared in captivity was investigated by comparing reproductive development of wild and captive-reared individuals. Wild and captive-reared breeders were sampled in the Mediterranean Sea during three different phases of the reproductive cycle: early gametogenesis (EARLY, late April-early May), advanced gametogenesis (ADVANCED, late May-early June) and spawning (SPAWNING, late June-July). Fish reproductive state was evaluated using the gonado-somatic index (GSI), histological analysis of the gonads and determination of sex steroid levels in the plasma, and correlated with leptin expression in the liver and gonad biochemical composition. The GSI and sex steroid levels were lower in captive-reared than in wild fish. During the ADVANCED period, when the wild greater amberjack breeders were already in spawning condition, ovaries of captive-reared breeders showed extensive atresia of late vitellogenic oocytes and spermatogenic activity ceased in the testes of half of the examined males. During the SPAWNING period, all captive-reared fish had regressed gonads, while wild breeders still displayed reproductive activity. Liver leptin expression and gonad proximate composition of wild and captive greater amberjack were similar. However, the gonads of captive-reared fish showed different total polar lipid contents, as well as specific lipid classes and fatty acid profiles with respect to wild individuals. This study underlines the need for an improvement in rearing technology for this species, which should include minimum handling during the reproductive season and the formulation of a specific diet to overcome the observed gonadal decrements of phospholipids, DHA (22:6n-3) and ARA (20:4n-6), compared to wild breeders.
Collapse
|
203
|
Hart RA, Dobos RC, Agnew LL, Smart NA, McFarlane JR. Leptin pharmacokinetics in male mice. Endocr Connect 2017; 6:20-26. [PMID: 27998953 PMCID: PMC5302164 DOI: 10.1530/ec-16-0089] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 12/19/2016] [Indexed: 11/09/2022]
Abstract
Pharmacokinetics of leptin in mammals has not been studied in detail and only one study has examined more than one time point in non-mutant mice and this was in a female mice. This is the first study to describe leptin distribution over a detailed time course in normal male mice. A physiologic dose (12 ng) of radiolabelled leptin was injected into adult male mice via the lateral tail vein and tissues were dissected out and measured for radioactivity over a time course of up to two hours. Major targets were the digestive tract, kidneys, skin and lungs. The brain was not a major target, and 0.15% of the total dose was recovered from the brain 5 min after administration. Major differences appear to exist in the distribution of leptin between the male and female mice, indicating a high degree of sexual dimorphism. Although the half-lives were similar between male and female mice, almost twice the proportion of leptin was recovered from the digestive tract of male mice in comparison to that reported previously for females. This would seem to indicate a major difference in leptin distribution and possibly function between males and females.
Collapse
Affiliation(s)
- Robert A Hart
- Centre for Bioactive Discovery in Health and AgeingUniversity of New England, Armidale, New South Wales, Australia
| | - Robin C Dobos
- NSW Department of Primary IndustriesArmidale, New South Wales, Australia
| | - Linda L Agnew
- Centre for Bioactive Discovery in Health and AgeingUniversity of New England, Armidale, New South Wales, Australia
| | - Neil A Smart
- Centre for Bioactive Discovery in Health and AgeingUniversity of New England, Armidale, New South Wales, Australia
| | - James R McFarlane
- Centre for Bioactive Discovery in Health and AgeingUniversity of New England, Armidale, New South Wales, Australia
| |
Collapse
|
204
|
Gomez JA, Rutkowski DT. Experimental reconstitution of chronic ER stress in the liver reveals feedback suppression of BiP mRNA expression. eLife 2016; 5. [PMID: 27938665 PMCID: PMC5179193 DOI: 10.7554/elife.20390] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/09/2016] [Indexed: 12/13/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is implicated in many chronic diseases, but very little is known about how the unfolded protein response (UPR) responds to persistent ER stress in vivo. Here, we experimentally reconstituted chronic ER stress in the mouse liver, using repeated injection of a low dose of the ER stressor tunicamycin. Paradoxically, this treatment led to feedback-mediated suppression of a select group of mRNAs, including those encoding the ER chaperones BiP and GRP94. This suppression was due to both silencing of the ATF6α pathway of UPR-dependent transcription and enhancement of mRNA degradation, possibly via regulated IRE1-dependent decay (RIDD). The suppression of mRNA encoding BiP was phenocopied by ectopic overexpression of BiP protein, and was also observed in obese mice. Our findings suggest that persistent cycles of UPR activation and deactivation create an altered, quasi-stable setpoint for UPR-dependent transcriptional regulation—an outcome that could be relevant to conditions such as metabolic syndrome. DOI:http://dx.doi.org/10.7554/eLife.20390.001 Toxic chemicals, extreme temperatures and other abnormal environmental conditions can cause the cells in our bodies to become stressed. Several kinds of stresses overwhelm a compartment in the cell called the endoplasmic reticulum, which is critical for processing new proteins so that they can work correctly. Endoplasmic reticulum stress has been linked to long-term diseases such as diabetes, cancer and neurodegenerative diseases. Most of what is known about how cells sense and respond to endoplasmic reticulum stress comes from studies on isolated cells that were subjected to harsh conditions that cells cannot tolerate for longer than a day or two. By contrast, little is known about how cells within whole organisms respond to milder but longer-lasting endoplasmic reticulum stress, which is closer to what occurs during disease. To investigate this issue, Gomez and Rutkowski treated mice repeatedly with a chemical that causes mild endoplasmic reticulum stress in the liver. The cells exposed to this persistent stress responded differently to those exposed to severe short-term stress. Whereas short-term stress causes liver cells to turn on genes that help the endoplasmic reticulum to process proteins more efficiently, long-term stress causes cells to turn off some of those genes. Further investigation revealed that cells in the livers of obese mice show similar patterns of gene activity as cells exposed to long-term endoplasmic reticulum stress. The findings presented by Gomez and Rutkowski could therefore also help us to understand more about the liver problems that often occur during obesity and diabetes. Further studies are now needed to examine exactly how long-lasting stress can shut off the cells’ protective mechanisms. Future experiments could also investigate whether other types of cells and organs respond to long-term endoplasmic reticulum stress in the same way as cells in the liver. DOI:http://dx.doi.org/10.7554/eLife.20390.002
Collapse
Affiliation(s)
- Javier A Gomez
- Graduate Program in Molecular and Cellular Biology, University of Iowa Carver College of Medicine, Iowa City, United States
| | - D Thomas Rutkowski
- Departments of Anatomy and Cell Biology and Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, United States
| |
Collapse
|
205
|
Cleary MP, Juneja SC, Phillips FC, Hu X, Grande JP, Maihle NJ. Leptin Receptor-Deficient MMTV-TGF-α/Leprdb Leprdb Female Mice Do Not Develop Oncogene-Induced Mammary Tumors. Exp Biol Med (Maywood) 2016; 229:182-93. [PMID: 14734797 DOI: 10.1177/153537020422900207] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Being overweight is a risk factor for postmenopausal breast cancer and is associated with an increased incidence and shortened latency of spontaneous and chemically Induced mammary tumors in rodents. However, leptin-deficient obese Lepob Lepob female mice have reduced incidences of spontaneous and oncogene-induced mammary tumors. Of interest, leptin enhances the proliferation of human breast cancer cell lines in which leptin receptors are expressed, which suggests that leptin signaling plays a role in tumor development. We evaluated oncogene-induced mammary tumor development in obese MMTV-TGF-α/Leprdb Leprdb mice that exhibit a defect in OB-Rb, which is considered to be the major signaling isoform of the leptin receptor. Lepr and MMTV-TGF-α mice were crossed, and the offspring were genotyped for oncogene expression and the determination of Lepr status. Lean MMTV-TGF-α/Lepr+ Lepr+ (homozygous) and MMTV-TGF-α/Lepr+ Leprdb (heterozygous) mice and obese MMTV-TGF-α/Leprdb Leprdb mice were monitored until age 104 weeks. Body weights of MMTV-TGF-α/Leprdb Leprdb mice were significantly heavier than those of the lean groups. No mammary tumors were detected in MMTV-TGF-α/LeprdbLeprdb mice, whereas the incidence of mammary tumors in MMTV-TGF-α/Lepr+ Lepr+ and MMTV-TGF-α/Lepr+ Leprdb mice was 69% and 82%, respectively. Examination of mammary tissue whole mounts indicated an absence of duct formation and branching for MMTV-TGF-α/Leprdb Leprdb mice. Both age at mammary tumor detection and tumor burden (tumors/mouse and tumor weights) were similar for the lean genotypes. Serum leptin levels of MMTV-TGF-α/Leprdb Leprdb mice were 12-20-fold higher than levels of lean mice. Thus, despite elevated serum leptin levels, leptin receptor-deficient MMTV-TGF-α/Leprdb Leprdb mice do not develop mammary tumors. This study provides additional evidence that leptin and its cognate receptor may be involved in mammary tumorigenesis.
Collapse
MESH Headings
- Animals
- Body Weight
- Disease Models, Animal
- Female
- Leptin/blood
- Leptin/metabolism
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mammary Tumor Virus, Mouse/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Obesity/genetics
- Obesity/pathology
- Oncogenes/genetics
- Receptors, Cell Surface/deficiency
- Receptors, Cell Surface/physiology
- Receptors, Leptin
- Survival Analysis
- Transforming Growth Factor alpha/genetics
Collapse
Affiliation(s)
- Margot P Cleary
- Hormel Institute, University of Minnesota, Austin, Minnesota 55912, USA.
| | | | | | | | | | | |
Collapse
|
206
|
Abstract
Adipose tissue plays a central role in regulating whole-body energy and glucose homeostasis through its subtle functions at both organ and systemic levels. On one hand, adipose tissue stores energy in the form of lipid and controls the lipid mobilization and distribution in the body. On the other hand, adipose tissue acts as an endocrine organ and produces numerous bioactive factors such as adipokines that communicate with other organs and modulate a range of metabolic pathways. Moreover, brown and beige adipose tissue burn lipid by dissipating energy in the form of heat to maintain euthermia, and have been considered as a new way to counteract obesity. Therefore, adipose tissue dysfunction plays a prominent role in the development of obesity and its related disorders such as insulin resistance, cardiovascular disease, diabetes, depression and cancer. In this review, we will summarize the recent findings of adipose tissue in the control of metabolism, focusing on its endocrine and thermogenic function.
