51
|
Ochsner SA, McKenna NJ. No Dataset Left Behind: Mechanistic Insights into Thyroid Receptor Signaling Through Transcriptomic Consensome Meta-Analysis. Thyroid 2020; 30:621-639. [PMID: 31910096 PMCID: PMC7187985 DOI: 10.1089/thy.2019.0307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background: Discovery-scale omics datasets relevant to thyroid receptors (TRs) and their physiological and synthetic bioactive small-molecule ligands allow for genome-wide interrogation of TR-regulated genes. These datasets have considerable collective value as a reference resource to allow researchers to routinely generate hypotheses addressing the mechanisms underlying the cell biology and physiology of TR signaling in normal and disease states. Methods: Here, we searched the Gene Expression Omnibus database to identify a population of publicly archived transcriptomic datasets involving genetic or pharmacological manipulation of either TR isoform in a mouse tissue or cell line. After initial quality control, samples were organized into contrasts (experiments), and transcript differential expression values and associated measures of significance were generated and committed to a consensome (for consensus omics) meta-analysis pipeline. To gain insight into tissue-selective functions of TRs, we generated liver- and central nervous system (CNS)-specific consensomes and identified evidence for genes that were selectively responsive to TR signaling in each organ. Results: The TR transcriptomic consensome ranks genes based on the frequency of their significant differential expression over the entire group of experiments. The TR consensome assigns elevated rankings both to known TR-regulated genes and to genes previously uncharacterized as TR-regulated, which shed mechanistic light on known cellular and physiological roles of TR signaling in different organs. We identify evidence for unreported genomic targets of TR signaling for which it exhibits strikingly distinct regulatory preferences in the liver and CNS. Moreover, the intersection of the TR consensome with consensomes for other cellular receptors sheds light on transcripts potentially mediating crosstalk between TRs and these other signaling paradigms. Conclusions: The mouse TR datasets and consensomes are freely available in the Signaling Pathways Project website for hypothesis generation, data validation, and modeling of novel mechanisms of TR regulation of gene expression. Our results demonstrate the insights into the mechanistic basis of thyroid hormone action that can arise from an ongoing commitment on the part of the research community to the deposition of discovery-scale datasets.
Collapse
Affiliation(s)
- Scott A. Ochsner
- The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Neil J. McKenna
- The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
- Address correspondence to: Neil J. McKenna, PhD, The Signaling Pathways Project, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
52
|
Iqbal J, Mascareno E, Chua S, Hussain MM. Leptin-mediated differential regulation of microsomal triglyceride transfer protein in the intestine and liver affects plasma lipids. J Biol Chem 2020; 295:4101-4113. [PMID: 32047110 PMCID: PMC7105304 DOI: 10.1074/jbc.ra119.011881] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/06/2020] [Indexed: 11/06/2022] Open
Abstract
The hormone leptin regulates fat storage and metabolism by signaling through the brain and peripheral tissues. Lipids delivered to peripheral tissues originate mostly from the intestine and liver via synthesis and secretion of apolipoprotein B (apoB)-containing lipoproteins. An intracellular chaperone, microsomal triglyceride transfer protein (MTP), is required for the biosynthesis of these lipoproteins, and its regulation determines fat mobilization to different tissues. Using cell culture and animal models, here we sought to identify the effects of leptin on MTP expression in the intestine and liver. Leptin decreased MTP expression in differentiated intestinal Caco-2 cells, but increased expression in hepatic Huh7 cells. Similarly, acute and chronic leptin treatment of chow diet-fed WT mice decreased MTP expression in the intestine, increased it in the liver, and lowered plasma triglyceride levels. These leptin effects required the presence of leptin receptors (LEPRs). Further experiments also suggested that leptin interacted with long-form LEPR (ObRb), highly expressed in the intestine, to down-regulate MTP. In contrast, in the liver, leptin interacted with short-form LEPR (ObRa) to increase MTP expression. Mechanistic experiments disclosed that leptin activates signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase (MAPK) signaling pathways in intestinal and hepatic cells, respectively, and thereby regulates divergent MTP expression. Our results also indicated that leptin-mediated MTP regulation in the intestine affects plasma lipid levels. In summary, our findings suggest that leptin regulates MTP expression differentially by engaging with different LEPR types and activating distinct signaling pathways in intestinal and hepatic cells.
Collapse
Affiliation(s)
- Jahangir Iqbal
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203; King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, Eastern Region, Ministry of National Guard Health Affairs, Al Ahsa 31982, Saudi Arabia.
| | - Eduardo Mascareno
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203
| | - Streamson Chua
- Department of Medicine and Neuroscience, Albert Einstein College of Medicine, Bronx, New York 10461
| | - M Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York 11203; Department of Foundations of Medicine, NYU Long Island School of Medicine and Diabetes and Obesity Research Center, NYU Winthrop Research Institute, Mineola, New York 11501; Veterans Affairs New York Harbor Healthcare System, Brooklyn, New York 11209.
| |
Collapse
|
53
|
Martinez-Pena Y Valenzuela I, Akaaboune M. The disassembly of the neuromuscular synapse in high-fat diet-induced obese male mice. Mol Metab 2020; 36:100979. [PMID: 32283080 PMCID: PMC7182767 DOI: 10.1016/j.molmet.2020.100979] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Objective A sustained high fat diet in mice mimics many features of human obesity. We used male and female Non-Swiss albino mice to investigate the impact of short and long-term high-fat diet-(HFD)-induced obesity on the peripheral neuromuscular junction (NMJ) and whether obesity-related synaptic structural alterations were reversible after switching obese mice from HFD to a standard fat diet (SD). Methods HFD-induced obese and age-matched control mice fed SD were used. We carried out in vivo time lapse imaging to monitor changes of synapses over time, quantitative fluorescence imaging to study the regulation of acetylcholine receptor number and density at neuromuscular junctions, and high resolution confocal microscope to study structural alterations in both the pre- and postsynaptic apparatus. Results Time-lapse imaging in vivo over a 9 month period revealed that NMJs of HFD obese male mice display a variety of obesity-related structural alterations, including the disappearance of large synaptic areas, significant reduction in the density/number of nicotinic acetylcholine receptor (AChRs), abnormal distribution of AChRs, high turnover rate of AChRs, retraction of axons from lost postsynaptic sites, and partially denervated synapses. The severity of these synaptic alterations is associated with the duration of obesity. However, no substantial alterations were observed at NMJs of age-matched HFD obese female mice or male mice fed with a standard or low fat diet. Intriguingly, when obese male mice were switched from HFD to a standard diet, receptor density and the abnormal pattern of AChR distribution were completely reversed to normal, whereas lost synaptic structures were not restored. Conclusions These results show that the obese male mice are more vulnerable than female mice to the impacts of long-term HFD on the NMJ damage and provide evidence that diet restriction can partially reverse obesity-related synaptic changes. Neuromuscular junctions of High-fat induced obese male mice display a variety of obesity-related structural alterations. The severity of alterations in neuromuscular junction morphology is associated with the duration of obesity. Neuromuscular junctions of High-fat diet induced obese female mice display no substantial morphological changes. Not all obesity-related synaptic alterations were reversible after switching male mice from High-fat diet to standard diet. Obese male mice are more vulnerable than female mice to the impacts of long-term HFD on the neuromuscular junction damage.
Collapse
Affiliation(s)
| | - Mohammed Akaaboune
- Department of Molecular, Cellular, and Developmental Biology, USA; Program in Neuroscience, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
54
|
Seoane-Collazo P, Martínez-Sánchez N, Milbank E, Contreras C. Incendiary Leptin. Nutrients 2020; 12:nu12020472. [PMID: 32069871 PMCID: PMC7071158 DOI: 10.3390/nu12020472] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/06/2020] [Accepted: 02/08/2020] [Indexed: 02/08/2023] Open
Abstract
Leptin is a hormone released by adipose tissue that plays a key role in the control of energy homeostasis through its binding to leptin receptors (LepR), mainly expressed in the hypothalamus. Most scientific evidence points to leptin’s satiating effect being due to its dual capacity to promote the expression of anorexigenic neuropeptides and to reduce orexigenic expression in the hypothalamus. However, it has also been demonstrated that leptin can stimulate (i) thermogenesis in brown adipose tissue (BAT) and (ii) the browning of white adipose tissue (WAT). Since the demonstration of the importance of BAT in humans 10 years ago, its study has aroused great interest, mainly in the improvement of obesity-associated metabolic disorders through the induction of thermogenesis. Consequently, several strategies targeting BAT activation (mainly in rodent models) have demonstrated great potential to improve hyperlipidemias, hepatic steatosis, insulin resistance and weight gain, leading to an overall healthier metabolic profile. Here, we review the potential therapeutic ability of leptin to correct obesity and other metabolic disorders, not only through its satiating effect, but by also utilizing its thermogenic properties.
Collapse
Affiliation(s)
- Patricia Seoane-Collazo
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Noelia Martínez-Sánchez
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, UK
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| | - Edward Milbank
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15782 Santiago de Compostela, Spain;
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706 Santiago de Compostela, Spain
| | - Cristina Contreras
- Department of Physiology, Pharmacy School, Complutense University of Madrid, 28040 Madrid, Spain
- Correspondence: (P.S.-C.); (N.M.-S.); (C.C.); Tel.: +81-298-533-301 (P.S.-C.); +34-913-941-650 (N.M.-S.); +44-01865285890 (C.C.)
| |
Collapse
|
55
|
Chohnan S, Matsuno S, Shimizu K, Tokutake Y, Kohari D, Toyoda A. Coenzyme A and Its Thioester Pools in Obese Zucker and Zucker Diabetic Fatty Rats. Nutrients 2020; 12:E417. [PMID: 32041091 PMCID: PMC7071249 DOI: 10.3390/nu12020417] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/29/2022] Open
Abstract
Feeding behavior is closely related to hypothalamic malonyl-CoA level in the brain and diet-induced obesity affects total CoA pools in liver. Herein, we performed a comprehensive analysis of the CoA pools formed in thirteen tissues of Zucker and Zucker diabetic fatty (ZDF) rats. Hypothalamic malonyl-CoA levels in obese rats remained low and were almost the same as those of lean rats, despite obese rats having much higher content of leptin, insulin, and glucose in their sera. Regardless of the fa-genotypes, larger total CoA pools were formed in the livers of ZDF rats and the size of hepatic total CoA pools in Zucker rats showed almost one tenth of the size of ZDF rats. The decreased total CoA pool sizes in Zucker rats was observed in the brown adipose tissues, while ZDF-fatty rats possessed 6% of total CoA pool in the lean rats in response to fa deficiency. This substantially lower CoA content in the obese rats would be disadvantageous to non-shivering thermogenesis. Thus, comparing the intracellular CoA behaviors between Zucker and ZDF rats, as well as the lean and fatty rats of each strain would help to elucidate features of obesity and type 2 diabetes in combination with result (s) of differential gene expression analysis and/or comparative genomics.