Collapse
Affiliation(s)
- Liping Luo
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
| | - Meilian Liu
- Department of Metabolism and EndocrinologyMetabolic Syndrome Research Center, Key Laboratory of Diabetes Immunology, National Clinical Research Center for Metabolic Diseases, The Second Xiangya Hospital,
Central South University, Changsha, Hunan, China
- Department of Biochemistry and Molecular BiologyUniversity of New Mexico Health Sciences Center,
Albuquerque, New Mexico, USA
| |
Collapse
|
207
|
Yu S, Wu X, Ferguson M, Simmen RC, Cleves MA, Simmen FA, Fang N. Diets Containing Shiitake Mushroom Reduce Serum Lipids and Serum Lipophilic Antioxidant Capacity in Rats. J Nutr 2016; 146:2491-2496. [PMID: 27798348 PMCID: PMC5118771 DOI: 10.3945/jn.116.239806] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 08/18/2016] [Accepted: 09/15/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND We previously reported that dietary intake of shiitake mushroom (SM; Lentinus edodes) decreased serum concentrations of polar lipids in male rats. OBJECTIVE This study evaluated the dietary effects of SM on serum cholesterol-related and serum antioxidant indexes in rats of both sexes. METHODS Sprague-Dawley rats [38 dams and their offspring (20 males and 20 females/diet)] were fed diets containing 0 (control), 1%, 4%, or 10% (wt:wt) SM powder from gestation day 4 through to postnatal day (PND) 126. Biochemical indexes were monitored during the midgrowth phase (PNDs 50-66). RESULTS The food consumption by offspring fed the control diet and diets supplemented with SM was not different when measured on PND 65. However, the 4% and 10% SM diets resulted in male rats with 7% lower body weights than those of the other 2 groups on PND 66. SM consumption dose-dependently decreased the concentrations of lipidemia-related factors in sera, irrespective of sex. At PND 50, serum concentrations of total cholesterol, HDL cholesterol, and non-HDL cholesterol in SM-fed male and female rats were generally lower (3-27%) than those in the corresponding control groups. Consumption of the 10% SM diet resulted in significantly decreased (55%) serum triglyceride concentrations relative to the control groups for both sexes. The 10% SM diet elicited a 62% reduction of serum leptin concentrations in females but not in males, and this same diet increased serum insulin (137%) and decreased serum glucose (15%) in males compared with controls. Serum lipophilic antioxidant capacity in males and females fed SM diets was generally lower (31-86%) than that in the control groups. CONCLUSION SM decreased the concentrations of lipidemia-related factors in rat sera irrespective of sex. The SM-elicited reduction of lipophilic antioxidant capacity irrespective of sex may reflect a lower pro-oxidative state and, hence, improved metabolic profile.
Collapse
Affiliation(s)
- Shanggong Yu
- Arkansas Children's Nutrition Center, Little Rock, AR
| | - Xianli Wu
- Beltsville Human Nutrition Research Center, USDA-Agricultural Research Service, Beltsville, MD; and
| | | | | | - Mario A Cleves
- Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | | | - Nianbai Fang
- Arkansas Children's Nutrition Center, Little Rock, AR;
- Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| |
Collapse
|
208
|
Abstract
Leptin is an adipose tissue hormone that functions as an afferent signal in a negative feedback loop that maintains homeostatic control of adipose tissue mass. This endocrine system thus serves a critical evolutionary function by protecting individuals from the risks associated with being too thin (starvation) or too obese (predation and temperature dysregulation). Mutations in leptin or its receptor cause massive obesity in mice and humans, and leptin can effectively treat obesity in leptin-deficient patients. Leptin acts on neurons in the hypothalamus and elsewhere to elicit its effects, and mutations that affect the function of this neural circuit cause Mendelian forms of obesity. Leptin levels fall during starvation and elicit adaptive responses in many other physiologic systems, the net effect of which is to reduce energy expenditure. These effects include cessation of menstruation, insulin resistance, alterations of immune function, and neuroendocrine dysfunction, among others. Some or all of these effects are also seen in patients with constitutively low leptin levels, such as occur in lipodystrophy. Leptin is an approved treatment for generalized lipodystrophy, a condition associated with severe metabolic disease, and has also shown potential for the treatment of other types of diabetes. In addition, leptin restores reproductive capacity and increases bone mineral density in patients with hypothalamic amenorrhea, an infertility syndrome in females. Most obese patients have high endogenous levels of leptin, in some instances as a result of mutations in the neural circuit on which leptin acts, though in most cases, the pathogenesis of leptin resistance is not known. Obese patients with leptin resistance show a variable response to exogenous leptin but may respond to a combination of leptin plus amylin. Overall, the identification of leptin has provided a framework for studying the pathogenesis of obesity in the general population, clarified the nature of the biologic response to starvation, and helped to advance our understanding of the neural mechanisms that control feeding.
Collapse
|
209
|
Cohen K, Waldman M, Abraham NG, Laniado-Schwartzman M, Gurfield D, Aravot D, Arad M, Hochhauser E. Caloric restriction ameliorates cardiomyopathy in animal model of diabetes. Exp Cell Res 2016; 350:147-153. [PMID: 27884680 DOI: 10.1016/j.yexcr.2016.11.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/16/2016] [Accepted: 11/19/2016] [Indexed: 01/08/2023]
Abstract
BACKGROUND The db/db mouse is an animal model of diabetes in which leptin receptor activity is deficient resulting accelerated cardiomyopathy when exposed to angiotensin (AT). Toll-like receptors 4 and 2 (TLR4, TLR2) are pattern recognition receptors, that recognize pathogen-associated molecular patterns and exacerbate and release inflammatory cytokines. Fetuin A (Fet A) is a fatty acid carrier which affects inflammation and insulin resistance in obese humans and animals through TLRs. The aim of this study was to investigate the effect of caloric restriction (CR) on free fatty acids (FFA) level and the inflammatory response in diabetic cardiomyopathy. METHODS AND RESULTS Left ventricular hypertrophy, increased fibrosis and leukocytes infiltration were observed in db/db AT treated hearts. Serum glucose, FFA, and cholesterol levels were elevated in db/db AT treated mice. Cardiac expression of PPARα increased while AKT phosphorylation was decreased. CONCLUSIONS Cumulatively, CR elevated cardiac PPARα improved the utilization of fatty acids, and reduced myocardial inflammation as seen by reduced levels of Fet A. Thus CR negated cardiomyopathy associated with AT in an animal model of diabetes suggesting that CR is an effective therapeutic approach in the treatment of diabetes and associated cardiomyopathy.
Collapse
Affiliation(s)
- Keren Cohen
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Israel; Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Israel
| | - Maayan Waldman
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Israel; Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Israel
| | - Nader G Abraham
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | | | - Danny Gurfield
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Israel
| | - Dan Aravot
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Israel
| | - Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Israel
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Israel.
| |
Collapse
|
210
|
Thon M, Hosoi T, Ozawa K. Possible Integrative Actions of Leptin and Insulin Signaling in the Hypothalamus Targeting Energy Homeostasis. Front Endocrinol (Lausanne) 2016; 7:138. [PMID: 27812350 PMCID: PMC5071376 DOI: 10.3389/fendo.2016.00138] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/07/2016] [Indexed: 12/17/2022] Open
Abstract
Obesity has emerged as one of the most burdensome conditions in modern society. In this context, understanding the mechanisms controlling food intake is critical. At present, the adipocyte-derived hormone leptin and the pancreatic β-cell-derived hormone insulin are considered the principal anorexigenic hormones. Although leptin and insulin signal transduction pathways are distinct, their regulation of body weight maintenance is concerted. Resistance to the central actions of leptin or insulin is linked to the emergence of obesity and diabetes mellitus. A growing body of evidence suggests a convergence of leptin and insulin intracellular signaling at the insulin-receptor-substrate-phosphatidylinositol-3-kinase level. Moreover, numerous factors mediating the pathophysiology of leptin resistance, a hallmark of obesity, such as endoplasmic reticulum stress, protein tyrosine phosphatase 1B, and suppressor of cytokine signaling 3 also contribute to insulin resistance. Recent studies have also indicated that insulin potentiates leptin-induced signaling. Thus, a greater understanding of the overlapping functions of leptin and insulin in the central nervous system is vital to understand the associated physiological and pathophysiological states. This mini-review focuses on the cross talk and integrative signaling of leptin and insulin in the regulation of energy homeostasis in the brain.
Collapse
Affiliation(s)
- Mina Thon
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Toru Hosoi
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koichiro Ozawa
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
211
|
Xu Y, Tong Q. Central leptin action on euglycemia restoration in type 1 diabetes: Restraining responses normally induced by fasting? Int J Biochem Cell Biol 2016; 88:198-203. [PMID: 27702650 DOI: 10.1016/j.biocel.2016.09.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 09/27/2016] [Accepted: 09/30/2016] [Indexed: 01/29/2023]
Abstract
Leptin monotherapy is sufficient to restore euglycemia in insulinopenic type 1 diabetes (T1D), yet the underlying mechanism remains poorly understood. Accumulating evidence demonstrates that the brain mediates the leptin action on euglycemia restoration. Here, we first review evidence supporting that symptoms in T1D resemble an uncontrolled response to fasting. Then, we discuss recent research progress on brain neurons and their neurotransmitters that potentially mediate the leptin action. Finally, peripheral effective pathways, which are normally involved in fasting responses and associated with leptin action on euglycemia restoration in T1D, will also be discussed. This summary complements several previous excellent reviews on this topic (Meek and Morton, 2016; Perry et al., 2016; Fujikawa and Coppari, 2015). A deep understanding of neurocircuitry and the peripheral effective pathways that mediate the leptin action on euglycemia restoration will likely lead to novel targets for an insulin-independent therapeutics against T1D.
Collapse
Affiliation(s)
- Yuanzhong Xu
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, The University of Texas Health Science Center at Houston, United States
| | - Qingchun Tong
- Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases of McGovern Medical School, The University of Texas Health Science Center at Houston, United States.
| |
Collapse
|
212
|
|
213
|
He Q, Scott Swindle C, Wan C, Flynn RJ, Oster RA, Chen D, Zhang F, Shu Y, Klug CA. Enhanced Hematopoietic Stem Cell Self-Renewal-Promoting Ability of Clonal Primary Mesenchymal Stromal/Stem cells Versus Their Osteogenic Progeny. Stem Cells 2016; 35:473-484. [PMID: 27539014 DOI: 10.1002/stem.2481] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 07/08/2016] [Accepted: 08/04/2016] [Indexed: 12/22/2022]
Abstract
Long-term self-renewing hematopoietic stem cell (LT-HSC) homeostasis within the bone marrow (BM) of adult mammals is regulated by complex interactions between LT-HSC and a number of niche-associated cell types including mesenchymal stromal/stem cells (MSC), osteoblasts (OB), macrophage, and neuronal cells in close proximity with the vasculature. Here, we cloned and functionally characterized a murine BM MSC subpopulation that was uniformly Nestin+ Lepr + Sca-1+ CD146+ and could be stably propagated with high colony-forming unit fibroblast re-cloning efficiency. MSC synergized with SCF and IL-11 to support a 20-fold expansion in true LT-HSC after 10-days of in vitro coculture. Optimal stimulation of LT-HSC expansion was minimally dependent on Notch signaling but was significantly enhanced by global inhibition of Wnt signaling. The self-renewal-promoting activity of MSC was progressively lost when MSC clones were differentiated into mature OB. This suggests that the stage of osteoblast development may significantly impact the ability of osteolineage cells to support LT-HSC homeostasis in vivo. Stem Cells 2017;35:473-484.
Collapse
Affiliation(s)
- Qiling He
- Department of Microbiology, University of Alabama-Birmingham, Alabama, USA
| | | | - Chao Wan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, P.R. China.,School of Biomedical Sciences Core Laboratory, Institute of Stem Cell, Genomics and Translational Research, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, P.R. China
| | - Robert J Flynn
- Department of Microbiology, University of Alabama-Birmingham, Alabama, USA
| | - Robert A Oster
- Division of Preventative Medicine, University of Alabama-Birmingham, Alabama, USA
| | - Dongquan Chen
- Division of Preventative Medicine, University of Alabama-Birmingham, Alabama, USA
| | - Fengjie Zhang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Yinglan Shu
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Christopher A Klug
- Department of Microbiology, University of Alabama-Birmingham, Alabama, USA
| |
Collapse
|
214
|
Tsukada S, Masuda H, Jung SY, Yun J, Kang S, Kim DY, Park JH, Ji ST, Kwon SM, Asahara T. Impaired development and dysfunction of endothelial progenitor cells in type 2 diabetic mice. DIABETES & METABOLISM 2016; 43:154-162. [PMID: 27638126 DOI: 10.1016/j.diabet.2016.07.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 07/02/2016] [Accepted: 07/23/2016] [Indexed: 12/16/2022]
Abstract
AIM Dysfunction of circulating endothelial progenitor cells (EPCs) has been shown to affect the development of microvascular diseases in diabetes patients. The aim of this study was to elucidate the development and mechanical dysfunction of EPCs in type 2 diabetes (T2D). METHODS The colony-forming capacity of EPCs and differentiation potential of bone marrow (BM) c-Kit(+)/Sca-I(+) lineage-negative mononuclear cells (KSL) were examined in T2D mice, db/db mice and KKAy mice, using EPC colony-forming assay (EPC-CFA). RESULTS T2D mice had fewer BM stem/progenitor cells, and proliferation of KSL was lowest in the BM of db/db mice. In T2D mice, the frequency of large colony-forming units (CFUs) derived from BM-KSL was highly reduced, indicating dysfunction of differentiation into mature EPCs. Only a small number of BM-derived progenitors [CD34(+) KSL cells], which contribute to the supply of EPCs for postnatal neovascularization, was also found. Furthermore, in terms of their plasticity to transdifferentiate into various cell types, BM-KSL exhibited a greater potential to differentiate into granulocyte macrophages (GMs) than into other cell types. CONCLUSION T2D affected EPC colony formation and differentiation of stem cells to mature EPCs or haematopoietic cells. These data suggest opposing regulatory mechanisms for differentiation into mature EPCs and GMs in T2D mice.
Collapse
Affiliation(s)
- S Tsukada
- Department Regenerative Medicine, Tokai University of Medicine, Kobe, Japan
| | - H Masuda
- Department Regenerative Medicine, Tokai University of Medicine, Kobe, Japan
| | - S Y Jung
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - J Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - S Kang
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - D Y Kim
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - J H Park
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - S T Ji
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - S-M Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Medical Research Institute, Department of Physiology, School of Medicine, Pusan National University, Yangsan, Republic of Korea; Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan National University, Yangsan, Republic of Korea.
| | - T Asahara
- Department Regenerative Medicine, Tokai University of Medicine, Kobe, Japan; Stem Cell Translational Research Laboratory, Center For Developmental Biology, RIKEN, Kobe, Japan; Kobe Institute of Biomedical Research and Innovation, Kobe, Japan.
| |
Collapse
|
215
|
Affiliation(s)
- Charles V Mobbs
- Departments of Neuroscience; Endocrinology, Diabetes and Bone Disease; and Geriatrics and Palliative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
216
|
Abstract
Carbohydrate, lipid, and protein metabolism are largely controlled by the interplay of various hormones, which includes those secreted by the pancreatic islets of Langerhans. While typically representing only 1% to 2% of the total pancreatic mass, the islets have a remarkable ability to adapt to disparate situations demanding a change in hormone release, such as peripheral insulin resistance. There are many different routes to the onset of insulin resistance, including obesity, lipodystrophy, glucocorticoid excess, and the chronic usage of atypical antipsychotic drugs. All of these situations are coupled to an increase in pancreatic islet size, often with a corresponding increase in insulin production. These adaptive responses within the islets are ultimately intended to maintain glycemic control and to promote macronutrient homeostasis during times of stress. Herein, we review the consequences of specific metabolic trauma that lead to insulin resistance and the corresponding adaptive alterations within the pancreatic islets.
Collapse
Affiliation(s)
- Susan J. Burke
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Michael D. Karlstad
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Knoxville, TN 37920
| | - J. Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Knoxville, TN 37920
| |
Collapse
|
217
|
Gong N, Johansson M, Björnsson BT. Impaired central leptin signaling and sensitivity in rainbow trout with high muscle adiposity. Gen Comp Endocrinol 2016; 235:48-56. [PMID: 27292790 DOI: 10.1016/j.ygcen.2016.06.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 06/05/2016] [Accepted: 06/08/2016] [Indexed: 11/22/2022]
Abstract
The hormone leptin has been identified in all vertebrate classes, but its physiological roles in non-mammalian vertebrates are not well defined. To elucidate leptin regulation in energy homeostasis in a teleost fish species, this study compares hypothalamic and pituitary leptin signaling systems in energetically divergent rainbow trout lines selected for low (lean line, LL) and high (fat line, FL) muscle adiposity under feeding and starvation conditions. In fed fish, hypothalamic gene expression and protein density of the full-functional leptin receptor (LepRL), as well as a leptin binding protein (LepBP) expression, are lower in FL than LL fish. The FL fish have also lower activation of leptin-relevant signaling pathways involving protein kinase B (Akt) and extracellular signal-related kinase. These observations suggests impaired central leptin action in FL fish. During fasting, hypothalamic LepRL and LepBP expression, as well as active Akt levels are downregulated after one week, while pituitary LepRL expression is upregulated, in the LL fish only. After four weeks, hypothalamic LepRL protein levels return to normal levels in both fish lines and Akt is reactivated, although not to the same extent in FL as in LL fish, indicating that FL fish have low leptin sensitivity to nutritional changes. Neuropeptide Y and orexin expression is downregulated to similar levels in both fish lines after one-week fasting. The divergent leptin system profiles between the two fish lines demonstrate that phenotypic selection for high muscle adiposity affects leptin endocrinology, indicating regulatory roles for leptin in rainbow trout energy homeostasis.
Collapse
Affiliation(s)
- Ningping Gong
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg, S-40590 Gothenburg, Sweden
| | - Marcus Johansson
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg, S-40590 Gothenburg, Sweden
| | - Björn Thrandur Björnsson
- Fish Endocrinology Laboratory, Department of Biological and Environmental Sciences, University of Gothenburg, S-40590 Gothenburg, Sweden.
| |
Collapse
|
218
|
Sundara Rajan S, Longhi MP. Dendritic cells and adipose tissue. Immunology 2016; 149:353-361. [PMID: 27479803 DOI: 10.1111/imm.12653] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022] Open
Abstract
Visceral adipose tissue inflammation in obesity is an established risk factor for metabolic syndrome, which can include insulin resistance, type 2 diabetes, hypertension and cardiovascular diseases. With obesity and related metabolic disorders reaching epidemic proportions globally, an understanding of the mechanisms of adipose tissue inflammation is crucial. Within the immune cell cohort, dendritic cells (DC) play a key role in balancing tolerance and immunity. Despite decades of research into the characterization of DC in lymphoid and non-lymphoid organs, their role in adipose tissue function is poorly understood. There is now an increasing interest in identification and characterization of DC in adipose tissue and understanding their function in regulating tissue metabolic homeostasis. This review provides an overview of the study of DC in adipose tissue, focusing on possible mechanisms by which DC may contribute to adipose tissue homeostasis.
Collapse
Affiliation(s)
- Sandeep Sundara Rajan
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - Maria Paula Longhi
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK.
| |
Collapse
|
219
|
Xu S, Zhu X, Li H, Hu Y, Zhou J, He D, Feng Y, Lu L, Du G, Hu Y, Liu T, Wang Z, Ding G, Chen J, Gao S, Wu F, Xue Z, Li Y, Fan G. The 14th Ile residue is essential for Leptin function in regulating energy homeostasis in rat. Sci Rep 2016; 6:28508. [PMID: 27378381 PMCID: PMC4932527 DOI: 10.1038/srep28508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/06/2016] [Indexed: 12/26/2022] Open
Abstract
LEPTIN (LEP) is a circulating hormone released primarily from white adipocytes and is crucial for regulating satiety and energy homeostasis in humans and animals. Using the CRISPR technology, we created a set of Lep mutant rats that carry either null mutations or a deletion of the 14(th) Ile (LEP(∆I14)) in the mature LEP protein. We examined the potential off-target sites (OTS) by whole-genome high-throughput sequencing and/or Sanger-sequencing analysis and found no OTS in mutant rats. Mature LEP(∆I14) is incessantly produced and released to blood at a much elevated level due to the feedback loop. Structure modeling of binding conformation between mutant LEP(∆I14) and LEPTIN receptor (LEPR) suggests that the conformation of LEP(∆I14) impairs its binding with LEPR, consistent with its inability to activate STAT3-binding element in the luciferase reporter assay. Phenotypic study demonstrated that Lep(∆I14) rats recapitulate phenotypes of Lep-null mutant rats including obesity, hyperinsulinemia, hepatic steatosis, nephropathy, and infertility. Compared to the existing ob/ob mouse models, this Lep(∆I14/∆I14) rat strain provides a robust tool for further dissecting the roles of LEP in the diabetes related kidney disease and reproduction problem, beyond its well established function in regulating energy homeostasis.