Collapse
Affiliation(s)
- Shigeru Chohnan
- Department of Food and Life Sciences, Ibaraki University College of Agriculture, 3-21-1 Chuo, Ami, Ibaraki 300-0393, Japan; (S.M.); (K.S.); (D.K.); (A.T.)
| | - Shiori Matsuno
- Department of Food and Life Sciences, Ibaraki University College of Agriculture, 3-21-1 Chuo, Ami, Ibaraki 300-0393, Japan; (S.M.); (K.S.); (D.K.); (A.T.)
| | - Kei Shimizu
- Department of Food and Life Sciences, Ibaraki University College of Agriculture, 3-21-1 Chuo, Ami, Ibaraki 300-0393, Japan; (S.M.); (K.S.); (D.K.); (A.T.)
| | - Yuka Tokutake
- Department of Applied Life Science, United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai, Fuchu, Tokyo 183-8509, Japan;
| | - Daisuke Kohari
- Department of Food and Life Sciences, Ibaraki University College of Agriculture, 3-21-1 Chuo, Ami, Ibaraki 300-0393, Japan; (S.M.); (K.S.); (D.K.); (A.T.)
| | - Atsushi Toyoda
- Department of Food and Life Sciences, Ibaraki University College of Agriculture, 3-21-1 Chuo, Ami, Ibaraki 300-0393, Japan; (S.M.); (K.S.); (D.K.); (A.T.)
| |
Collapse
|
56
|
Iqbal N, Zhu LI, Chua SC. Neuronal Cell Cycle Events Link Caloric Intake to Obesity. Trends Endocrinol Metab 2020; 31:46-52. [PMID: 31629614 PMCID: PMC7064044 DOI: 10.1016/j.tem.2019.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/03/2019] [Accepted: 09/05/2019] [Indexed: 02/03/2023]
Abstract
Obesity is a neurological disorder that operates by favoring energy storage within adipose depots and increased caloric intake. Most cases of human obesity are acquired without any underlying genetic basis. Here, we suggest that obesity can impair the function of some hypothalamic neurons critical to body weight regulation. Genetic ablation of the retinoblastoma (Rb) gene within pro-opiomelanocortin (POMC) neurons leads to death of the neurons and subsequent obesity. The Rb protein (pRb), a key inhibitor of the cell cycle, can also be inactivated by cyclin dependent kinase (CDK)-mediated phosphorylation. Extensive development led to the production of FDA-approved CDK4/6 inhibitors. Based on our own results, we propose that maintaining or re-instating pRb function using CDK4/6 inhibitors are potentially effective treatments of diet-induced obesity (DIO).
Collapse
Affiliation(s)
- Niloy Iqbal
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10462, USA
| | - LIang Zhu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10462, USA
| | - Streamson C Chua
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10462, USA; Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10462, USA.
| |
Collapse
|
57
|
Abstract
Many animal models that are currently used in appetite and obesity research share at least some main features of human obesity and its comorbidities. Hence, even though no animal model replicates all aspects of "common" human obesity, animal models are imperative in studying the control of energy balance and reasons for its imbalance that may eventually lead to overt obesity. The most frequently used animal models are small rodents that may be based on mutations or manipulations of individual or several genes and on the exposure to obesogenic diets or other manipulations that predispose the animals to gaining or maintaining excessive weight. Characteristics include hyperphagia or changes in energy metabolism and at least in some models the frequent comorbidities of obesity, like hyperglycemia, insulin resistance, or diabetes-like syndromes. Some of the most frequently used animal models of obesity research involve animals with monogenic mutations of the leptin pathway which in fact are useful to study specific mechanistic aspects of eating controls, but typically do not recapitulate "common" obesity in the human population. Hence, this review will mention advantages and disadvantages of respective animal models in order to build a basis for the most appropriate use in biomedical research.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland.
| |
Collapse
|
58
|
Fetissov SO, Meguid MM. Food intake and meal pattern in response to hyperosmotic-induced dehydration in obese and lean Zucker rats. Nutrition 2020; 70S:100011. [DOI: 10.1016/j.nutx.2020.100011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/16/2020] [Accepted: 06/21/2020] [Indexed: 10/23/2022]
|
59
|
Eftekhari S, Westgate CSJ, Uldall MS, Jensen RH. Preclinical update on regulation of intracranial pressure in relation to idiopathic intracranial hypertension. Fluids Barriers CNS 2019; 16:35. [PMID: 31767019 PMCID: PMC6878629 DOI: 10.1186/s12987-019-0155-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Elevated intracranial pressure (ICP) is observed in association with a range of brain disorders. One of these challenging disorders is idiopathic intracranial hypertension (IIH), characterized by raised ICP of unknown cause with significant morbidity and limited therapeutic options. In this review, special focus is put on the preclinical research performed in order to understand the pathophysiology behind ICP regulation and IIH. This includes cerebrospinal fluid dynamics, molecular mechanisms underlying disturbances in brain fluids leading to elevated ICP, role of obesity in IIH, development of an IIH model and ICP measurements in rodents. The review also discusses existing and new drug targets for IIH that have been evaluated in vivo. CONCLUSIONS ICP monitoring in rodents is challenging and different methods have been applied. Some of these methods are invasive, depend on use of anesthesia and only allow short-term monitoring. Long-term ICP recordings are needed to study IIH but existing methods are hampered by several limitations. As obesity is one of the most common risk factors for IIH, a rodent obese model has been developed that mimics some key aspects of IIH. The most commonly used drugs for IIH have been evaluated in vivo for their efficacy at lowering ICP in the existing animal models. These studies suggest these drugs, including acetazolamide, might have limited or no reducing effect on ICP. Two drug targets that can impact ICP in healthy rodents are topiramate and a glucagon-like peptide-1 receptor (GLP-1R) agonist. However, it remains to evaluate their effect in an IIH model with more precise and valid ICP monitoring system. Therefore, continued evaluation in the preclinical research with refined tools is of great importance to further understand the pathophysiology behind disorders with raised ICP and to explore new drug targets.
Collapse
Affiliation(s)
- Sajedeh Eftekhari
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600 Glostrup, Denmark
| | - Connar Stanley James Westgate
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600 Glostrup, Denmark
| | - Maria Schmidt Uldall
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600 Glostrup, Denmark
| | - Rigmor Hoejland Jensen
- Danish Headache Center, Department of Neurology, Glostrup Research Institute, Rigshospitalet-Glostrup, University of Copenhagen, Nordstjernevej 42, 2600 Glostrup, Denmark
| |
Collapse
|
60
|
Chellappa K, Brinkman JA, Mukherjee S, Morrison M, Alotaibi MI, Carbajal KA, Alhadeff AL, Perron IJ, Yao R, Purdy CS, DeFelice DM, Wakai MH, Tomasiewicz J, Lin A, Meyer E, Peng Y, Arriola Apelo SI, Puglielli L, Betley JN, Paschos GK, Baur JA, Lamming DW. Hypothalamic mTORC2 is essential for metabolic health and longevity. Aging Cell 2019; 18:e13014. [PMID: 31373126 PMCID: PMC6718533 DOI: 10.1111/acel.13014] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 05/26/2019] [Accepted: 07/03/2019] [Indexed: 12/16/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is an evolutionarily conserved protein kinase that regulates growth and metabolism. mTOR is found in two protein complexes, mTORC1 and mTORC2, that have distinct components and substrates and are both inhibited by rapamycin, a macrolide drug that robustly extends lifespan in multiple species including worms and mice. Although the beneficial effect of rapamycin on longevity is generally attributed to reduced mTORC1 signaling, disruption of mTORC2 signaling can also influence the longevity of worms, either positively or negatively depending on the temperature and food source. Here, we show that loss of hypothalamic mTORC2 signaling in mice decreases activity level, increases the set point for adiposity, and renders the animals susceptible to diet-induced obesity. Hypothalamic mTORC2 signaling normally increases with age, and mice lacking this pathway display higher fat mass and impaired glucose homeostasis throughout life, become more frail with age, and have decreased overall survival. We conclude that hypothalamic mTORC2 is essential for the normal metabolic health, fitness, and lifespan of mice. Our results have implications for the use of mTORC2-inhibiting pharmaceuticals in the treatment of brain cancer and diseases of aging.
Collapse
Affiliation(s)
- Karthikeyani Chellappa
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Jacqueline A. Brinkman
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Sarmistha Mukherjee
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Mark Morrison
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Mohammed I. Alotaibi
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Endocrinology and Reproductive Physiology Graduate Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Kathryn A. Carbajal
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Amber L. Alhadeff
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Isaac J. Perron
- Center for Sleep and Circadian Neurobiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Rebecca Yao
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Cole S. Purdy
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Denise M. DeFelice
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Matthew H. Wakai
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
| | - Jay Tomasiewicz
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Amy Lin
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Emma Meyer
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Yajing Peng
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Sebastian I. Arriola Apelo
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Department of Dairy ScienceUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - Luigi Puglielli
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Waisman CenterUniversity of Wisconsin‐MadisonMadisonWIUSA
| | - J. Nicholas Betley
- Department of Biology, School of Arts and SciencesUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Georgios K. Paschos
- Center for Sleep and Circadian Neurobiology, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
- The Institute for Translational Medicine and Therapeutics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Joseph A. Baur
- Department of Physiology and Institute for Diabetes, Obesity and Metabolism, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Dudley W. Lamming
- Department of MedicineUniversity of Wisconsin‐MadisonMadisonWIUSA
- William S. Middleton Memorial Veterans HospitalMadisonWIUSA
- Endocrinology and Reproductive Physiology Graduate Training ProgramUniversity of Wisconsin‐MadisonMadisonWIUSA
| |
Collapse
|
61
|
Liu S, Li X, Yang J, Zhu R, Fan Z, Xu X, Feng W, Cui J, Sun J, Liu M. Misfolded proinsulin impairs processing of precursor of insulin receptor and insulin signaling in β cells. FASEB J 2019; 33:11338-11348. [PMID: 31311313 PMCID: PMC6766638 DOI: 10.1096/fj.201900442r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Insulin resistance in classic insulin-responsive tissues is a hallmark of type 2 diabetes (T2D). However, the pathologic significance of β-cell insulin resistance and the underlying mechanisms contributing to defective insulin signaling in β cells remain largely unknown. Emerging evidence indicates that proinsulin misfolding is not only the molecular basis of mutant INS-gene–induced diabetes of youth (MIDY) but also an important contributor in the development and progression of T2D. However, the molecular basis of β-cell failure caused by misfolded proinsulin is still incompletely understood. Herein, using Akita mice expressing diabetes-causing mutant proinsulin, we found that misfolded proinsulin abnormally interacted with the precursor of insulin receptor (ProIR) in the endoplasmic reticulum (ER), impaired ProIR maturation to insulin receptor (IR), and decreased insulin signaling in β cells. Importantly, using db/db insulin-resistant mice, we found that oversynthesis of proinsulin led to an increased proinsulin misfolding, which resulted in impairments of ProIR processing and insulin signaling in β cells. These results reveal for the first time that misfolded proinsulin can interact with ProIR in the ER, impairing intracellular processing of ProIR and leading to defective insulin signaling that may contribute to β-cell failure in both MIDY and T2D.—Liu, S., Li, X., Yang, J., Zhu, R., Fan, Z., Xu, X., Feng, W., Cui, J., Sun, J., Liu, M. Misfolded proinsulin impairs processing of precursor of insulin receptor and insulin signaling in β cells.