Collapse
Affiliation(s)
- Shuyang Xu
- Tongji University, School of Life Sciences and Technology, 1239 Siping Road, Shanghai 200092, China
| | - Xianmin Zhu
- Tongji University, School of Life Sciences and Technology, 1239 Siping Road, Shanghai 200092, China
| | - Hong Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Youtian Hu
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Jinping Zhou
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai 200065, China
| | - Di He
- Tongji University, School of Life Sciences and Technology, 1239 Siping Road, Shanghai 200092, China
| | - Yun Feng
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai 200065, China
| | - Lina Lu
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai 200065, China
| | - Guizhen Du
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, CA 90095 USA
| | - Youjin Hu
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai 200065, China.,Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, CA 90095 USA
| | - Tiancheng Liu
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhen Wang
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guohui Ding
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jiayu Chen
- Tongji University, School of Life Sciences and Technology, 1239 Siping Road, Shanghai 200092, China
| | - Shaorong Gao
- Tongji University, School of Life Sciences and Technology, 1239 Siping Road, Shanghai 200092, China
| | - Fang Wu
- Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 800 Dong Chuan Road, Shanghai 200240, China
| | - Zhigang Xue
- Translational Center for Stem Cell Research, Tongji Hospital, Department of Regenerative Medicine, Tongji University School of Medicine, Shanghai 200065, China
| | - Yixue Li
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guoping Fan
- Tongji University, School of Life Sciences and Technology, 1239 Siping Road, Shanghai 200092, China.,Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, CA 90095 USA
| |
Collapse
|
220
|
Denroche HC, Glavas MM, Tudurí E, Karunakaran S, Quong WL, Philippe M, Britton HM, Clee SM, Kieffer TJ. Disrupted Leptin Signaling in the Lateral Hypothalamus and Ventral Premammillary Nucleus Alters Insulin and Glucagon Secretion and Protects Against Diet-Induced Obesity. Endocrinology 2016; 157:2671-85. [PMID: 27183315 DOI: 10.1210/en.2015-1998] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Leptin signaling in the central nervous system, and particularly the arcuate hypothalamic nucleus, is important for regulating energy and glucose homeostasis. However, the roles of extra-arcuate leptin responsive neurons are less defined. In the current study, we generated mice with widespread inactivation of the long leptin receptor isoform in the central nervous system via Synapsin promoter-driven Cre (Lepr(flox/flox) Syn-cre mice). Within the hypothalamus, leptin signaling was disrupted in the lateral hypothalamic area (LHA) and ventral premammillary nucleus (PMV) but remained intact in the arcuate hypothalamic nucleus and ventromedial hypothalamic nucleus, dorsomedial hypothalamic nucleus, and nucleus of the tractus solitarius. To investigate the role of LHA/PMV neuronal leptin signaling, we examined glucose and energy homeostasis in Lepr(flox/flox) Syn-cre mice and Lepr(flox/flox) littermates under basal and diet-induced obese conditions and tested the role of LHA/PMV neurons in leptin-mediated glucose lowering in streptozotocin-induced diabetes. Lepr(flox/flox) Syn-cre mice did not have altered body weight or blood glucose levels but were hyperinsulinemic and had enhanced glucagon secretion in response to experimental hypoglycemia. Surprisingly, when placed on a high-fat diet, Lepr(flox/flox) Syn-cre mice were protected from weight gain, glucose intolerance, and diet-induced hyperinsulinemia. Peripheral leptin administration lowered blood glucose in streptozotocin-induced diabetic Lepr(flox/flox) Syn-cre mice as effectively as in Lepr(flox/flox) littermate controls. Collectively these findings suggest that leptin signaling in LHA/PMV neurons is not critical for regulating glucose levels but has an indispensable role in the regulation of insulin and glucagon levels and, may promote the development of diet-induced hyperinsulinemia and weight gain.
Collapse
Affiliation(s)
- Heather C Denroche
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Maria M Glavas
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Eva Tudurí
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Subashini Karunakaran
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Whitney L Quong
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Marion Philippe
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Heidi M Britton
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Susanne M Clee
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| | - Timothy J Kieffer
- Laboratory of Molecular and Cellular Medicine (H.C.D., M.M.G., E.T., W.L.Q., M.P., H.M.B., T.J.K.) and Laboratory of the Genetics of Obesity and Diabetes (S.K., S.M.C.), Department of Cellular and Physiological Sciences, Life Sciences Institute, and Department of Surgery (T.J.K.), University of British Columbia, Vancouver, British Columbia, Canada V6T 1Z3
| |
Collapse
|
221
|
Alliouachene S, Bilanges B, Chaussade C, Pearce W, Foukas LC, Scudamore CL, Moniz LS, Vanhaesebroeck B. Inactivation of class II PI3K-C2α induces leptin resistance, age-dependent insulin resistance and obesity in male mice. Diabetologia 2016; 59:1503-1512. [PMID: 27138914 PMCID: PMC4901096 DOI: 10.1007/s00125-016-3963-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 03/24/2016] [Indexed: 01/08/2023]
Abstract
AIMS/HYPOTHESIS While the class I phosphoinositide 3-kinases (PI3Ks) are well-documented positive regulators of metabolism, the involvement of class II PI3K isoforms (PI3K-C2α, -C2β and -C2γ) in metabolic regulation is just emerging. Organismal inactivation of PI3K-C2β increases insulin signalling and sensitivity, whereas PI3K-C2γ inactivation has a negative metabolic impact. In contrast, the role of PI3K-C2α in organismal metabolism remains unexplored. In this study, we investigated whether kinase inactivation of PI3K-C2α affects glucose metabolism in mice. METHODS We have generated and characterised a mouse line with a constitutive inactivating knock-in (KI) mutation in the kinase domain of the gene encoding PI3K-C2α (Pik3c2a). RESULTS While homozygosity for kinase-dead PI3K-C2α was embryonic lethal, heterozygous PI3K-C2α KI mice were viable and fertile, with no significant histopathological findings. However, male heterozygous mice showed early onset leptin resistance, with a defect in leptin signalling in the hypothalamus, correlating with a mild, age-dependent obesity, insulin resistance and glucose intolerance. Insulin signalling was unaffected in insulin target tissues of PI3K-C2α KI mice, in contrast to previous reports in which downregulation of PI3K-C2α in cell lines was shown to dampen insulin signalling. Interestingly, no metabolic phenotypes were detected in female PI3K-C2α KI mice at any age. CONCLUSIONS/INTERPRETATION Our data uncover a sex-dependent role for PI3K-C2α in the modulation of hypothalamic leptin action and systemic glucose homeostasis. ACCESS TO RESEARCH MATERIALS All reagents are available upon request.
Collapse
Affiliation(s)
- Samira Alliouachene
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| | - Benoit Bilanges
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| | - Claire Chaussade
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
- Galderma R&D, Sophia Antipolis Cedex, France
| | - Wayne Pearce
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Cheryl L Scudamore
- Mary Lyon Centre, MRC Harwell, Harwell Science and Innovation Campus, Harwell, UK
| | - Larissa S Moniz
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK
| | - Bart Vanhaesebroeck
- UCL Cancer Institute, University College London, 72 Huntley Street, London, WC1E 6DD, UK.
| |
Collapse
|
222
|
Cornejo MP, Hentges ST, Maliqueo M, Coirini H, Becu-Villalobos D, Elias CF. Neuroendocrine Regulation of Metabolism. J Neuroendocrinol 2016; 28:10.1111/jne.12395. [PMID: 27114114 PMCID: PMC4956544 DOI: 10.1111/jne.12395] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/31/2016] [Accepted: 04/21/2016] [Indexed: 12/29/2022]
Abstract
Given the current environment in most developed countries, it is a challenge to maintain a good balance between calories consumed and calories burned, although maintenance of metabolic balance is key to good health. Therefore, understanding how metabolic regulation is achieved and how the dysregulation of metabolism affects health is an area of intense research. Most studies focus on the hypothalamus, which is a brain area that acts as a key regulator of metabolism. Among the nuclei that comprise the hypothalamus, the arcuate nucleus is one of the major mediators in the regulation of food intake. The regulation of energy balance is also a key factor ensuring the maintenance of any species as a result of the dependence of reproduction on energy stores. Adequate levels of energy reserves are necessary for the proper functioning of the hypothalamic-pituitary-gonadal axis. This review discusses valuable data presented in the 2015 edition of the International Workshop of Neuroendocrinology concerning the fundamental nature of the hormonal regulation of the hypothalamus and the impact on energy balance and reproduction.
Collapse
Affiliation(s)
- Maria P. Cornejo
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE, dependent on the Argentine Research Council (CONICET), Scientific Research Commission, Province of Buenos Aires (CIC-PBA) and National University of La Plata (UNLP)], La Plata, Argentina
| | - Shane T. Hentges
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Manuel Maliqueo
- Endocrinology and Metabolism Laboratory, Department of Medicine West Division, School of Medicine University of Chile, Santiago de Chile, Chile
| | - Hector Coirini
- Laboratory of Neurobiology, Institute of Biology and Experimental Medicine [(IBYME), dependent on CONICET] and Department of Human Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Damasia Becu-Villalobos
- Laboratory of Pituitary Regulation, Institute of Biology and Experimental Medicine [(IBYME), dependent on CONICET], Buenos Aires, Argentina
| | - Carol F. Elias
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
223
|
Abstract
The domestication and urbanization of dogs and cats has dramatically altered their environment and behavior. Human and pet obesity is a global concern, particularly in developed countries. An increased incidence of chronic disease is associated with obesity secondary to low-grade systemic inflammation. This article reviews current research into the genetic, dietary, and physiologic factors associated with obesity, along with use of "omics" technology to better understand and characterize this disease.
Collapse
Affiliation(s)
- Beth Hamper
- Hamper Veterinary Nutritional Consulting, 9160 Crestview Drive, Indianapolis, IN 46240, USA.
| |
Collapse
|
224
|
Jyotaki M, Sanematsu K, Shigemura N, Yoshida R, Ninomiya Y. Leptin suppresses sweet taste responses of enteroendocrine STC-1 cells. Neuroscience 2016; 332:76-87. [PMID: 27353597 DOI: 10.1016/j.neuroscience.2016.06.036] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 06/17/2016] [Accepted: 06/22/2016] [Indexed: 12/26/2022]
Abstract
Leptin is an important hormone that regulates food intake and energy homeostasis by acting on central and peripheral targets. In the gustatory system, leptin is known to selectively suppress sweet responses by inhibiting the activation of sweet sensitive taste cells. Sweet taste receptor (T1R2+T1R3) is also expressed in gut enteroendocrine cells and contributes to nutrient sensing, hormone release and glucose absorption. Because of the similarities in expression patterns between enteroendocrine and taste receptor cells, we hypothesized that they may also share similar mechanisms used to modify/regulate the sweet responsiveness of these cells by leptin. Here, we used mouse enteroendocrine cell line STC-1 and examined potential effect of leptin on Ca(2+) responses of STC-1 cells to various taste compounds. Ca(2+) responses to sweet compounds in STC-1 cells were suppressed by a rodent T1R3 inhibitor gurmarin, suggesting the involvement of T1R3-dependent receptors in detection of sweet compounds. Responses to sweet substances were suppressed by ⩾1ng/ml leptin without affecting responses to bitter, umami and salty compounds. This effect was inhibited by a leptin antagonist (mutant L39A/D40A/F41A) and by ATP gated K(+) (KATP) channel closer glibenclamide, suggesting that leptin affects sweet taste responses of enteroendocrine cells via activation of leptin receptor and KATP channel expressed in these cells. Moreover, leptin selectively inhibited sweet-induced but not bitter-induced glucagon-like peptide-1 (GLP-1) secretion from STC-1 cells. These results suggest that leptin modulates sweet taste responses of enteroendocrine cells to regulate nutrient sensing, hormone release and glucose absorption in the gut.
Collapse
Affiliation(s)
- Masafumi Jyotaki
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka 812-8582, Japan; Monell Chemical Senses Center, Philadelphia, PA, United States
| | - Keisuke Sanematsu
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryusuke Yoshida
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka 812-8582, Japan; OBT Research Center, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yuzo Ninomiya
- Section of Oral Neuroscience, Graduate School of Dental Sciences, Kyushu University, Fukuoka 812-8582, Japan; Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan; Monell Chemical Senses Center, Philadelphia, PA, United States.
| |
Collapse
|
225
|
Wan J, Li P, Liu DW, Chen Y, Mo HZ, Liu BG, Chen WJ, Lu XQ, Guo J, Zhang Q, Qiao YJ, Liu ZS, Wan GR. GSK-3β inhibitor attenuates urinary albumin excretion in type 2 diabetic db/db mice, and delays epithelial-to-mesenchymal transition in mouse kidneys and podocytes. Mol Med Rep 2016; 14:1771-84. [PMID: 27357417 DOI: 10.3892/mmr.2016.5441] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 02/02/2016] [Indexed: 11/05/2022] Open
Abstract
The mechanism underlying epithelial‑to‑mesenchymal transition (EMT) caused by high glucose (HG) stimulation in diabetic nephropathy (DN) remains to be fully elucidated. The present study investigated the effects of HG on EMT and the activity of glycogen synthase kinase 3β (GSK‑3β) in podocytes and the kidneys of db/db mice, and assessed the effects of (2'Z, 3'E)‑6‑bromoindirubin‑3'‑oxime (BIO), an inhibitor of GSK‑3β, on EMT and glomerular injury. The resulting data showed that the activity of GSK‑3β was upregulated by HG and downregulated by BIO in the podocytes and the renal cortex. The expression levels of epithelial markers, including nephrin, podocin and synaptopodin, were decreased by HG and increased by BIO, whereas the reverse were true for mesenchymal markers, including α‑smooth muscle actin (α‑SMA) and fibronectin. The expression levels of β‑catenin and Snail, in contrast to current understanding of the Wnt signaling pathway, were increased by HG and decreased by BIO. In addition, expression of the vitamin D receptor (VDR) was decreased by HG and increased by BIO. In conclusion, the present study revealed that the mechanism by which BIO inhibited HG‑mediated EMT in podocytes and the renal cortex was primarily due to the VDR. Treatment with BIO protected renal function by maintaining the integrity of the filtration membrane and decreasing UAE, but not by regulating blood glucose. Therefore, GSK‑3β may be used as a sensitive biomarker of DN, and its inhibition by BIO may be effective in the treatment of DN.
Collapse
Affiliation(s)
- Jia Wan
- Henan Food and Drug Administration, Zhengzhou, Henan 450012, P.R. China
| | - Peng Li
- Pharmaceutical College, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Dong-Wei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450012, P.R. China
| | - Ying Chen
- School of Life Science and Technology, Henan Institute of Science and Technology, Xinxiang, Henan 453003, P.R. China
| | - Hai-Zhen Mo
- Department of Food Science, Henan Institute of Science and Technology, Xinxiang, Henan 453003, P.R. China
| | - Ben-Guo Liu
- Department of Food Science, Henan Institute of Science and Technology, Xinxiang, Henan 453003, P.R. China
| | - Wen-Jie Chen
- Modern Education Technology Center, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xiao-Qing Lu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450012, P.R. China
| | - Jia Guo
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450012, P.R. China
| | - Qian Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450012, P.R. China
| | - Ying-Jin Qiao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450012, P.R. China
| | - Zhang-Suo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450012, P.R. China
| | - Guang-Rui Wan
- Pharmaceutical College, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
226
|
Wasim M, Awan FR, Najam SS, Khan AR, Khan HN. Role of Leptin Deficiency, Inefficiency, and Leptin Receptors in Obesity. Biochem Genet 2016; 54:565-72. [PMID: 27313173 DOI: 10.1007/s10528-016-9751-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 05/27/2016] [Indexed: 11/25/2022]
Abstract
Leptin protein consists of 167 amino acids, which is mainly secreted from the white adipose tissue. This protein acts on the hypothalamic regions of the brain which control eating behavior, thus playing a significant role in maintaining body's metabolism. Leptin receptors belong to glycoprotein 130 (gp130) family of cytokine receptors and exist in six isoforms (LEPR a-f), and all the isoforms are encoded by LEPR gene; out of these isoforms, the LEPR-b receptor is the 'longest form,' and in most of the cases, mutations in this isoform cause severe obesity. Also, mutations in the leptin gene (LEP) or its receptors gene can lead to obesity. Some biochemical pathways affect the bioactivity of leptin and/or its receptors. To date, eleven pathogenic mutations have been reported in the LEP which are p.L72S, p.N103K, p.R105W, p.H118L, p.S141C, p.W121X c.104_106delTCA, c.135del3bp, c.398delG, c.481_482delCT, and c.163C>T. Different mutations in the LEPR have also been reported as c.2396-1 G>T, c.1675 G>A, p.P316T, etc. In some studies, where leptin was deficient, leptin replacement therapy has shown positive impact by preventing weight gain and obesity.
Collapse
Affiliation(s)
- Muhammad Wasim
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan.
| | - Fazli Rabbi Awan
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Syeda Sadia Najam
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Abdul Rehman Khan
- Obesity and Diabetes Research Laboratory, Department of Chemistry, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| | - Haq Nawaz Khan
- Diabetes and Cardio-Metabolic Disorders Laboratory, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| |
Collapse
|
227
|
Reshma R, Mishra SR, Thakur N, Parmar MS, Somal A, Bharti MK, Pandey S, Chandra V, Chouhan VS, Verma MR, Singh G, Sharma GT, Maurya VP, Sarkar M. Modulatory role of leptin on ovarian functions in water buffalo (Bubalus bubalis). Theriogenology 2016; 86:1720-39. [PMID: 27381558 DOI: 10.1016/j.theriogenology.2016.05.029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/11/2022]
Abstract
The aim of the present study was to demonstrate the modulatory role of leptin on bubaline granulosa cells (GCs) and luteal cells (LCs) functions using an in vitro cell culture system and to establish a cross talk between leptin and insulin-like growth factor-1 (IGF-1). GCs were collected from group IV follicles (>13 mm size) and LCs from mid-luteal phase corpus luteum and were grown in serum-containing media supplemented with leptin at three different dose rates (0.1, 1, and 10 ng/mL) and time durations (24, 48, and 72 hours). We evaluated the production and secretion of estradiol (E2) and progesterone (P4) using RIA and the mRNA expression of steroidogenic acute regulatory protein (STARD1), cytochrome P450 cholesterol side-chain cleavage (CYP11A1), 3β-hydroxysteroid dehydrogenase (3β-HSD), cytochrome P450 aromatase (CYP19A1), sterol regulatory element-binding protein 1 (SREBP1), steroidogenic factor-1 (SF1), anti-apoptotic gene PCNA, pro-apoptotic gene caspase 3 and endothelial cell marker, Von Willebrand factor (vWF), using quantitative real-time polymerase chain reaction. The results depicted a direct inhibitory action of leptin on GCs steroidogenesis in a time-dependent manner (P < 0.05), whereas in the presence of IGF-1 the inhibitory effect was reverted. Furthermore, leptin augmented both cellular proliferation (PCNA) and apoptosis (caspase 3). On the other hand, in LCs, leptin alone showed an apparent stimulatory effect on steroidogenesis (P < 0.05); however, in the presence of IGF-1, an antagonistic effect was witnessed. Moreover, leptin had an inhibitory effect on apoptosis while promoted cellular proliferation and angiogenesis. These findings were further strengthened by immunocytochemistry. To conclude, these observations for the first time reported that in buffaloes leptin has a direct dose-, time-, and tissue-dependent effect on ovarian steroidogenesis, angiogenesis, and cytoprotection, and furthermore, it can regulate the effect of systemic factors like IGF-1. Hence, this in vitro study provides an insight into the putative roles of leptin alone and its interactions in vivo.
Collapse
Affiliation(s)
- R Reshma
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - S R Mishra
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - N Thakur
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - M S Parmar
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - A Somal
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - M K Bharti
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - S Pandey
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - V Chandra
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - V S Chouhan
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - M R Verma
- Division of Livestock Economics, Statistics and Information Technology, Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - G Singh
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - G T Sharma
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - V P Maurya
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - M Sarkar
- Physiology and Climatology, Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
| |
Collapse
|
228
|
Vasam G, Joshi S, Jarajapu YPR. Impaired Mobilization of Vascular Reparative Bone Marrow Cells in Streptozotocin-Induced Diabetes but not in Leptin Receptor-Deficient db/db Mice. Sci Rep 2016; 6:26131. [PMID: 27188595 PMCID: PMC4870646 DOI: 10.1038/srep26131] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/27/2016] [Indexed: 01/10/2023] Open
Abstract
Diabetes is associated with impaired mobilization of bone marrow stem/progenitor cells that accelerate vascularization of ischemic areas. This study characterized mobilization of vascular reparative bone marrow progenitor cells in mouse models of diabetes. Age-matched control or streptozotocin (STZ)-induced diabetic, and db/db mice with lean-controls were studied. Mobilization induced by G-CSF, AMD3100 or ischemia was evaluated by flow cytometric enumeration of circulating Lin(-)Sca-1(+)cKit(+) (LSK) cells, and by colony forming unit (CFU) assay. The circulating WBCs and LSKs, and CFUs were reduced in both models with a shorter duration (10-12 weeks) of diabetes compared to their respective controls. Longer duration of STZ-diabetes (≥20 weeks) induced impairment of G-CSF- or AMD3100-mobilization (P < 0.01, n = 8). In db/db mice, mobilization by G-CSF or AMD3100 was either increased or unaffected (P < 0.05, n = 6 to 8). Proliferation, migration, and ischemia-induced mobilization, of LSK cells were impaired in both models. Leptin receptor antagonist, PESLAN-1, increased G-CSF- or AMD3100-mobilization of WBCs and LSKs, compared to the untreated. Leptin increased basal WBCs, decreased basal and AMD3100-mobilized LSK cells, and had no effect on G-CSF. These results suggest that mobilopathy is apparent in STZ-diabetes but not in db/db mice. Leptin receptor antagonism would be a promising approach for reversing diabetic bone marrow mobilopathy.