Collapse
Affiliation(s)
- Shiqun Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Yang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruimin Zhu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenqian Fan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoxi Xu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenli Feng
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingqiu Cui
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinhong Sun
- Department of Health Management, Tianjin Medical University General Hospital, Tianjin, China
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
62
|
Garcia-Galiano D, Borges BC, Allen SJ, Elias CF. PI3K signalling in leptin receptor cells: Role in growth and reproduction. J Neuroendocrinol 2019; 31:e12685. [PMID: 30618188 PMCID: PMC6533139 DOI: 10.1111/jne.12685] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/04/2019] [Accepted: 01/04/2019] [Indexed: 12/15/2022]
Abstract
Nutrition and growth are important signals for pubertal development, although how they are perceived and integrated in brain circuits has not been well defined. Growth hormones and metabolic cues both recruit phosphatidylinositol 3-kinase (PI3K) signalling in hypothalamic sites, although whether they converge into the same neuronal population(s) is also not known. In this review, we discuss recent findings from our laboratory showing the role of PI3K subunits in cells directly responsive to the adipocyte-derived hormone leptin in the coordination of growth, pubertal development and fertility. Mice with deletion of PI3K p110α and p110β catalytic subunits in leptin receptor cells (LRΔα+β ) have a lean phenotype associated with increased energy expenditure, locomotor activity and thermogenesis. The LRΔα+β mice also show deficient growth and delayed puberty. Deletion of a single subunit (ie, p110α) in LR cells (LRΔα ) causes a similar phenotype of increased energy expenditure, deficient growth and delayed pubertal development, indicating that these functions are preferably controlled by p110α. The LRΔα mice show enhanced leptin sensitivity in metabolic regulation but, remarkably, these mice are unresponsive to the effects of leptin on growth and puberty. PI3K is also recruited by insulin and a subpopulation of LR neurones is responsive to i.c.v. insulin administration. Deletion of insulin receptor in LR cells causes no changes in body weight or linear growth and induces only a mild delay in pubertal completion. Our findings demonstrate that PI3K in LR cells plays an essential role in growth and reproduction. We will also discuss the potential neural pathways underlying these effects.
Collapse
Affiliation(s)
- David Garcia-Galiano
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Beatriz C. Borges
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Kresge Hearing Research Institute and Department of Otolaryngology - Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Susan J. Allen
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Carol F. Elias
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
63
|
Suarez AN, Noble EE, Kanoski SE. Regulation of Memory Function by Feeding-Relevant Biological Systems: Following the Breadcrumbs to the Hippocampus. Front Mol Neurosci 2019; 12:101. [PMID: 31057368 PMCID: PMC6482164 DOI: 10.3389/fnmol.2019.00101] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022] Open
Abstract
The hippocampus (HPC) controls fundamental learning and memory processes, including memory for visuospatial navigation (spatial memory) and flexible memory for facts and autobiographical events (declarative memory). Emerging evidence reveals that hippocampal-dependent memory function is regulated by various peripheral biological systems that are traditionally known for their roles in appetite and body weight regulation. Here, we argue that these effects are consistent with a framework that it is evolutionarily advantageous to encode and recall critical features surrounding feeding behavior, including the spatial location of a food source, social factors, post-absorptive processing, and other episodic elements of a meal. We review evidence that gut-to-brain communication from the vagus nerve and from feeding-relevant endocrine systems, including ghrelin, insulin, leptin, and glucagon-like peptide-1 (GLP-1), promote hippocampal-dependent spatial and declarative memory via neurotrophic and neurogenic mechanisms. The collective literature reviewed herein supports a model in which various stages of feeding behavior and hippocampal-dependent memory function are closely linked.
Collapse
Affiliation(s)
| | | | - Scott E. Kanoski
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
64
|
Baldini G, Phelan KD. The melanocortin pathway and control of appetite-progress and therapeutic implications. J Endocrinol 2019; 241:R1-R33. [PMID: 30812013 PMCID: PMC6500576 DOI: 10.1530/joe-18-0596] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 01/22/2019] [Indexed: 12/19/2022]
Abstract
The initial discovery that ob/ob mice become obese because of a recessive mutation of the leptin gene has been crucial to discover the melanocortin pathway to control appetite. In the melanocortin pathway, the fed state is signaled by abundance of circulating hormones such as leptin and insulin, which bind to receptors expressed at the surface of pro-opiomelanocortin (POMC) neurons to promote processing of POMC to the mature hormone α-melanocyte-stimulating hormone (α-MSH). The α-MSH released by POMC neurons then signals to decrease energy intake by binding to melanocortin-4 receptor (MC4R) expressed by MC4R neurons to the paraventricular nucleus (PVN). Conversely, in the 'starved state' activity of agouti-related neuropeptide (AgRP) and of neuropeptide Y (NPY)-expressing neurons is increased by decreased levels of circulating leptin and insulin and by the orexigenic hormone ghrelin to promote food intake. This initial understanding of the melanocortin pathway has recently been implemented by the description of the complex neuronal circuit that controls the activity of POMC, AgRP/NPY and MC4R neurons and downstream signaling by these neurons. This review summarizes the progress done on the melanocortin pathway and describes how obesity alters this pathway to disrupt energy homeostasis. We also describe progress on how leptin and insulin receptors signal in POMC neurons, how MC4R signals and how altered expression and traffic of MC4R change the acute signaling and desensitization properties of the receptor. We also describe how the discovery of the melanocortin pathway has led to the use of melanocortin agonists to treat obesity derived from genetic disorders.
Collapse
Affiliation(s)
- Giulia Baldini
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kevin D. Phelan
- Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| |
Collapse
|
65
|
Borges BC, Han X, Allen SJ, Garcia-Galiano D, Elias CF. Insulin signaling in LepR cells modulates fat and glucose homeostasis independent of leptin. Am J Physiol Endocrinol Metab 2019; 316:E121-E134. [PMID: 30376348 PMCID: PMC6417687 DOI: 10.1152/ajpendo.00287.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hypothalamic neurons detect changes in circulating hormones such as leptin and insulin and put forward outputs to sustain energy and glucose homeostasis. Because leptin and insulin receptors colocalize in ~40-60% of neurons in the hypothalamus, we characterized the metabolic phenotype of mice with selective deletion of the insulin receptor (InsR) in LepR cells. LRΔInsR mice presented no difference in body weight and insulin levels but increased fat mass. In the light phase, LRΔInsR mice exhibited increased food intake, locomotor activity, carbon dioxide production, and respiratory exchange rate. These mice showed reduced fat oxidation and reduced expression of cluster of differentiation 36 and AMP-activated protein kinase-α1 in the liver, increased glucose oxidation in the light phase, and overall reduced basal glucose levels. To verify the impact of InsR deletion in LepR cells in obesity, we generated ob/ ob InsRfl, ob/ ob LRcre, and ob/ ob LRΔInsR mice. The ob/ ob LRΔInsR mice had higher body weight, fat mass, and expression of genes related to fat metabolism in the liver. No difference in food intake despite increased neuropeptide Y and agouti-related peptide expression, and no difference in energy expenditure, fat, or glucose oxidation was found in ob/ ob LRΔInsR compared with LRcre or LRΔInsR controls. Remarkably, basal glucose levels were reduced, and the expression of genes associated with glucose metabolism in the liver was higher. Insulin signaling in LepR cells is required for the proper fat and glucose oxidation. These effects are independent of leptin given that the leptin-deficient ob/ ob LRΔInsR mice also presented reduced glycemia and higher adiposity. The mechanisms underlying these responses remain to be unveiled.
Collapse
Affiliation(s)
- Beatriz C Borges
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
- Department of Physiology, School of Medicine of Ribeirao Preto, University of Sao Paulo , Brazil
| | - Xingfa Han
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
- Isotope Research Laboratory, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Susan J Allen
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| | - David Garcia-Galiano
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
| | - Carol F Elias
- Department of Molecular and Integrative Physiology, University of Michigan , Ann Arbor, Michigan
- Department of Obstetrics and Gynecology, University of Michigan , Ann Arbor, Michigan
| |
Collapse
|
66
|
Nam SM, Kwon HJ, Kim W, Kim JW, Hahn KR, Jung HY, Kim DW, Yoo DY, Seong JK, Hwang IK, Yoon YS. Changes of myelin basic protein in the hippocampus of an animal model of type 2 diabetes. Lab Anim Res 2018; 34:176-184. [PMID: 30671103 PMCID: PMC6333608 DOI: 10.5625/lar.2018.34.4.176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 12/31/2022] Open
Abstract
In this study, we observed chronological changes in the immunoreactivity and expression level of myelin basic protein (MBP), one of the most abundant proteins in the central nervous system, in the hippocampus of Zucker diabetic fatty (ZDF) rats and their control littermates (Zucker lean control; ZLC). In the ZLC group, body weight steadily increased with age; the body weight of the ZDF group, however, peaked at 30 weeks of age, and subsequently decreased. Based on the changes of body weight, animals were divided into the following six groups: early (12-week), middle (30-week), and chronic (52-week) diabetic groups and their controls. MBP immunoreactivity was found in the alveus, strata pyramidale, and lacunosum-moleculare of the CA1 region, strata pyramidale and radiatum of the CA3 region, and subgranular zone, polymorphic layer, and molecular layer of the dentate gyrus. MBP immunoreactivity was lowest in the hippocampus of 12-week-old rats in the ZLC group, and highest in 12-week-old rats in the ZDF group. Diabetes increased MBP levels in the 12-week-old group, while MBP immunoreactivity decreased in the 30-week-old group. In the 52-week-old ZLC and ZDF groups, MBP immunoreactivity was detected in the hippocampus, similar to the 30-week-old ZDF group. Western blot results corroborated with immunohistochemical results. These results suggested that changes in the immunoreactivity and expression of MBP in the hippocampus might be a compensatory response to aging, while the sustained levels of MBP in diabetic animals could be attributed to a loss of compensatory responses in oligodendrocytes.