Collapse
Affiliation(s)
- Goutham Vasam
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Shrinidh Joshi
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| | - Yagna P. R. Jarajapu
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota, USA
| |
Collapse
|
229
|
Abstract
PURPOSE OF REVIEW Sodium-glucose cotransporters (SGLTs) are important mediators of glucose uptake across apical cell membranes. SGLT1 mediates almost all sodium-dependent glucose uptake in the small intestine, while in the kidney SGLT2, and to a lesser extent SGLT1, account for more than 90% and nearly 3%, respectively, of glucose reabsorption from the glomerular ultrafiltrate. Although the recent availability of SGLT2 inhibitors for the treatment of diabetes mellitus has increased the number of clinical studies, this review has a focus on mechanisms contributing to the cellular regulation of SGLTs. RECENT FINDINGS Studies have focused on the regulation of SGLT expression under different physiological/pathophysiological conditions, for example diet, age or diabetes mellitus. Several studies provide evidence of SGLT regulation via cyclic adenosine monophosphate/protein kinase A, protein kinase C, glucagon-like peptide 2, insulin, leptin, signal transducer and activator of transcription-3 (STAT3), phosphoinositide-3 kinase (PI3K)/Akt, mitogen-activated protein kinases (MAPKs), nuclear factor-kappaB (NF-kappaB), with-no-K[Lys] kinases/STE20/SPS1-related proline/alanine-rich kinase (Wnk/SPAK) and regulatory solute carrier protein 1 (RS1) pathways. SUMMARY SGLT inhibitors are important drugs for glycemic control in diabetes mellitus. Although the contribution of SGLT1 for absorption of glucose from the intestine as well as SGLT2/SGLT1 for renal glucose reabsorption has been comprehensively defined, this review provides an up-to-date outline for the mechanistic regulation of SGLT1/SGLT2.
Collapse
|
230
|
Shpakov AO. The brain leptin signaling system and its functional state in metabolic syndrome and type 2 diabetes mellitus. J EVOL BIOCHEM PHYS+ 2016; 52:177-195. [DOI: 10.1134/s0022093016030017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
231
|
Abstract
Obesity and its major comorbidities, including type 2 diabetes mellitus, nonalcoholic fatty liver disease (NAFLD), obesity cardiomyopathy, and certain cancers, have caused life expectancy in the United States to decline in recent years. Obesity is the increased accumulation of triglycerides (TG), which are synthesized from glycerol and long-chain fatty acids (LCFA) throughout the body. LCFA enter adipocytes, hepatocytes, and cardiomyocytes via specific, facilitated transport processes. Metabolism of increased cellular TG content in obesity may lead to comorbidities such as NAFLD and cardiomyopathy. Better understanding of LCFA transport processes may lead to successful treatment of obesity and NAFLD.
Collapse
Affiliation(s)
- Paul D Berk
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia College of Physicians and Surgeons, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA; Division of Preventive Medicine, Department of Medicine, Columbia College of Physicians and Surgeons, Columbia University Medical Center, William Black Building, 650 West 168 Street, Room 1006, Box 57A, New York, NY 10032, USA.
| | - Elizabeth C Verna
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia College of Physicians and Surgeons, Columbia University Medical Center, 650 West 168 Street, New York, NY 10032, USA
| |
Collapse
|
232
|
Bodhankar SL, Kandhare AD, Patil MV. Ameliorative Effect of Alkaloidal Fraction of Leaves of Alstonia scholaris Against Acetic Acid Induced Colitis via Modulation of Oxido-nitrosative and Pro-inflammatory Cytokines. ACTA ACUST UNITED AC 2016. [DOI: 10.5567/pharmacologia.2016.170.181] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
233
|
Escobar S, Rocha A, Felip A, Carrillo M, Zanuy S, Kah O, Servili A. Leptin receptor gene in the European sea bass (Dicentrarchus labrax): Cloning, phylogeny, tissue distribution and neuroanatomical organization. Gen Comp Endocrinol 2016; 229:100-11. [PMID: 26979276 DOI: 10.1016/j.ygcen.2016.03.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 03/07/2016] [Accepted: 03/11/2016] [Indexed: 11/25/2022]
Abstract
In this study, we report the cloning of three transcripts for leptin receptor in the European sea bass, a marine teleost of economic interest. The two shortest variants, generated by different splice sites, encode all functional extracellular and intracellular domains but missed the transmembrane domain. The resulting proteins are therefore potential soluble binding proteins for leptin. The longest transcript (3605bp), termed sblepr, includes all the essential domains for binding and transduction of the signal. Thus, it is proposed as the ortholog for the human LEPR gene, the main responsible for leptin signaling. Phylogenetic analysis shows the sblepr clustered within the teleost leptin receptor group in 100% of the bootstrap replicates. The neuroanatomical localization of sblepr expressing cells has been assessed by in situ hybridization in brains of sea bass of both sexes during their first sexual maturation. At histological level, the distribution pattern of sblepr expressing cells in the brain shows no clear differences regarding sex or reproductive season. Transcripts of the sblepr have a widespread distribution throughout the forebrain and midbrain until the caudal portion of the hypothalamus. A high hybridization signal is detected in the telencephalon, preoptic area, medial basal and caudal hypothalamus and in the pituitary gland. In a more caudal region, sblepr expressing cells are identified in the longitudinal torus. The expression pattern observed for sblepr suggests that in sea bass, leptin is very likely to be involved in the control of food intake, energy reserves and reproduction.
Collapse
Affiliation(s)
- Sebastián Escobar
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Ana Rocha
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Alicia Felip
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Manuel Carrillo
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain
| | - Silvia Zanuy
- Department of Fish Physiology and Biotechnology, Instituto de Acuicultura de Torre la Sal (CSIC), Ribera de Cabanes s/n, 12595 Torre la Sal, Castellón, Spain.
| | - Olivier Kah
- Team NEED, Research Institute for Health, Environment and Occupation, INSERM U1085, SFR Biosit, Université de Rennes 1, France.
| | - Arianna Servili
- Team NEED, Research Institute for Health, Environment and Occupation, INSERM U1085, SFR Biosit, Université de Rennes 1, France
| |
Collapse
|
234
|
Chowen JA, Argente-Arizón P, Freire-Regatillo A, Frago LM, Horvath TL, Argente J. The role of astrocytes in the hypothalamic response and adaptation to metabolic signals. Prog Neurobiol 2016; 144:68-87. [PMID: 27000556 DOI: 10.1016/j.pneurobio.2016.03.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 02/09/2016] [Accepted: 03/04/2016] [Indexed: 12/19/2022]
Abstract
The hypothalamus is crucial in the regulation of homeostatic functions in mammals, with the disruption of hypothalamic circuits contributing to chronic conditions such as obesity, diabetes mellitus, hypertension, and infertility. Metabolic signals and hormonal inputs drive functional and morphological changes in the hypothalamus in attempt to maintain metabolic homeostasis. However, the dramatic increase in the incidence of obesity and its secondary complications, such as type 2 diabetes, have evidenced the need to better understand how this system functions and how it can go awry. Growing evidence points to a critical role of astrocytes in orchestrating the hypothalamic response to metabolic cues by participating in processes of synaptic transmission, synaptic plasticity and nutrient sensing. These glial cells express receptors for important metabolic signals, such as the anorexigenic hormone leptin, and determine the type and quantity of nutrients reaching their neighboring neurons. Understanding the mechanisms by which astrocytes participate in hypothalamic adaptations to changes in dietary and metabolic signals is fundamental for understanding the neuroendocrine control of metabolism and key in the search for adequate treatments of metabolic diseases.
Collapse
Affiliation(s)
- Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain.
| | - Pilar Argente-Arizón
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura M Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Tamas L Horvath
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Section of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, CIBER de Obesidad Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III, Madrid, Spain; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
235
|
Leptin signaling regulates glucose homeostasis, but not adipostasis, in the zebrafish. Proc Natl Acad Sci U S A 2016; 113:3084-9. [PMID: 26903647 DOI: 10.1073/pnas.1513212113] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Leptin is the primary adipostatic factor in mammals. Produced largely by adipocytes in proportion to total adipose mass, the hormone informs the brain regarding total energy stored as triglycerides in fat cells. The hormone acts on multiple circuits in the brain to regulate food intake, autonomic outflow, and endocrine function to maintain energy balance. In addition to regulating adipose mass, mammalian leptin also plays a role in the regulation of glucose homeostasis and as a gating factor in reproductive competence. Leptin-deficient mice and people exhibit early onset profound hyperphagia and obesity, diabetes, and infertility. Although leptin and the leptin receptor are found in fish, the hormone is not expressed in adipose tissue, but is found in liver and other tissues. Here, we show that adult zebrafish lacking a functional leptin receptor do not exhibit hyperphagia or increased adiposity, and exhibit normal fertility. However, leptin receptor-deficient larvae have increased numbers of β-cells and increased levels of insulin mRNA. Furthermore, larval zebrafish have been shown to exhibit β-cell hyperplasia in response to high fat feeding or peripheral insulin resistance, and we show here that leptin receptor is required for this response. Adult zebrafish also have increased levels of insulin mRNA and other alterations in glucose homeostasis. Thus, a role for leptin in the regulation of β-cell mass and glucose homeostasis appears to be conserved across vertebrates, whereas its role as an adipostatic factor is likely to be a secondary role acquired during the evolution of mammals.
Collapse
|
236
|
Utoyama M, Akieda-Asai S, Koda S, Nunoi H, Date Y. Role of the neural pathway from hindbrain to hypothalamus in the regulation of energy homeostasis in rats. Neurosci Lett 2016; 614:83-8. [PMID: 26773865 DOI: 10.1016/j.neulet.2016.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 01/22/2023]
Abstract
Recent evidence suggests that neural pathways from the hindbrain to the hypothalamus are important for informing the hypothalamus of the body's condition with regard to energy metabolism. Here we examined energy metabolism in rats with transections of the midbrain that severed the neural pathway from the hindbrain to the hypothalamus, and then investigated the levels of various molecules associated with control of energy metabolism in these rats. Food intake and body weight were higher in the midbrain-transected rats than in sham-operated rats. In addition, the midbrain-transected rats showed insulin resistance and hyperleptinemia. Furthermore, the hypothalamic mRNA levels of anorectic proopiomelanocortin and cocaine- and amphetamine-related transcript were significantly lower in midbrain-transected rats than in sham-operated rats. Our findings elucidate the mechanisms of food intake and energy balance from the perspective of multifactorial regulatory systems that underlie functions such as neurohormonal integration.