Collapse
Affiliation(s)
- Sung Min Nam
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Anatomy, College of Veterinary Medicine, Konkuk University, Seoul, Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, Korea
| | - Woosuk Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, Korea
| | - Dae Young Yoo
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- Department of Anatomy, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Je Kyung Seong
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, Korea
- KMPC (Korea Mouse Phenotyping Center), Seoul National University, Seoul, Korea
| |
Collapse
|
67
|
Niazi RK, Gjesing AP, Hollensted M, Have CT, Grarup N, Pedersen O, Ullah A, Shahid G, Ahmad W, Gul A, Hansen T. Identification of novel LEPR mutations in Pakistani families with morbid childhood obesity. BMC MEDICAL GENETICS 2018; 19:199. [PMID: 30442103 PMCID: PMC6238292 DOI: 10.1186/s12881-018-0710-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 10/25/2018] [Indexed: 12/30/2022]
Abstract
Background Mutations in the genes encoding leptin (LEP), the leptin receptor (LEPR), and the melanocortin 4 receptor (MC4R) are known to cause severe early-onset childhood obesity. The aim of the current study was to examine the prevalence of damaging LEP, LEPR, and MC4R mutations in Pakistani families having a recessive heritance of early-onset obesity. Methods Using targeted resequencing, the presence of rare mutations in LEP, LEPR, and MC4R, was investigated in individuals from 25 families suspected of having autosomal recessive early-onset obesity. Segregation patterns of variants were assessed based on chip-based genotyping. Results Homozygous LEPR variants were identified in two probands. One carried a deletion (c.3260AG) resulting in the frameshift mutation p.Ser1090Trpfs*6, and the second carried a substitution (c.2675C > G) resulting in the missense mutation p.Pro892Arg. Both mutations were located within regions of homozygosity shared only among affected individuals. Both probands displayed early-onset obesity, hyperphagia and diabetes. No mutations were found in LEP and MC4R. Conclusions The current study highlights the implication of LEPR mutations in cases of severe early-onset obesity in consanguineous Pakistani families. Through targeted resequencing, we identified novel damaging mutations, and our approach may therefore be utilized in clinical testing or diagnosis of known forms of monogenic obesity with the aim of optimizing obesity treatment. Electronic supplementary material The online version of this article (10.1186/s12881-018-0710-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Robina Khan Niazi
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan.,Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Children Hospital, Pakistan Institute of Medical Sciences, Islamabad, Pakistan.,Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Anette P Gjesing
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Hollensted
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Christian Theil Have
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Asmat Ullah
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Gulbin Shahid
- Children Hospital, Pakistan Institute of Medical Sciences, Islamabad, Pakistan
| | - Wasim Ahmad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Asma Gul
- Department of Bioinformatics and Biotechnology, International Islamic University, Islamabad, Pakistan
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
68
|
Reddon H, Patel Y, Turcotte M, Pigeyre M, Meyre D. Revisiting the evolutionary origins of obesity: lazy versus peppy-thrifty genotype hypothesis. Obes Rev 2018; 19:1525-1543. [PMID: 30261552 DOI: 10.1111/obr.12742] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/26/2018] [Accepted: 07/01/2018] [Indexed: 12/31/2022]
Abstract
The recent global obesity epidemic is attributed to major societal and environmental changes, such as excessive energy intake and sedentary lifestyle. However, exposure to 'obesogenic' environments does not necessarily result in obesity at the individual level, as 40-75% of body mass index variation in population is attributed to genetic differences. The thrifty genotype theory posits that genetic variants promoting efficient food sequestering and optimal deposition of fat during periods of food abundance were evolutionarily advantageous for the early hunter-gatherer and were positively selected. However, the thrifty genotype is likely too simplistic and fails to provide a justification for the complex distribution of obesity predisposing gene variants and for the broad range of body mass index observed in diverse ethnic groups. This review proposes that gene pleiotropy may better account for the variability in the distribution of obesity susceptibility alleles across modern populations. We outline the lazy-thrifty versus peppy-thrifty genotype hypothesis and detail the body of evidence in the literature in support of this novel concept. Future population genetics and mathematical modelling studies that account for pleiotropy may further improve our understanding of the evolutionary origins of the current obesity epidemic.
Collapse
Affiliation(s)
- H Reddon
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Canada
| | - Y Patel
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Canada
| | - M Turcotte
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Canada
| | - M Pigeyre
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - D Meyre
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| |
Collapse
|
69
|
Fuchs T, Loureiro MDP, Macedo LE, Nocca D, Nedelcu M, Costa-Casagrande TA. Modelos animais na síndrome metabólica. Rev Col Bras Cir 2018; 45:e1975. [PMID: 30379216 DOI: 10.1590/0100-6991e-20181975] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/20/2018] [Indexed: 12/19/2022] Open
Abstract
RESUMO O conhecimento sobre modelos animais para estudo metabólico representa a base da pesquisa nessa área. Este trabalho tem por objetivo revisar os principais modelos animais a serem utilizados no estudo da obesidade e da síndrome metabólica. Para isso, pesquisa no banco de dados Pubmed foi realizada usando as palavras-chave “animal models”, “obesity”, "metabolic syndrome”, e “bariatric surgery”. Várias espécies de animais podem ser usadas para o estudo de distúrbios metabólicos, no entanto, os roedores, tanto modelos monogênicos quanto modelos de obesidade induzida por dieta (DIO), são os animais mais utilizados nessa área. Animais monogênicos são a melhor escolha se apenas um aspecto estiver sendo avaliado. Animais DIO tendem a demonstrar melhor a interação entre doença, ambiente e gene. No entanto, eles ainda não são totalmente eficazes para a compreensão de todos os mecanismos dessa doença.
Collapse
|
70
|
Duffy S, Lutz TA, Boyle CN. Rodent models of leptin receptor deficiency are less sensitive to amylin. Am J Physiol Regul Integr Comp Physiol 2018; 315:R856-R865. [DOI: 10.1152/ajpregu.00179.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The pancreatic hormone amylin is released from beta cells following nutrient ingestion and contributes to the control of body weight and glucose homeostasis. Amylin reduces food intake by activating neurons in the area postrema (AP). Amylin was also shown to synergize with the adipokine leptin, with combination therapy producing greater weight loss and food intake reduction than either hormone alone. Although amylin and leptin were initially thought to interact downstream of the AP in the hypothalamus, recent findings show that the two hormones can act on the same AP neurons, suggesting a more direct relationship. The objective of this study was to determine whether amylin action depends on functional leptin signaling. We tested the ability of amylin to induce satiation and to activate its primary target neurons in the AP in two rodent models of LepR deficiency, the db/db mouse and the Zucker diabetic fatty (ZDF) rat. When compared with wild-type (WT) mice, db/db mice exhibited reduced amylin-induced satiation, reduced amylin-induced Fos in the AP, and a lower expression of calcitonin receptor (CTR) protein, the core component of all amylin receptors. ZDF rats also showed no reduction in food intake following amylin treatment; however, unlike the db/db mice, levels of amylin-induced Fos and CTR in the AP were no different than WT rats. Our results suggest that LepR expression is required for the full anorexic effect of amylin; however, the neuronal activation in the AP seems to depend on the type of LepR mutation.
Collapse
Affiliation(s)
- Sonya Duffy
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
| | - Thomas A. Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
- Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Christina N. Boyle
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland
| |
Collapse
|
71
|
Sex-specific differences in hepatic steatosis in obese spontaneously hypertensive (SHROB) rats. Biol Sex Differ 2018; 9:40. [PMID: 30201044 PMCID: PMC6131947 DOI: 10.1186/s13293-018-0202-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 08/31/2018] [Indexed: 12/12/2022] Open
Abstract
Background Patients with metabolic syndrome, who are characterized by co-existence of insulin resistance, hypertension, hyperlipidemia, and obesity, are also prone to develop non-alcoholic fatty liver disease (NAFLD). Although the prevalence and severity of NAFLD is significantly greater in men than women, the mechanisms by which gender modulates the pathogenesis of hepatic steatosis are poorly defined. The obese spontaneously hypertensive (SHROB) rats represent an attractive model of metabolic syndrome without overt type 2 diabetes. Although pathological manifestation caused by the absence of a functional leptin receptor has been extensively studied in SHROB rats, it is unknown whether these animals elicited sex-specific differences in the development of hepatic steatosis. Methods We compared hepatic pathology in male and female SHROB rats. Additionally, we examined key biochemical and molecular parameters of signaling pathways linked with hyperinsulinemia and hyperlipidemia. Finally, using methods of quantitative polymerase chain reaction (qPCR) and western blot analysis, we quantified expression of 45 genes related to lipid biosynthesis and metabolism in the livers of male and female SHROB rats. Results We show that all SHROB rats developed hepatic steatosis that was accompanied by enhanced expression of SREBP1, SREBP2, ACC1, and FASN proteins. The livers of male rats also elicited higher induction of Pparg, Ppara, Slc2a4, Atox1, Skp1, Angptl3, and Pnpla3 mRNAs. In contrast, the livers of female SHROB rats elicited constitutively higher levels of phosphorylated JNK and AMPK and enhanced expression of Cd36. Conclusion Based on these data, we conclude that the severity of hepatic steatosis in male and female SHROB rats was mainly driven by increased de novo lipogenesis. Moreover, male and female SHROB rats also elicited differential severity of hepatic steatosis that was coupled with sex-specific differences in fatty acid transport and esterification. Electronic supplementary material The online version of this article (10.1186/s13293-018-0202-x) contains supplementary material, which is available to authorized users.
Collapse
|
72
|
Mazor R, Friedmann-Morvinski D, Alsaigh T, Kleifeld O, Kistler EB, Rousso-Noori L, Huang C, Li JB, Verma IM, Schmid-Schönbein GW. Cleavage of the leptin receptor by matrix metalloproteinase-2 promotes leptin resistance and obesity in mice. Sci Transl Med 2018; 10:eaah6324. [PMID: 30135249 PMCID: PMC9678493 DOI: 10.1126/scitranslmed.aah6324] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 05/01/2017] [Accepted: 03/22/2018] [Indexed: 08/08/2023]
Abstract
Obesity and related morbidities pose a major health threat. Obesity is associated with increased blood concentrations of the anorexigenic hormone leptin; however, obese individuals are resistant to its anorexigenic effects. We examined the phenomenon of reduced leptin signaling in a high-fat diet-induced obesity model in mice. Obesity promoted matrix metalloproteinase-2 (Mmp-2) activation in the hypothalamus, which cleaved the leptin receptor's extracellular domain and impaired leptin-mediated signaling. Deletion of Mmp-2 restored leptin receptor expression and reduced circulating leptin concentrations in obese mice. Lentiviral delivery of short hairpin RNA to silence Mmp-2 in the hypothalamus of wild-type mice prevented leptin receptor cleavage and reduced fat accumulation. In contrast, lentiviral delivery of Mmp-2 in the hypothalamus of Mmp-2-/- mice promoted leptin receptor cleavage and higher body weight. In a genetic mouse model of obesity, transduction of cleavage-resistant leptin receptor in the hypothalamus reduced the rate of weight gain compared to uninfected mice or mice infected with the wild-type receptor. Immunofluorescence analysis showed that astrocytes and agouti-related peptide neurons were responsible for Mmp-2 secretion in mice fed a high-fat diet. These results suggest a mechanism for leptin resistance through activation of Mmp-2 and subsequent cleavage of the extracellular domain of the leptin receptor.
Collapse
Affiliation(s)
- Rafi Mazor
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Dinorah Friedmann-Morvinski
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tom Alsaigh
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Oded Kleifeld
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Erik B Kistler
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92093, USA
- Department of Anesthesiology and Critical Care, University of California, San Diego, La Jolla, CA 92093, USA
| | - Liat Rousso-Noori
- Department of Biochemistry and Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neurosciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Cheng Huang
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria 3800, Australia
| | - Joyce B Li
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Geert W Schmid-Schönbein
- Department of Bioengineering, Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
73
|
Abstract
PURPOSE OF REVIEW This review aims to present current information on genes underlying severe obesity, with the main emphasis on the three genes LEP, LEPR and MC4R. RECENT FINDINGS There is a substantial amount of evidence that variants in at least ten different genes are the cause of severe monogenic obesity. The majority of these are involved in the leptin-melanocortin signalling pathway. Due to the frequency of some of the identified variants, it is clear that monogenic variants also make a significant contribution to common obesity. The artificial distinction between rare monogenic obesity and common polygenic obesity is now obsolete with the identification of MC4R variants of strong effect in the general population.
Collapse
Affiliation(s)
- Una Fairbrother
- School of Human Sciences, London Metropolitan University, North Campus, 166-220 Holloway Road, London, N7 8DB, UK
| | - Elliot Kidd
- School of Human Sciences, London Metropolitan University, North Campus, 166-220 Holloway Road, London, N7 8DB, UK
| | - Tanya Malagamuwa
- Institute of Medical and Biomedical Education, St George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE, UK
| | - Andrew Walley
- Institute of Medical and Biomedical Education, St George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE, UK.
| |
Collapse
|
74
|
Rai A, Mishra R, Ganesh S. Suppression of leptin signaling reduces polyglucosan inclusions and seizure susceptibility in a mouse model for Lafora disease. Hum Mol Genet 2018; 26:4778-4785. [PMID: 28973665 DOI: 10.1093/hmg/ddx357] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 09/12/2017] [Indexed: 01/10/2023] Open
Abstract
Lafora disease (LD) represents a fatal form of neurodegenerative disorder characterized by the presence of abnormally large number of polyglucosan bodies-called the Lafora bodies-in neurons and other tissues of the affected patients. The disease is caused by defects in the EPM2A gene coding for a protein phosphatase (laforin) or the NHLRC1 gene coding for an ubiquitin ligase (malin). Studies have shown that inhibition of glycogen synthesis in the brain could prevent the formation of Lafora bodies in the neurons and reduce seizure susceptibility in laforin-deficient mouse, an established animal model for LD. Since increased glucose uptake is thought to underlie increased glycogen in LD, and since the adipocyte hormone leptin is known to positively regulate the glucose uptake in neurons, we reasoned that blocking leptin signaling might reduce the neuronal glucose uptake and ameliorate the LD pathology. We demonstrate here that mice that were deficient for both laforin and leptin receptor showed a reduction in the glycogen level, Lafora bodies and gliosis in the brain, and displayed reduced susceptibility to induced seizures as compared to animals that were deficient only for laforin. Thus, blocking leptin signaling could be a one of the effective therapeutic strategies in LD.
Collapse
Affiliation(s)
- Anupama Rai
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Rohit Mishra
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Subramaniam Ganesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| |
Collapse
|
75
|
Fabelo C, Hernandez J, Chang R, Seng S, Alicea N, Tian S, Conde K, Wagner EJ. Endocannabinoid Signaling at Hypothalamic Steroidogenic Factor-1/Proopiomelanocortin Synapses Is Sex- and Diet-Sensitive. Front Mol Neurosci 2018; 11:214. [PMID: 29973869 PMCID: PMC6020785 DOI: 10.3389/fnmol.2018.00214] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 05/30/2018] [Indexed: 01/05/2023] Open
Abstract
We tested the hypotheses that steroidogenic factor (SF)-1 neurons in the hypothalamic ventromedial nucleus (VMN) provide sexually disparate, endocannabinoid (EC)- and diet-sensitive glutamatergic input onto proopiomelanocortin (POMC) neurons. Electrophysiological recordings were performed in hypothalamic slices from intact and castrated guinea pigs, along with in vitro optogenetic experiments in intact male as well as cycling and ovariectomized female NR5A1-Cre mice. In slices from castrated male and female guinea pigs, depolarized-induced suppression of excitation (DSE) time-dependently reduced the amplitude of evoked excitatory postsynaptic currents (eEPSCs) in POMC neurons generated by electrically stimulating the dorsomedial VMN. Androgen stimulation rapidly enhanced this DSE, which was also found in insulin-resistant, high-fat diet (HFD)-fed males. By contrast, retrograde signaling at VMN/ARC POMC synapses was markedly attenuated in periovulatory females. HFD potentiated central cannabinoid-induced hyperphagia in both males and females, but exerted differential influences on cannabinoid-induced increases in energy expenditure. In NR5A1-Cre mice, the reduction in light-evoked EPSC amplitude caused by postsynaptic depolarization in cycling females was modest in comparison to that seen in intact males. Estradiol attenuated the DSE in light-evoked EPSC amplitude in slices from ovariectomized females. Moreover, the retrograde inhibition of transmission was further accentuated in HFD-fed males. Chemogenetic activation of SF-1 neurons suppressed appetite and increased energy expenditure in males, effects which were attenuated by HFD. Conversely, energy expenditure was increased in estradiol- but not vehicle-treated ovariectomized females. Together with our previous studies indicating that DSE in POMC neurons is EC-mediated, these findings indicate that VMN SF-1/ARC POMC synapses represent a sexually differentiated, EC- and diet-sensitive anorexigenic component within the hypothalamic energy balance circuitry.
Collapse
Affiliation(s)
- Carolina Fabelo
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Jennifer Hernandez
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Rachel Chang
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Sakara Seng
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Natalia Alicea
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Sharon Tian
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Kristie Conde
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Edward J Wagner
- Department of Basic Medical Sciences, College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA, United States.,Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
76
|
Carreras NL, Martorell P, Chenoll E, Genovés S, Ramón D, Aleixandre A. Anti-obesity properties of the strain Bifidobacterium animalis subsp. lactis CECT 8145 in Zücker fatty rats. Benef Microbes 2018; 9:629-641. [DOI: 10.3920/bm2017.0141] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
We evaluated the effect of oral administration of Bifidobacterium animalis subsp. lactis CECT 8145 strain in Zücker fatty rats. The Zücker fatty rats were randomly divided into two groups (n=10 each) and administered either B. animalis subsp. lactis CECT 8145 (1010 cfu/day) suspended in skim milk, or skim milk alone (control group). Each treatment was administered in drinking bottles from week 5 until week 17 of age. A lean Zücker rat group (standard group) was included to provide normal values for the Zücker strain. This group was administered skim milk in the drinking bottle for the same experimental period as Zücker fatty rats. Body weight gain was greater in the fatty control group than in the fatty rats treated daily with B. animalis subsp. lactis CECT 8145. Furthermore, dry and liquid food intake significantly decreased in the treated Zücker fatty group and these rats also showed decreased plasma ghrelin levels as compared with the Zücker fatty control group. B. animalis subsp. lactis CECT 8145 intake also decreased plasma tumour necrosis factor-α (a proinflammatory cytokine) and plasma malondialdehyde (a biomarker of oxidative stress). Moreover, the ratio plasma total cholesterol/plasma cholesterol transported by high-density lipoproteins, considered as an index for cardiovascular disease, also significantly decreased in the Zücker fatty rats treated with B. animalis subsp. lactis CECT 8145. By contrast, this bacterial strain significantly increased plasma adiponectin (an insulin-sensitising adipokine), but did not produce significant effects on triglyceride levels or glucose metabolism biomarkers. Although further research is required to confirm B. animalis subsp. lactis CECT 8145 is an efficient anti-obesity treatment in humans, the results obtained in this study are promising and point to the health and anti-obesity properties of this bacterial strain.
Collapse
Affiliation(s)
- N. López Carreras
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| | - P. Martorell
- Department of Food Biotechnology; Biópolis S.L. Parc Científic Universitat De València, Edif. 2, C/Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - E. Chenoll
- Department of Food Biotechnology; Biópolis S.L. Parc Científic Universitat De València, Edif. 2, C/Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - S. Genovés
- Department of Food Biotechnology; Biópolis S.L. Parc Científic Universitat De València, Edif. 2, C/Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - D. Ramón
- Department of Food Biotechnology; Biópolis S.L. Parc Científic Universitat De València, Edif. 2, C/Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - A. Aleixandre
- Departamento de Farmacología, Facultad de Medicina, Universidad Complutense de Madrid, Avenida Complutense s/n, 28040 Madrid, Spain
| |
Collapse
|
77
|
Tyree SM, Borniger JC, de Lecea L. Hypocretin as a Hub for Arousal and Motivation. Front Neurol 2018; 9:413. [PMID: 29928253 PMCID: PMC5997825 DOI: 10.3389/fneur.2018.00413] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023] Open
Abstract
The lateral hypothalamus is comprised of a heterogeneous mix of neurons that serve to integrate and regulate sleep, feeding, stress, energy balance, reward, and motivated behavior. Within these populations, the hypocretin/orexin neurons are among the most well studied. Here, we provide an overview on how these neurons act as a central hub integrating sensory and physiological information to tune arousal and motivated behavior accordingly. We give special attention to their role in sleep-wake states and conditions of hyper-arousal, as is the case with stress-induced anxiety. We further discuss their roles in feeding, drug-seeking, and sexual behavior, which are all dependent on the motivational state of the animal. We further emphasize the application of powerful techniques, such as optogenetics, chemogenetics, and fiber photometry, to delineate the role these neurons play in lateral hypothalamic functions.
Collapse
Affiliation(s)
- Susan M Tyree
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Jeremy C Borniger
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| | - Luis de Lecea
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, United States
| |
Collapse
|
78
|
Short-Term Soy Protein Isolate Feeding Prevents Liver Steatosis and Reduces Serum ALT and AST Levels in Obese Female Zucker Rats. Biomedicines 2018; 6:biomedicines6020055. [PMID: 29757972 PMCID: PMC6027420 DOI: 10.3390/biomedicines6020055] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 05/04/2018] [Accepted: 05/09/2018] [Indexed: 01/22/2023] Open
Abstract
Non-alcoholic fatty liver disease is a common liver disorder worldwide and is associated with obesity. We investigated effects of obesity and short-term intake of soy protein with isoflavones (SPI) on body weight change, energy intake, liver steatosis, and serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), and leptin levels. Seventeen lean and seventeen obese (fa/fa) female Zucker rats were randomly assigned to either casein or SPI diet for 8 weeks. Body weight was recorded twice weekly; feed intake was measured weekly. Livers were examined histologically, and serum AST, ALT, and leptin levels were measured. Obese soy-fed (OS) rats gained more weight but had lower liver steatosis than obese casein-fed (OC) rats. Energy intake for OS versus OC rats were only different at weeks 2 and 3. Serum AST and ALT levels were lower in OS versus OC rats. Obesity increased serum leptin levels for both diets. In summary, short-term SPI intake reduced liver steatosis, and the only time points at which the mean energy intakes of OS and OC rats differed were at weeks 2 and 3, where OS rats had a higher mean energy intake, which may have accounted for the increased body weight in OS rats.
Collapse
|
79
|
Coleman JRI, Krapohl E, Eley TC, Breen G. Individual and shared effects of social environment and polygenic risk scores on adolescent body mass index. Sci Rep 2018; 8:6344. [PMID: 29679049 PMCID: PMC5910407 DOI: 10.1038/s41598-018-24774-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 03/15/2018] [Indexed: 11/20/2022] Open
Abstract
Juvenile obesity is associated with adverse health outcomes. Understanding genetic and environmental influences on body mass index (BMI) during adolescence could inform interventions. We investigated independent and interactive effects of parenting, socioeconomic status (SES) and polygenic risk on BMI pre-adolescence, and on the rate of change in BMI across adolescence. Genome-wide genotype data, BMI and child perceptions of parental warmth and punitive discipline were available at 11 years old, and parental SES was available from birth on 3,414 unrelated participants. Linear models were used to test the effects of social environment and polygenic risk on pre-adolescent BMI. Change in BMI across adolescence was assessed in a subset (N = 1943). Sex-specific effects were assessed. Higher genetic risk was associated with increased BMI pre-adolescence and across adolescence (p < 0.00417, corrected for multiple tests). Negative parenting was not significantly associated with either phenotype, but lower SES was associated with increased BMI pre-adolescence. No interactions passed correction for multiple testing. Polygenic risk scores from adult GWAS meta-analyses are associated with BMI in juveniles, suggesting a stable genetic component. Pre-adolescent BMI was associated with social environment, but parental style has, at most, a small effect.