Collapse
Affiliation(s)
- Maiko Utoyama
- Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan; Department of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Sayaka Akieda-Asai
- Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Shuichi Koda
- Psychiartric & Neurological Disease Field, Asubio Pharma, Kobe 650-0047, Japan
| | - Hiroyuki Nunoi
- Department of Pediatrics, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yukari Date
- Frontier Science Research Center, University of Miyazaki, Miyazaki 889-1692, Japan.
| |
Collapse
|
237
|
Iwakura H, Dote K, Bando M, Koyama H, Hosoda K, Kangawa K, Nakao K. Establishment of Leptin-Responsive Cell Lines from Adult Mouse Hypothalamus. PLoS One 2016; 11:e0148639. [PMID: 26849804 PMCID: PMC4744015 DOI: 10.1371/journal.pone.0148639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 01/21/2016] [Indexed: 12/03/2022] Open
Abstract
Leptin resistance is considered to be the primary cause of obesity. However, the cause of leptin resistance remains incompletely understood, and there is currently no cure for the leptin-resistant state. In order to identify novel drug-target molecules that could overcome leptin resistance, it would be useful to develop in vitro assay systems for evaluating leptin resistance. In this study, we established immortalized adult mouse hypothalamus—derived cell lines, termed adult mouse hypothalamus (AMH) cells, by developing transgenic mice in which SV40 Tag was overexpressed in chromogranin A—positive cells in a tamoxifen-dependent manner. In order to obtain leptin-responsive clones, we selected clones based on the phosphorylation levels of STAT3 induced by leptin. The selected clones were fairly responsive to leptin in terms of STAT3, ERK, and Akt phosphorylation and induction of c-Fos mRNA induction. Pretreatment with leptin, insulin, and palmitate attenuated the c-Fos mRNA response to leptin, suggesting that certain aspects of leptin resistance might be reconstituted in this cellular model. These cell lines are useful tools for understanding the molecular nature of the signal disturbance in the leptin-resistant state and for identifying potential target molecules for drugs that relieve leptin resistance, although they have drawbacks including de-differentiated nature and lack of long-time stability.
Collapse
Affiliation(s)
- Hiroshi Iwakura
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- * E-mail:
| | - Katsuko Dote
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mika Bando
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Koyama
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Diabetes and Clinical Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kiminori Hosoda
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Human Health Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kangawa
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
238
|
Seroussi E, Cinnamon Y, Yosefi S, Genin O, Smith JG, Rafati N, Bornelöv S, Andersson L, Friedman-Einat M. Identification of the Long-Sought Leptin in Chicken and Duck: Expression Pattern of the Highly GC-Rich Avian leptin Fits an Autocrine/Paracrine Rather Than Endocrine Function. Endocrinology 2016; 157:737-51. [PMID: 26587783 DOI: 10.1210/en.2015-1634] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
More than 20 years after characterization of the key regulator of mammalian energy balance, leptin, we identified the leptin (LEP) genes of chicken (Gallus gallus) and duck (Anas platyrhynchos). The extreme guanine-cytosine content (∼70%), the location in a genomic region with low-complexity repetitive and palindromic sequence elements, the relatively low sequence conservation, and low level of expression have hampered the identification of these genes until now. In vitro-expressed chicken and duck leptins specifically activated signaling through the chicken leptin receptor in cell culture. In situ hybridization demonstrated expression of LEP mRNA in granular and Purkinje cells of the cerebellum, anterior pituitary, and in embryonic limb buds, somites, and branchial arches, suggesting roles in adult brain control of energy balance and during embryonic development. The expression patterns of LEP and the leptin receptor (LEPR) were explored in chicken, duck, and quail (Coturnix japonica) using RNA-sequencing experiments available in the Short Read Archive and by quantitative RT-PCR. In adipose tissue, LEP and LEPR were scarcely transcribed, and the expression level was not correlated to adiposity. Our identification of the leptin genes in chicken and duck genomes resolves a long lasting controversy regarding the existence of leptin genes in these species. This identification was confirmed by sequence and structural similarity, conserved exon-intron boundaries, detection in numerous genomic, and transcriptomic datasets and characterization by PCR, quantitative RT-PCR, in situ hybridization, and bioassays. Our results point to an autocrine/paracrine mode of action for bird leptin instead of being a circulating hormone as in mammals.
Collapse
Affiliation(s)
- Eyal Seroussi
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Yuval Cinnamon
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Sara Yosefi
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Olga Genin
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Julia Gage Smith
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Nima Rafati
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Susanne Bornelöv
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Leif Andersson
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| | - Miriam Friedman-Einat
- Agricultural Research Organization (E.S., Y.C., S.Y., O.G., J.G.-S., M.F.-E.), Volcani Center, 50250 Bet-Dagan, Israel; Department of Medical Biochemistry and Microbiology (N.R., S.B., L.A.), Uppsala University, SE-75123 Uppsala, Sweden; Department of Animal Breeding and Genetics (L.A.), Swedish University of Agricultural Sciences, SE-75007 Uppsala, Sweden; and Department of Veterinary Integrative Biosciences (L.A.), College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas 77843-4458
| |
Collapse
|
239
|
Rieg JAD, Chirasani VR, Koepsell H, Senapati S, Mahata SK, Rieg T. Regulation of intestinal SGLT1 by catestatin in hyperleptinemic type 2 diabetic mice. J Transl Med 2016; 96:98-111. [PMID: 26552046 PMCID: PMC4695279 DOI: 10.1038/labinvest.2015.129] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 09/10/2015] [Accepted: 09/24/2015] [Indexed: 01/23/2023] Open
Abstract
The small intestine is the major site for nutrient absorption that is critical in maintenance of euglycemia. Leptin, a key hormone involved in energy homeostasis, directly affects nutrient transport across the intestinal epithelium. Catestatin (CST), a 21-amino acid peptide derived from proprotein chromogranin A, has been shown to modulate leptin signaling. Therefore, we reasoned that leptin and CST could modulate intestinal Na(+)-glucose transporter 1 (SGLT1) expression in the context of obesity and diabetes. We found that hyperleptinemic db/db mice exhibit increased mucosal mass, associated with an enhanced proliferative response and decreased apoptosis in intestinal crypts, a finding absent in leptin-deficient ob/ob mice. Intestinal SGLT1 abundance was significantly decreased in hyperleptinemic but not leptin-deficient mice, indicating leptin regulation of SGLT1 expression. Phlorizin, a SGLT1/2 inhibitor, was without effect in an oral glucose tolerance test in db/db mice. The alterations in architecture and SGLT1 abundance were not accompanied by changes in the localization of intestinal alkaline phosphatase, indicating intact differentiation. Treatment of db/db mice with CST restored intestinal SGLT1 abundance and intestinal turnover, suggesting a cross-talk between leptin and CST, without affecting plasma leptin levels. Consistent with this hypothesis, we identified structural homology between CST and the AB-loop of leptin and protein-protein docking revealed binding of CST and leptin with the Ig-like binding site-III of the leptin receptor. In summary, downregulation of SGLT1 in an obese type 2 diabetic mouse model with hyperleptinemia is presumably mediated via the short form of the leptin receptor and reduces overt hyperglycemia.
Collapse
Affiliation(s)
- Jessica A. Dominguez Rieg
- Department of Basic Sciences, Bastyr University California, San Diego, CA, USA,VA San Diego Healthcare System, San Diego, California; CA, USA
| | | | - Hermann Koepsell
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs-Institute, University of Würzburg, Würzburg, Germany
| | - Sanjib Senapati
- Department of Biotechnology, Institute of Technology Madras, Chennai, India
| | - Sushil K. Mahata
- VA San Diego Healthcare System, San Diego, California; CA, USA,Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Timo Rieg
- VA San Diego Healthcare System, San Diego, California; CA, USA,Division of Nephrology-Hypertension, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
240
|
|
241
|
Lampropoulou E, Lymperopoulou A, Charonis A. Reduced expression of ERp46 under diabetic conditions in β-cells and the effect of liraglutide. Metabolism 2016; 65:7-15. [PMID: 26683792 DOI: 10.1016/j.metabol.2015.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/11/2015] [Accepted: 09/12/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Diabetes mellitus is characterized by peripheral insulin resistance, hyperglycemia and defective insulin secretion. Insulin producing pancreatic β-cells are equipped with a highly developed endoplasmic reticulum (ER) and thus are affected by ER stress under hyperglycemic conditions. We have previously studied the influence of high glucose on cultured β-cells in vitro. Proteomic analysis revealed a number of proteins involved in glucose toxicity, while further biochemical analysis identified the endoplasmic reticulum protein ERp46 as a molecule with a possible role in insulin production at the post-translational level. In addition, the involvement of incretin hormone glucagon-like peptide 1 (GLP-1) in diabetes proposes that incretin-mimetic compounds may be among the optimal choices in future therapeutic interventions; therefore their effects on various aspects of the pathogenesis of diabetes mellitus should be explored in detail. Based on the above, we examined the possible involvement of ERp46 in insulin production and the effect of the GLP-1 analogue liraglutide on the expression of ERp46 in vitro, in β-cells cultured under high glucose conditions and in vivo, in the mouse db/db diabetic model, where pronounced hyperglycemia is a key characteristic. RESULTS Confocal microscopy revealed areas of co-localization of ERp46 and pro-insulin in pancreatic islets. In order to explore the possible interaction between ERp46 and insulin immunoprecipitation was used. In extracts from cultured β-cells, antibodies against pro-insulin co-precipitated ERp46 and antibodies against ERp46 co-precipitated pro-insulin, as shown by Western blotting. Furthermore, data from a proximity ligation assay positioned these two molecules closer than 30nm in distance. When pancreatic β-cells were cultured in high glucose conditions they exhibited a decrease in ERp46 expression, while treatment with the GLP-1 analogue liraglutide restored ERp46 levels, leading to a significant increase of ERp46 in comparison to hyperglycemic conditions. In the diabetic mouse model db(-)/db, ERp46 expression was reduced in pancreatic islets, as documented by morphological and biochemical techniques. This decrease was abolished after treatment with the GLP-1 analogue in a dose-dependent manner. In an attempt to understand the underlying mechanism, we examined the sequence of the promoter of ERp46 and found consensus motifs that can be recognized by transcription factors ATF6 and XBP1. Subsequently, we performed chromatin immunoprecipitation assay and demonstrated that treatment of β-TC-6 cells with 25mmol/L glucose decreases gradually the binding enrichment of ATF6 and XBP1 in ERp46 gene promoter. CONCLUSIONS We propose that since ERp46 is a member of the disulfide isomerases family, it is likely to play a key role in insulin biosynthesis and its reduction under high glucose conditions may be a novel contributor to the glucotoxicity of β-cells. In addition, the GLP-1 analogue liraglutide seems to interfere in this process and may exert its beneficial effects in diabetes by affecting insulin production via restoration of ERp46 expression.