Collapse
Affiliation(s)
- Jonathan R I Coleman
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK.,National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK
| | - Eva Krapohl
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK
| | - Thalia C Eley
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK. .,National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK.
| | - Gerome Breen
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, UK. .,National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, UK.
| |
Collapse
|
80
|
Pan W, Adams JM, Allison MB, Patterson C, Flak JN, Jones J, Strohbehn G, Trevaskis J, Rhodes CJ, Olson DP, Myers MG. Essential Role for Hypothalamic Calcitonin Receptor‒Expressing Neurons in the Control of Food Intake by Leptin. Endocrinology 2018; 159:1860-1872. [PMID: 29522093 PMCID: PMC5888224 DOI: 10.1210/en.2017-03259] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/28/2018] [Indexed: 01/07/2023]
Abstract
The adipocyte-derived hormone leptin acts via its receptor (LepRb) on central nervous system neurons to communicate the repletion of long-term energy stores, to decrease food intake, and to promote energy expenditure. We generated mice that express Cre recombinase from the calcitonin receptor (Calcr) locus (Calcrcre mice) to study Calcr-expressing LepRb (LepRbCalcr) neurons, which reside predominantly in the arcuate nucleus (ARC). Calcrcre-mediated ablation of LepRb in LepRbCalcrknockout (KO) mice caused hyperphagic obesity. Because LepRb-mediated transcriptional control plays a crucial role in leptin action, we used translating ribosome affinity purification followed by RNA sequencing to define the transcriptome of hypothalamic Calcr neurons, along with its alteration in LepRbCalcrKO mice. We found that ARC LepRbCalcr cells include neuropeptide Y (NPY)/agouti-related peptide (AgRP)/γ-aminobutyric acid (GABA) ("NAG") cells as well as non-NAG cells that are distinct from pro-opiomelanocortin cells. Furthermore, although LepRbCalcrKO mice exhibited dysregulated expression of several genes involved in energy balance, neither the expression of Agrp and Npy nor the activity of NAG cells was altered in vivo. Thus, although direct leptin action via LepRbCalcr cells plays an important role in leptin action, our data also suggest that leptin indirectly, as well as directly, regulates these cells.
Collapse
Affiliation(s)
- Warren Pan
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
| | - Jessica M Adams
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Margaret B Allison
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| | - Christa Patterson
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jonathan N Flak
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Justin Jones
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Garth Strohbehn
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | | | | | - David P Olson
- Division of Endocrinology, Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Martin G Myers
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
81
|
Dobrian AD, Huyck RW, Glenn L, Gottipati V, Haynes BA, Hansson GI, Marley A, McPheat WL, Nadler JL. Activation of the 12/15 lipoxygenase pathway accompanies metabolic decline in db/db pre-diabetic mice. Prostaglandins Other Lipid Mediat 2018; 136:23-32. [PMID: 29605541 DOI: 10.1016/j.prostaglandins.2018.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/06/2018] [Accepted: 03/15/2018] [Indexed: 12/17/2022]
Abstract
The 12-lipoxygenase (12LO) pathway is a promising target to reduce islet dysfunction, adipose tissue (AT) inflammation and insulin resistance. Optimal pre-clinical models for the investigation of selective12LO inhibitors in this context have not yet been identified. The objective of this study was to characterize the time course of 12LO isoform expression and metabolite production in pancreatic islets and AT of C57BLKS/J-db/db obese diabetic mouse in a pre-diabetic state in order to establish a suitable therapeutic window for intervention with selective lipoxygenase inhibitors. Mice have 2 major 12LO isoforms -the leukocyte type (12/15LO) and the platelet type (p12LO) and both are expressed in islets and AT. We found a sharp increase in protein expression of 12/15LO in the pancreatic islets of 10-week old db-/- mice compared to 8- week old counterparts. Immunohistochemistry showed that the increase in islet 12/15LO parallels a decline in islet number. Analysis of 12- and 15-hydroperoxytetraeicosanoid acids (HETE)s showed a 2-3 fold increase especially in 12(S)-HETE that mirrored the increase in 12/15LO expression in islets. Analysis of AT and stromal vascular fraction (SVF) showed a significant increase of platelet 12LO gene expression along with 12- and 15- HETEs. The data demonstrate that the db/db mouse is a suitable model for investigation of 12/15LO inhibitors in the development of inflammatory mediated type 2 diabetes, with a narrow window of therapeutic intervention prior to 8 weeks of age.
Collapse
Affiliation(s)
- Anca D Dobrian
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, United States.
| | - Ryan W Huyck
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Lindsey Glenn
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Vijay Gottipati
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Bronson A Haynes
- Department of Physiological Sciences, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Göran I Hansson
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Anna Marley
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca,Alderley Park, Macclesfield, Cheshire, SK10 4TG, UK
| | - William L McPheat
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Pepparedsleden 1, Mölndal, 431 83, Sweden
| | - Jerry L Nadler
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, United States
| |
Collapse
|
82
|
Arends D, Hesse D, Brockmann GA. Invited review: Genetic and genomic mouse models for livestock research. Arch Anim Breed 2018. [DOI: 10.5194/aab-61-87-2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Abstract. Knowledge about the function and functioning of single or
multiple interacting genes is of the utmost significance for understanding the
organism as a whole and for accurate livestock improvement through genomic
selection. This includes, but is not limited to, understanding the
ontogenetic and environmentally driven regulation of gene action
contributing to simple and complex traits. Genetically modified mice, in
which
the functions of single genes are annotated; mice with reduced genetic
complexity; and simplified structured populations are tools to gain
fundamental knowledge of inheritance patterns and whole system genetics and
genomics. In this review, we briefly describe existing mouse resources and
discuss their value for fundamental and applied research in livestock.
Collapse
|
83
|
Abstract
Obesity represents the single most important risk factor for early disability and death in developed societies, and the incidence of obesity remains at staggering levels. CNS systems that modulate energy intake and expenditure in response to changes in body energy stores serve to maintain constant body adiposity; the adipocyte-derived hormone leptin and its receptor (LEPR) represent crucial regulators of these systems. As in the case of insulin resistance, a variety of mechanisms (including feedback inhibition, inflammation, gliosis and endoplasmic reticulum stress) have been proposed to interfere with leptin action and impede the systems that control body energy homeostasis to promote or maintain obesity, although the relative importance and contribution of each of these remain unclear. However, LEPR signalling may be increased (rather than impaired) in common obesity, suggesting that any obesity-associated defects in leptin action must result from lesions somewhere other than the initial LEPR signal. It is also possible that increased LEPR signalling could mediate some of the obesity-associated changes in hypothalamic function.
Collapse
|
84
|
Yamazaki T, Li W, Yang L, Li P, Cao H, Motegi SI, Udey MC, Bernhard E, Nakamura T, Mukouyama YS. Whole-Mount Adult Ear Skin Imaging Reveals Defective Neuro-Vascular Branching Morphogenesis in Obese and Type 2 Diabetic Mouse Models. Sci Rep 2018; 8:430. [PMID: 29323138 PMCID: PMC5764985 DOI: 10.1038/s41598-017-18581-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/14/2017] [Indexed: 12/13/2022] Open
Abstract
Obesity and type 2 diabetes are frequently associated with peripheral neuropathy. Though there are multiple methods for diagnosis and analysis of morphological changes of peripheral nerves and blood vessels, three-dimensional high-resolution imaging is necessary to appreciate the pathogenesis with an anatomically recognizable branching morphogenesis and patterning. Here we established a novel technique for whole-mount imaging of adult mouse ear skin to visualize branching morphogenesis and patterning of peripheral nerves and blood vessels. Whole-mount immunostaining of adult mouse ear skin showed that peripheral sensory and sympathetic nerves align with large-diameter blood vessels. Diet-induced obesity (DIO) mice exhibit defective vascular smooth muscle cells (VSMCs) coverage, while there is no significant change in the amount of peripheral nerves. The leptin receptor-deficient db/db mice, a severe obese and type 2 diabetic mouse model, exhibit defective VSMC coverage and a large increase in the amount of smaller-diameter nerve bundles with myelin sheath and unmyelinated nerve fibers. Interestingly, an increase in the amount of myeloid immune cells was observed in the DIO but not db/db mouse skin. These data suggest that our whole-mount imaging method enables us to investigate the neuro-vascular and neuro-immune phenotypes in the animal models of obesity and diabetes.
Collapse
Affiliation(s)
- Tomoko Yamazaki
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA.,Earle A. Chiles Research Institute, Robert W. Franz Cancer Center, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Wenling Li
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Ling Yang
- Laboratory of Obesity and Metabolic Diseases, Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Ping Li
- Laboratory of Obesity and Metabolic Diseases, Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Haiming Cao
- Laboratory of Obesity and Metabolic Diseases, Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Sei-Ichiro Motegi
- Dermatology Branch, National Cancer Institute, 10 Center Drive, Bethesda, MD, 20892, USA.,Department of Dermatology, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan
| | - Mark C Udey
- Dermatology Branch, National Cancer Institute, 10 Center Drive, Bethesda, MD, 20892, USA.,Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Elise Bernhard
- Divisions of Endocrinology, Cincinnati Children's Hospital, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Takahisa Nakamura
- Divisions of Endocrinology, Cincinnati Children's Hospital, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA.,Divisions of Developmental Biology, Cincinnati Children's Hospital, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Yoh-Suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, Genetics and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
85
|
Qiao S, Mao G, Li H, Ma Z, Hong L, Zhang H, Wang C, An J. DPP-4 Inhibitor Sitagliptin Improves Cardiac Function and Glucose Homeostasis and Ameliorates β-Cell Dysfunction Together with Reducing S6K1 Activation and IRS-1 and IRS-2 Degradation in Obesity Female Mice. J Diabetes Res 2018; 2018:3641516. [PMID: 30116740 PMCID: PMC6079488 DOI: 10.1155/2018/3641516] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/08/2018] [Accepted: 06/14/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chronic overnutrition leads to cardiac dysfunction and insulin (INS) resistance. Dipeptidyl peptidase-4 (DPP-4) improves glucose metabolism and insulin sensitivity in both human and animal models. In this study, we explored whether DPP-4 inhibitor sitagliptin (SIT) is involved in the protection of cardiac function and β-cell function using an obesity female mouse model. METHODS Six-week-old C57BL6/J mice were fed a high fat and fructose Western diet with DPP-4 inhibitor SIT for 12 weeks. Cardiac function was examined by echocardiography. Body weight, plasma glucose, and insulin concentrations were measured. The contents of total S6 kinase 1 (S6K1), phosphorylation of S6K1 activation, and INS docking proteins INS receptor substrates 1 and 2 (IRS-1, IRS-2) were assayed, and histology of heart tissue was performed. RESULTS Chronic Western diet consumption elevated plasma glucose and insulin and caused obesity, diastolic dysfunction, and β-cell dysfunction. DPP-4 inhibition with SIT resulted in reduction in body weight, fasting glucose, and plasma insulin, and improved cardiac diastolic dysfunction. SIT also decreased mTOR/S6K1 activation and prevented the degradation of IRS-1 and IRS-2. CONCLUSIONS This study revealed pleiotropic protective effects of DPP-4 inhibitor SIT on cardiac function, glycemia, and β-cell function together with reducing S6K1 activation and IRS-1 and IRS-2 degradation in the obesity female mouse model.
Collapse
Affiliation(s)
- Shigang Qiao
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, No. 199 Renai Road, Suzhou 215123, China
| | - Guofang Mao
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Hua Li
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Zhimin Ma
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Lei Hong
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Huiling Zhang
- Department of Pharmacology, Laboratory of Cerebrovascular Pharmacology, College of Pharmaceutical Science, Soochow University, No. 199 Renai Road, Suzhou 215123, China
| | - Chen Wang
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
- Department of Anesthesiology and Perioperative Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Science and Technology Town Hospital, No. 1 Lijiang Road, Suzhou 215153, China
| |
Collapse
|
86
|
Lee JM, Govindarajah V, Goddard B, Hinge A, Muench DE, Filippi MD, Aronow B, Cancelas JA, Salomonis N, Grimes HL, Reynaud D. Obesity alters the long-term fitness of the hematopoietic stem cell compartment through modulation of Gfi1 expression. J Exp Med 2017; 215:627-644. [PMID: 29282250 PMCID: PMC5789409 DOI: 10.1084/jem.20170690] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/31/2017] [Accepted: 11/16/2017] [Indexed: 12/13/2022] Open
Abstract
Lee et al. show that established obesity alters the composition and long-term fitness of the hematopoietic stem cell (HSC) compartment, in part through a Gfi1-dependent HSC regulatory program that is activated by the chronic oxidative stress associated with this condition. Obesity is a chronic organismal stress that disrupts multiple systemic and tissue-specific functions. In this study, we describe the impact of obesity on the activity of the hematopoietic stem cell (HSC) compartment. We show that obesity alters the composition of the HSC compartment and its activity in response to hematopoietic stress. The impact of obesity on HSC function is progressively acquired but persists after weight loss or transplantation into a normal environment. Mechanistically, we establish that the oxidative stress induced by obesity dysregulates the expression of the transcription factor Gfi1 and that increased Gfi1 expression is required for the abnormal HSC function induced by obesity. These results demonstrate that obesity produces durable changes in HSC function and phenotype and that elevation of Gfi1 expression in response to the oxidative environment is a key driver of the altered HSC properties observed in obesity. Altogether, these data provide phenotypic and mechanistic insight into durable hematopoietic dysregulations resulting from obesity.
Collapse
Affiliation(s)
- Jung-Mi Lee
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Vinothini Govindarajah
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Bryan Goddard
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Ashwini Hinge
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - David E Muench
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Marie-Dominique Filippi
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Bruce Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Jose A Cancelas
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.,Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Nathan Salomonis
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Damien Reynaud
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
87
|
|
88
|
Abstract
Understanding of the neural and physiological substrates of hunger and satiety has increased rapidly over the last three decades, and pharmacological targets have already been identified for the treatment of obesity that has moved from pre-clinical screening to therapies approved by regulatory authorities. Initially, this review describes the way in which physiological signals of energy availability interact with hedonic and rewarding properties of food to modulate the neural circuitry that supports eating behaviour. This is followed by a brief account of current and promising targets for drug development and a review of the wide range of preclinical paradigms that model important influences on human eating behaviour, and can be used to guide early stages of the drug development process.
Collapse
|
89
|
Sun L, Sun Q, Qi J. Adult hippocampal neurogenesis: an important target associated with antidepressant effects of exercise. Rev Neurosci 2017; 28:693-703. [PMID: 28422706 DOI: 10.1515/revneuro-2016-0076] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/08/2017] [Indexed: 12/28/2022]
Abstract
AbstractDepression is a prevalent devastating mental disorder that affects the normal life of patients and brings a heavy burden to whole society. Although many efforts have been made to attenuate depressive/anxiety symptoms, the current clinic antidepressants have limited effects. Scientists have long been making attempts to find some new strategies that can be applied as the alternative antidepressant therapy. Exercise, a widely recognized healthy lifestyle, has been suggested as a therapy that can relieve psychiatric stress. However, how exercise improves the brain functions and reaches the antidepressant target needs systematic summarization due to the complexity and heterogeneous feature of depression. Brain plasticity, especially adult neurogenesis in the hippocampus, is an important neurophysiology to facilitate animals for neurogenesis can occur in not only humans. Many studies indicated that an appropriate level of exercise can promote neurogenesis in the adult brains. In this article, we provide information about the antidepressant effects of exercise and its implications in adult neurogenesis. From the neurogenesis perspective, we summarize evidence about the effects of exercise in enhancing neurogenesis in the hippocampus through regulating growth factors, neurotrophins, neurotransmitters and metabolism as well as inflammations. Taken together, a large number of published works indicate the multiple benefits of exercise in the brain functions of animals, particularly brain plasticity like neurogenesis and synaptogenesis. Therefore, a new treatment method for depression therapy can be developed by regulating the exercise activity.
Collapse
Affiliation(s)
- Lina Sun
- School of PE No. 79, Taiyuan University of Technology, Yingze West Avenue, Taiyuan 030024, Shanxi, China; and Department of Physiology, Shanxi Medical University, China
| | - Qingshan Sun
- School of PE No. 79, Taiyuan University of Technology, Taiyuan 030024, Shanxi, China
| | - Jinshun Qi
- Department of Physiology, Shanxi Medical University, Taiyuan 030024, Shanxi, China
| |
Collapse
|
90
|
Chen H, Xu X, Yang XY, Ling BY, Sun HP, Liu C, Zhang YQ, Cao H, Xu L. Systemic dexmedetomidine attenuates mechanical allodynia through extracellular sign db type 2 diabetic mice. Neurosci Lett 2017; 657:126-133. [PMID: 28757391 DOI: 10.1016/j.neulet.2017.07.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 07/23/2017] [Accepted: 07/26/2017] [Indexed: 10/19/2022]
Abstract
Painful diabetic neuropathy (PDN) is a common complication of diabetes mellitus. However, the treatment for PDN is limited in clinical practice. In the present study, we investigated the effect of systemic administration dexmedetomidine (DEX), a selective alpha 2 adrenergic receptor (α2AR) agonist, on mechanical allodynia and its underlying mechanism in db/db mice, an animal model of type 2 diabetes mellitus. Our data demonstrated that db/db mice develop mechanical allodynia at the early stage of diabetes. During the period of mechanical allodynia, we detected increased release of norepinephrine (NE) and decreased levels of α2A-Adrenoceptors in db/db mice. Immunohistochemistry showed that the α2A-Adrenoceptor is predominantly expressed in neurons in the spinal cord. Acute injection of dexmedetomidine significantly decreased mechanical allodynia, which was blocked by its selective antagonist BRL44408. Furthermore, the upregulation of pERK1 and pERK2 in db/db mice were attenuated by preadministration of dexmedetomidine. We provide the first evidence that the functional alternation of spinal noradrenergic system might underlie exaggerated nociception in PDN. Systemic dexmedetomidine inhibits the mechanical allodynia which is related to ERK signaling pathway in type 2 diabetes, implying that the α2-Adrenoceptor might be a potential therapeutic strategy for PDN.
Collapse
Affiliation(s)
- Hui Chen
- Department of Endocrinology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Xiang Xu
- Department of Endocrinology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Xiao-Yu Yang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 200032, China
| | - Bing-Yu Ling
- Department of Endocrinology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - He-Ping Sun
- Department of Endocrinology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Chao Liu
- Department of Endocrinology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, 214023, China
| | - Yu Qiu Zhang
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 200032, China; Institutes of Integrative Medicine, Fudan University, 200032, China
| | - Hong Cao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and Collaborative Innovation Center for Brain Science, Fudan University, 200032, China; Institutes of Integrative Medicine, Fudan University, 200032, China.
| | - Lan Xu
- Department of Endocrinology, Wuxi People's Hospital, Nanjing Medical University, Wuxi, Jiangsu, 214023, China.
| |
Collapse
|
91
|
Karkhanis A, Holleran KM, Jones SR. Dynorphin/Kappa Opioid Receptor Signaling in Preclinical Models of Alcohol, Drug, and Food Addiction. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:53-88. [PMID: 29056156 DOI: 10.1016/bs.irn.2017.08.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The dynorphin/kappa opioid receptor (KOR) system is implicated in the "dark side" of addiction, in which stress exacerbates maladaptive responses to drug and alcohol exposure. For example, acute stress and acute ethanol exposure result in an elevation in dynorphin, the KOR endogenous ligand. Activation of KORs results in modulation of several neurotransmitters; however, this chapter will focus on its regulatory effects on dopamine in mesolimbic areas. Specifically, KOR activation has an inhibitory effect on dopamine release, thereby influencing reward processing. Repeated stimulation of KORs, for example, via chronic drug and/or stress exposure, results in increased function of the dynorphin/KOR system. This augmentation in KOR function shifts the homeostatic balance in favor of an overall reduction in dopamine signaling via either by reducing dopamine release or by increasing dopamine transporter function. This chapter examines the effects of chronic ethanol exposure on KOR function and the downstream effects on dopamine transmission. Additionally, the impact of chronic cocaine exposure and its effects on KOR function will be explored. Further, KORs may also be involved in driving excessive consumption of food, contributing to the risk of developing obesity. While some studies have shown that KOR agonists reduce drug intake, other studies have shown that antagonists reduce addiction-like behaviors, demonstrating therapeutic potential. For example, KOR inhibition reduces ethanol intake in dependent animals, motivation to self-administer cocaine in chronic stress-exposed animals, and food consumption in obese animals. This chapter will delve into the mechanisms by which modulation of the dynorphin/KOR system may be therapeutic.
Collapse
Affiliation(s)
| | | | - Sara R Jones
- Wake Forest School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
92
|
Abstract
Obesity is a major global epidemic that sets the stage for diverse multiple pathologies, including cardiovascular disease. The obesity-related low-grade chronic inflamed milieu is more pronounced in aging and responsive to cardiac dysfunction in heart failure pathology. Metabolic dysregulation of obesity integrates with immune reservoir in spleen and kidney network. Therefore, an integrative systems biology approach is necessary to delay progressive cardiac alternations. The purpose of this comprehensive review is to largely discuss the impact of obesity on the cardiovascular pathobiology in the context of problems and challenges, with major emphasis on the diversified models, and to study cardiac remodeling in obesity. The information in this article is immensely helpful in teaching advanced undergraduate, graduate, and medical students about the advancement and impact of obesity on cardiovascular health. © 2017 American Physiological Society. Compr Physiol 7:1463-1477, 2017.
Collapse
Affiliation(s)
- Ganesh V Halade
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Alabama, USA
| | - Vasundhara Kain
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
93
|
Andermann ML, Lowell BB. Toward a Wiring Diagram Understanding of Appetite Control. Neuron 2017; 95:757-778. [PMID: 28817798 DOI: 10.1016/j.neuron.2017.06.014] [Citation(s) in RCA: 373] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 01/26/2023]
Abstract
Prior mouse genetic research has set the stage for a deep understanding of appetite regulation. This goal is now being realized through the use of recent technological advances, such as the ability to map connectivity between neurons, manipulate neural activity in real time, and measure neural activity during behavior. Indeed, major progress has been made with regard to meal-related gut control of appetite, arcuate nucleus-based hypothalamic circuits linking energy state to the motivational drive, hunger, and, finally, limbic and cognitive processes that bring about hunger-mediated increases in reward value and perception of food. Unexpected findings are also being made; for example, the rapid regulation of homeostatic neurons by cues that predict future food consumption. The aim of this review is to cover the major underpinnings of appetite regulation, describe recent advances resulting from new technologies, and synthesize these findings into an updated view of appetite regulation.