Collapse
Affiliation(s)
- Eugenia Lampropoulou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efessiou 4, Athens 115 27, Greece.
| | - Anna Lymperopoulou
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efessiou 4, Athens 115 27, Greece.
| | - Aristidis Charonis
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Soranou Efessiou 4, Athens 115 27, Greece.
| |
Collapse
|
242
|
Hoang Do O, Thorn P. Insulin secretion from beta cells within intact islets: location matters. Clin Exp Pharmacol Physiol 2015; 42:406-14. [PMID: 25676261 PMCID: PMC4418378 DOI: 10.1111/1440-1681.12368] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 12/21/2014] [Accepted: 01/06/2015] [Indexed: 12/17/2022]
Abstract
The control of hormone secretion is central to body homeostasis, and its dysfunction is important in many diseases. The key cellular steps that lead to hormone secretion have been identified, and the stimulus-secretion pathway is understood in outline for many endocrine cells. In the case of insulin secretion from pancreatic beta cells, this pathway involves the uptake of glucose, cell depolarization, calcium entry, and the triggering of the fusion of insulin-containing granules with the cell membrane. The wealth of information on the control of insulin secretion has largely been obtained from isolated single-cell studies. However, physiologically, beta cells exist within the islets of Langerhans, with structural and functional specializations that are not preserved in single-cell cultures. This review focuses on recent work that is revealing distinct aspects of insulin secretion from beta cells within the islet.
Collapse
Affiliation(s)
- Oanh Hoang Do
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Qld, Australia
| | | |
Collapse
|
243
|
Effects of angiotensin II on leptin and downstream leptin signaling in the carotid body during acute intermittent hypoxia. Neuroscience 2015; 310:430-41. [DOI: 10.1016/j.neuroscience.2015.09.066] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 08/11/2015] [Accepted: 09/24/2015] [Indexed: 11/19/2022]
|
244
|
Preliminary Characterization of a Leptin Receptor Knockout Rat Created by CRISPR/Cas9 System. Sci Rep 2015; 5:15942. [PMID: 26537785 PMCID: PMC4633582 DOI: 10.1038/srep15942] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 09/29/2015] [Indexed: 12/28/2022] Open
Abstract
Leptin receptor, which is encoded by the diabetes (db) gene and is highly expressed in the choroid plexus, regulatesenergy homeostasis, the balance between food intake and energy expenditure, fertility and bone mass. Here, using CRISPR/Cas9 technology, we created the leptin receptor knockout rat. Homozygous leptin receptor null rats are characterized by obesity, hyperphagia, hyperglycemia, glucose intolerance, hyperinsulinemia and dyslipidemia. Due to long-term poor glycemic control, the leptin receptor knockout rats also develop some diabetic complications such as pancreatic, hepatic and renal lesions. In addition, the leptin receptor knockout rats show a significant decrease in bone volume and bone mineral density of the femur compared with their wild-type littermates. Our model has rescued some deficiency of the existing rodent models, such as the transient hyperglycemia of db/db mice in the C57BL/6J genetic background and the delayed onset of glucose intolerance in the Zucker rats, and it is proven to be a useful animal model for biomedical and pharmacological research on obesity and diabetes.
Collapse
|
245
|
Yoshida R, Noguchi K, Shigemura N, Jyotaki M, Takahashi I, Margolskee RF, Ninomiya Y. Leptin Suppresses Mouse Taste Cell Responses to Sweet Compounds. Diabetes 2015; 64:3751-62. [PMID: 26116698 PMCID: PMC4876703 DOI: 10.2337/db14-1462] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 06/16/2015] [Indexed: 01/19/2023]
Abstract
Leptin is known to selectively suppress neural and behavioral responses to sweet-tasting compounds. However, the molecular basis for the effect of leptin on sweet taste is not known. Here, we report that leptin suppresses sweet taste via leptin receptors (Ob-Rb) and KATP channels expressed selectively in sweet-sensitive taste cells. Ob-Rb was more often expressed in taste cells that expressed T1R3 (a sweet receptor component) than in those that expressed glutamate-aspartate transporter (a marker for Type I taste cells) or GAD67 (a marker for Type III taste cells). Systemically administered leptin suppressed taste cell responses to sweet but not to bitter or sour compounds. This effect was blocked by a leptin antagonist and was absent in leptin receptor-deficient db/db mice and mice with diet-induced obesity. Blocking the KATP channel subunit sulfonylurea receptor 1, which was frequently coexpressed with Ob-Rb in T1R3-expressing taste cells, eliminated the effect of leptin on sweet taste. In contrast, activating the KATP channel with diazoxide mimicked the sweet-suppressing effect of leptin. These results indicate that leptin acts via Ob-Rb and KATP channels that are present in T1R3-expressing taste cells to selectively suppress their responses to sweet compounds.
Collapse
Affiliation(s)
- Ryusuke Yoshida
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Kenshi Noguchi
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan Section of Orthodontics and Dentofacial Orthopedics, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Noriatsu Shigemura
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masafumi Jyotaki
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | - Ichiro Takahashi
- Section of Orthodontics and Dentofacial Orthopedics, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan
| | | | - Yuzo Ninomiya
- Section of Oral Neuroscience, Graduate School of Dental Science, Kyushu University, Fukuoka, Japan Division of Sensory Physiology, Research and Development Center for Taste and Odor Sensing, Kyushu University, Fukuoka, Japan
| |
Collapse
|
246
|
Friedman JM. Editorial. Life Sci 2015; 140:1-2. [DOI: 10.1016/j.lfs.2015.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
247
|
Jo YS, Ryu D, Maida A, Wang X, Evans RM, Schoonjans K, Auwerx J. Phosphorylation of the nuclear receptor corepressor 1 by protein kinase B switches its corepressor targets in the liver in mice. Hepatology 2015; 62:1606-18. [PMID: 25998209 PMCID: PMC4618256 DOI: 10.1002/hep.27907] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 05/17/2015] [Indexed: 01/22/2023]
Abstract
UNLABELLED Nuclear receptor corepressor 1 (NCoR1) is a transcriptional coregulator that has wide-ranging effects on gene expression patterns. In the liver, NCoR1 represses lipid synthesis in the fasting state, whereas it inhibits activation of peroxisome proliferator-activated receptor alpha (PPARα) upon feeding, thereby blunting ketogenesis. Here, we show that insulin by activation of protein kinase B induces phosphorylation of NCoR1 on serine 1460, which selectively favors its interaction with PPARα and estrogen-related receptor alpha (ERRα) over liver X receptor alpha (LXRα). Phosphorylation of NCoR1 on S1460 selectively derepresses LXRα target genes, resulting in increased lipogenesis, whereas, at the same time, it inhibits PPARα and ERRα targets, thereby attenuating oxidative metabolism in the liver. Phosphorylation-gated differential recruitment of NCoR1 to different nuclear receptors explains the apparent paradox that liver-specific deletion of NCoR1 concurrently induces both lipogenesis and oxidative metabolism owing to a global derepression of LXRα, PPARα, and ERRα activity. CONCLUSION Phosphorylation-mediated recruitment switch of NCoR1 between nuclear receptor subsets provides a mechanism by which corepressors can selectively modulate liver energy metabolism during the fasting-feeding transition.
Collapse
Affiliation(s)
- Young Suk Jo
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
- Division of Endocrinology, Department of Internal Medicine, Yonsei University College of Medicine, 50 Yonsei-ro Seodaemun-gu, Seoul, 120-752, South Korea
| | - Dongryeol Ryu
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Adriano Maida
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Xu Wang
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Ronald M. Evans
- Gene Expression Laboratory, Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Kristina Schoonjans
- Metabolic Signaling, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| |
Collapse
|
248
|
Hart RA, Dobos RC, Agnew LL, Tellam RL, McFarlane JR. Pharmacokinetics of leptin in female mice. Physiol Res 2015; 65:311-20. [PMID: 26447522 DOI: 10.33549/physiolres.933053] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Pharmacokinetics of leptin in mammals has received limited attention and only one study has examined more than two time points and this was in ob/ob mice. This study is the first to observe the distribution of leptin over a time course in female mice. A physiologic dose (12 ng) of radiolabelled leptin was injected in adult female mice via the lateral tail vein and tissues were dissected out and measured for radioactivity over a time course up to two hours. Major targets for administered leptin included the liver, kidneys, gastrointestinal tract and the skin while the lungs had high concentrations of administered leptin per gram of tissue. Leptin was also found to enter the lumen of the digestive tract intact from the plasma. Very little of the dose (<1 %) was recovered from the brain at any time. Consequently we confirm that the brain is not a major target for leptin from the periphery, although it may be very sensitive to leptin that does get to the hypothalamus. Several of the major targets (GI tract, skin and lungs) for leptin form the interface for the body with the environment, and given the ability of leptin to modulate immune function, this may represent a priming effect for tissues to respond to damage and infection.
Collapse
Affiliation(s)
- R A Hart
- Centre for Bioactive Discovery in Health and Ageing, University of New England, Armidale, NSW 2351, Australia.
| | | | | | | | | |
Collapse
|
249
|
Szewczyk-Golec K, Woźniak A, Reiter RJ. Inter-relationships of the chronobiotic, melatonin, with leptin and adiponectin: implications for obesity. J Pineal Res 2015; 59:277-91. [PMID: 26103557 DOI: 10.1111/jpi.12257] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 06/19/2015] [Indexed: 12/15/2022]
Abstract
Obesity and its medical complications represent a significant problem throughout the world. In recent decades, mechanisms underlying the progression of obesity have been intensively examined. The involvement of both the behavioral aspects, such as calorie-rich diet, low physical activity and sleep deprivation, and the intrinsic factors, including adipose tissue deregulation, chronic inflammation, oxidative stress, and chronodisruption, has been identified. The circadian disturbances of the adipose tissue endocrine function have been correlated with obesity. Leptin and adiponectin are adipokines strongly associated with glucose and lipid metabolism and with energy balance. Their synthesis and secretion display circadian rhythms that are disturbed in the obese state. Hyperleptinemia resulting in leptin resistance, and hypo-adiponectinemia have been linked to the pathophysiology of the obesity-related disorders. A deficiency of melatonin, one of the consequences of sleep deprivation, has also been demonstrated to correlate with obesity. Melatonin is a pineal secretory product involved in numerous actions, such as regulation of internal biological clocks and energy metabolism, and it functions as an antioxidant and as an anti-inflammatory agent. There exists a substantial amount of evidence supporting the beneficial effects of melatonin supplementation on obesity and its complications. In the current review, the results of studies related to the interactions between melatonin, and both leptin and adiponectin are discussed. Despite the existence of some inconsistencies, melatonin has been found to normalize the expression and secretion patterns of both adipokines. These results support the concept of melatonin as a potential therapeutic agent for obesity and related disorders.
Collapse
Affiliation(s)
- Karolina Szewczyk-Golec
- The Chair of Medical Biology, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Alina Woźniak
- The Chair of Medical Biology, Nicolaus Copernicus University, Ludwik Rydygier Collegium Medicum, Bydgoszcz, Poland
| | - Russel J Reiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| |
Collapse
|
250
|
Candlish M, Angelis RD, Götz V, Boehm U. Gene Targeting in Neuroendocrinology. Compr Physiol 2015; 5:1645-76. [DOI: 10.1002/cphy.c140079] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|