Collapse
Affiliation(s)
- Mark L Andermann
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
94
|
Yin W, Qin W, Gao Y. Urine glucose levels are disordered before blood glucose level increase was observed in Zucker diabetic fatty rats. SCIENCE CHINA-LIFE SCIENCES 2017; 61:844-848. [DOI: 10.1007/s11427-017-9134-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 06/23/2017] [Indexed: 01/19/2023]
|
95
|
Epigenetic Regulation of Adipokines. Int J Mol Sci 2017; 18:ijms18081740. [PMID: 28796178 PMCID: PMC5578130 DOI: 10.3390/ijms18081740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 08/04/2017] [Accepted: 08/08/2017] [Indexed: 12/29/2022] Open
Abstract
Adipose tissue expansion in obesity leads to changes in the expression of adipokines, adipocyte-specific hormones that can regulate whole body energy metabolism. Epigenetic regulation of gene expression is a mechanism by which cells can alter gene expression through the modifications of DNA and histones. Epigenetic mechanisms, such as DNA methylation and histone modifications, are intimately tied to energy metabolism due to their dependence on metabolic intermediates such as S-adenosylmethionine and acetyl-CoA. Altered expression of adipokines in obesity may be due to epigenetic changes. The goal of this review is to highlight current knowledge of epigenetic regulation of adipokines.
Collapse
|
96
|
Hayashi Y, Toyomasu Y, Saravanaperumal SA, Bardsley MR, Smestad JA, Lorincz A, Eisenman ST, Cipriani G, Nelson Holte MH, Al Khazal FJ, Syed SA, Gajdos GB, Choi KM, Stoltz GJ, Miller KE, Kendrick ML, Rubin BP, Gibbons SJ, Bharucha AE, Linden DR, Maher LJ, Farrugia G, Ordog T. Hyperglycemia Increases Interstitial Cells of Cajal via MAPK1 and MAPK3 Signaling to ETV1 and KIT, Leading to Rapid Gastric Emptying. Gastroenterology 2017; 153:521-535.e20. [PMID: 28438610 PMCID: PMC5526732 DOI: 10.1053/j.gastro.2017.04.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 04/14/2017] [Accepted: 04/17/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND & AIMS Depletion of interstitial cells of Cajal (ICCs) is common in diabetic gastroparesis. However, in approximately 20% of patients with diabetes, gastric emptying (GE) is accelerated. GE also occurs faster in obese individuals, and is associated with increased blood levels of glucose in patients with type 2 diabetes. To understand the fate of ICCs in hyperinsulinemic, hyperglycemic states characterized by rapid GE, we studied mice with mutation of the leptin receptor (Leprdb/db), which in our colony had accelerated GE. We also investigated hyperglycemia-induced signaling in the ICC lineage and ICC dependence on glucose oxidative metabolism in mice with disruption of the succinate dehydrogenase complex, subunit C gene (Sdhc). METHODS Mice were given breath tests to analyze GE of solids. ICCs were studied by flow cytometry, intracellular electrophysiology, isometric contractility measurement, reverse-transcription polymerase chain reaction, immunoblot, immunohistochemistry, enzyme-linked immunosorbent assays, and metabolite assays; cells and tissues were manipulated pharmacologically and by RNA interference. Viable cell counts, proliferation, and apoptosis were determined by methyltetrazolium, Ki-67, proliferating cell nuclear antigen, bromodeoxyuridine, and caspase-Glo 3/7 assays. Sdhc was disrupted in 2 different strains of mice via cre recombinase. RESULTS In obese, hyperglycemic, hyperinsulinemic female Leprdb/db mice, GE was accelerated and gastric ICC and phasic cholinergic responses were increased. Female KitK641E/+ mice, which have genetically induced hyperplasia of ICCs, also had accelerated GE. In isolated cells of the ICC lineage and gastric organotypic cultures, hyperglycemia stimulated proliferation by mitogen-activated protein kinase 1 (MAPK1)- and MAPK3-dependent stabilization of ets variant 1-a master transcription factor for ICCs-and consequent up-regulation of v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) receptor tyrosine kinase. Opposite changes occurred in mice with disruption of Sdhc. CONCLUSIONS Hyperglycemia increases ICCs via oxidative metabolism-dependent, MAPK1- and MAPK3-mediated stabilization of ets variant 1 and increased expression of KIT, causing rapid GE. Increases in ICCs might contribute to the acceleration in GE observed in some patients with diabetes.
Collapse
Affiliation(s)
- Yujiro Hayashi
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Yoshitaka Toyomasu
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Siva Arumugam Saravanaperumal
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Michael R. Bardsley
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - John A. Smestad
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Andrea Lorincz
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Fatimah J. Al Khazal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Sabriya A. Syed
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Gabriella B. Gajdos
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota,Gastroenterology Research Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Kyoung Moo Choi
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota
| | - Gary J. Stoltz
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Katie E. Miller
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | | | - Brian P. Rubin
- Departments of Anatomic Pathology and Cancer Biology, Robert J. Tomsich Pathology and Laboratory Medicine Institute, Lerner Research Institute and Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Adil E. Bharucha
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - David R. Linden
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Louis James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | | | - Tamas Ordog
- Enteric Neuroscience Program, Mayo Clinic, Rochester, Minnesota; Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota; Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota.
| |
Collapse
|
97
|
Deletion of Suppressor of Cytokine Signaling 3 from Forebrain Neurons Delays Infertility and Onset of Hypothalamic Leptin Resistance in Response to a High Caloric Diet. J Neurosci 2017; 36:7142-53. [PMID: 27383590 DOI: 10.1523/jneurosci.2714-14.2016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 05/27/2016] [Indexed: 12/19/2022] Open
Abstract
UNLABELLED The cellular processes that cause high caloric diet (HCD)-induced infertility are poorly understood but may involve upregulation of suppressor of cytokine signaling (SOCS-3) proteins that are associated with hypothalamic leptin resistance. Deletion of SOCS-3 from brain cells is known to protect mice from diet-induced obesity, but the effects on HCD-induced infertility are unknown. We used neuron-specific SOCS3 knock-out mice to elucidate this and the effects on regional hypothalamic leptin resistance. As expected, male and female neuron-specific SOCS3 knock-out mice were protected from HCD-induced obesity. While female wild-type mice became infertile after 4 months of HCD feeding, infertility onset in knock-out females was delayed by 4 weeks. Similarly, knock-out mice had delayed leptin resistance development in the medial preoptic area and anteroventral periventricular nucleus, regions important for generation of the surge of GnRH and LH that induces ovulation. We therefore tested whether the suppressive effects of HCD on the estradiol-induced GnRH/LH surge were overcome by neuron-specific SOCS3 knock-out. Although only 20% of control HCD-mice experienced a preovulatory-like LH surge, LH surges could be induced in almost all neuron-specific SOCS3 knock-out mice on this diet. In contrast to females, HCD-fed male mice did not exhibit any fertility decline compared with low caloric diet-fed males despite their resistance to the satiety effects of leptin. These data show that deletion of SOCS3 delays the onset of leptin resistance and infertility in HCD-fed female mice, but given continued HCD feeding this state does eventually occur, presumably in response to other mechanisms inhibiting leptin signal transduction. SIGNIFICANCE STATEMENT Obesity is commonly associated with infertility in humans and other animals. Treatments for human infertility show a decreased success rate with increasing body mass index. A hallmark of obesity is an increase in circulating leptin levels; despite this, the brain responds as if there were low levels of leptin, leading to increased appetite and suppressed fertility. Here we show that leptin resistant infertility is caused in part by the leptin signaling molecule SOCS3. Deletion of SOCS3 from brain neurons delays the onset of diet-induced infertility.
Collapse
|
98
|
Assessment of gut microbiota populations in lean and obese Zucker rats. PLoS One 2017; 12:e0181451. [PMID: 28704522 PMCID: PMC5509373 DOI: 10.1371/journal.pone.0181451] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/01/2017] [Indexed: 12/28/2022] Open
Abstract
Obesity has been on the rise in the US and worldwide for the last several decades. Obesity has been associated with chronic disease development, such as certain types of cancer, type 2 diabetes, cardiovascular disease, and liver diseases. Previously, we reported that obesity promotes DMBA-induced mammary tumor development using the obese Zucker rat model. The intestinal microbiota is composed of a diverse population of obligate and facultative anaerobic microorganisms, and these organisms carry out a broad range of metabolic activities. Obesity has been linked to changes in the intestinal microbiota, but the composition of the bacterial populations in lean and obese Zucker rats has not been carefully studied. Therefore, the objective of this study was to determine the effects of obesity on the gut microbiota in this model. Lean and obese female Zucker rats (n = 16) were fed an AIN-93G-like diet for 8 weeks. Rats were weighed twice weekly, and fecal samples were collected at the beginning and end of the experiment. 16S rRNA gene sequencing was used to evaluate the composition of the fecal bacterial populations. At the outset of the study, the lean rats exhibited much lower ratios of the Firmicutes to Bacteroidetes phyla than the obese rats, but after 60 days, this ratio in the lean rats exceeded that of the obese. This shift was associated with reductions in the Bacteroidaceae, S24-7 and Paraprevotellaceae families in the lean rats. Obese rats also showed increased levels of the genus Akkermansia at day 60. PCoA plots of beta diversity showed clustering of the different test groups, indicating clear differences in intestinal microbiota populations associated with both the time point of the study and the lean or obese status in the Zucker rat model for obesity.
Collapse
|
99
|
Gao C, Wang Q, Chung SK, Shen J. Crosstalk of metabolic factors and neurogenic signaling in adult neurogenesis: Implication of metabolic regulation for mental and neurological diseases. Neurochem Int 2017; 106:24-36. [DOI: 10.1016/j.neuint.2017.02.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/10/2017] [Accepted: 02/03/2017] [Indexed: 12/31/2022]
|
100
|
Ramos-Lobo AM, Donato J. The role of leptin in health and disease. Temperature (Austin) 2017; 4:258-291. [PMID: 28944270 DOI: 10.1080/23328940.2017.1327003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Leptin is a master regulator of energy balance and body adiposity. Additionally, leptin exerts important control on glucose homeostasis, thermogenesis, autonomic nervous system and neuroendocrine axes. In metabolic diseases, such as obesity and diabetes mellitus, leptin signaling may be compromised, indicating the important role of this hormone in the etiology and pathophysiological manifestations of these conditions. In the present manuscript, we reviewed important concepts of leptin signaling, as well as about the effects of leptin on several biologic functions. We also discussed the possible therapeutic use of leptin administration and how our current obesogenic environment contributes to the development of leptin resistance. Our objective was to provide a comprehensive and state-of-the-art review about the importance of leptin to maintain the homeostasis and during pathological conditions.
Collapse
Affiliation(s)
- Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|