1
|
Srour N, Caron A, Michael NJ. Do POMC neurons have a sweet tooth for leptin? Special issue: Role of nutrients in nervous control of energy balance. Biochimie 2024; 223:179-187. [PMID: 36122808 DOI: 10.1016/j.biochi.2022.09.006] [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: 05/21/2022] [Revised: 08/29/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022]
Abstract
Coordinated detection of changes in metabolic state by the nervous system is fundamental for survival. Hypothalamic pro-opiomelanocortin (POMC) neurons play a critical role in integrating metabolic signals, including leptin levels. They also coordinate adaptative responses and thus represent an important relay in the regulation of energy balance. Despite a plethora of work documenting the effects of individual hormones, nutrients, and neuropeptides on POMC neurons, the importance for crosstalk and additive effects between such signaling molecules is still underexplored. The ability of the metabolic state and the concentrations of nutrients, such as glucose, to influence leptin's effects on POMC neurons appears critical for understanding the function and complexity of this regulatory network. Here, we summarize the current knowledge on the effects of leptin on POMC neuron electrical excitability and discuss factors potentially contributing to variability in these effects, with a particular focus on the mouse models that have been developed and the importance of extracellular glucose levels. This review highlights the importance of the metabolic "environment" for determining hypothalamic neuronal responsiveness to metabolic cues and for determining the fundamental effects of leptin on the activity of hypothalamic POMC neurons.
Collapse
Affiliation(s)
- Nader Srour
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada; Montreal Diabetes Research Center, QC, Canada.
| | - Natalie Jane Michael
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, 2725 chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada; Faculté de Pharmacie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
2
|
Purnell JQ, le Roux CW. Hypothalamic control of body fat mass by food intake: The key to understanding why obesity should be treated as a disease. Diabetes Obes Metab 2024; 26 Suppl 2:3-12. [PMID: 38351898 DOI: 10.1111/dom.15478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/06/2024] [Accepted: 01/18/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Hypothalamic centres have been recognized to play a central role in body weight regulation for nearly 70 years. AIMS In this review, we will explore the current undersanding of the role the hypothalamus plays in controlling food intake behaviours. MATERIALS AND METHODS Review of relevant literature from PubMed searches and review article citations. RESULTS Beginning with autopsy studies showing destructive hypothalamic lesions in patients manifesting hyperphagia and rapid weight gain, followed by animal lesioning studies pinpointing adjacent hypothalamic sites as the 'satiety' centre and the 'feeding' centre of the brain, the neurocircuitry that governs our body weight is now understood to consist of a complex, interconnected network, including the hypothalamus and extending to cortical sites, reward centres and brainstem. Neurons in these sites receive afferent signals from the gastrointestinal tract and adipose tissue indicating food availability, calorie content, as well as body fat mass. DISCUSSION Integration of these complex signals leads to modulation of the two prime effector systems that defend a body fat mass set point: food intake and energy expenditure. CONCLUSION Understanding the hypothalamic control of food intake forms the foundation for understanding and managing obesity as a chronic disease.
Collapse
Affiliation(s)
- Jonathan Q Purnell
- Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Carel W le Roux
- School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
3
|
Luo M, Guo L, Wu C, Hao M, Gu J, Li X, Wang Q. Effects of dietary copper intake on blood lipids in women of childbearing age and the potential role of gut microbiota. Front Nutr 2024; 11:1368730. [PMID: 38505268 PMCID: PMC10948407 DOI: 10.3389/fnut.2024.1368730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/16/2024] [Indexed: 03/21/2024] Open
Abstract
Background Copper (Cu) is a vital trace element involved in numerous physiological processes, including glycolysis and lipid metabolism. Imbalances in Cu homeostasis can contribute to various diseases. However, current research on the impact of Cu on lipid metabolism has yielded inconsistent findings. Moreover, studies investigating the effects of dietary Cu intake on blood lipids among women of childbearing age are rare. Understanding of this relationship could enhance lipid management, given that most women obtain Cu through their diet. Additionally, the gut microbiota may play a role in this process. This study aims to investigate the effects of dietary Cu intake on blood lipids in women of childbearing age and to analyze the role of gut microbiota in this process. Methods This study utilized data from the National Health and Nutrition Examination Survey (NHANES) to conduct a preliminary analysis of the correlation between dietary Cu levels and blood lipid indicators in women of childbearing age. Subsequently, an on-site research was conducted to further investigate this relationship, followed by animal experiments to verify the effect of different Cu doses on blood lipid levels. Multiple linear regression models, ANOVA, XGBOOST were employed to analyze the impact of Cu on blood lipids and the role of intestinal microbiota in this process. Results In the population study, the NHANES results were consistent with on-site findings. The TG, and TC levels in women with childbearing were increased with higher dietary Cu intake. Animal experiments have shown that as Cu intake increases, TC levels increase. Furthermore, when the Cu intake reached 8 mg/day (the recommended dietary Cu intake limit of China, RDI), the TG levels in the research animals decrease, alongside a reduction in the abundance of Weissella cibaria (probiotics related to lipid metabolism), and the levels of LPS and IL-6 increase. Conclusion The blood lipid levels of women of childbearing age increase with higher dietary Cu intake. RDI of 8 mg/day for women of childbearing age in China may need to be appropriately reduced. Regulating the gut microbiota, especially by increasing the abundance of Weissella cibaria may be an effective intervention for blood lipids.
Collapse
Affiliation(s)
- Mingming Luo
- Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Jiangxi Cancer Institute, Nanchang, China
| | - Linmei Guo
- Department of Health Statistics, School of Public Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Chunmei Wu
- Department of Health Statistics, School of Public Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Ming Hao
- Department of Health Statistics, School of Public Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Junwang Gu
- Department of Health Statistics, School of Public Health and Health Management, Gannan Medical University, Ganzhou, China
| | - Xuhuan Li
- The Fourth Affiliated Hospital of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qi Wang
- Department of Health Statistics, School of Public Health and Health Management, Gannan Medical University, Ganzhou, China
| |
Collapse
|
4
|
Riera CE. Wiring the Brain for Wellness: Sensory Integration in Feeding and Thermogenesis: A Report on Research Supported by Pathway to Stop Diabetes. Diabetes 2024; 73:338-347. [PMID: 38377445 PMCID: PMC10882152 DOI: 10.2337/db23-0706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/06/2023] [Indexed: 02/22/2024]
Abstract
The recognition of sensory signals from within the body (interoceptive) and from the external environment (exteroceptive), along with the integration of these cues by the central nervous system, plays a crucial role in maintaining metabolic balance. This orchestration is vital for regulating processes related to both food intake and energy expenditure. Animal model studies indicate that manipulating specific populations of neurons in the central nervous system which influence these processes can effectively modify energy balance. This body of work presents an opportunity for the development of innovative weight loss therapies for the treatment of obesity and type 2 diabetes. In this overview, we delve into the sensory cues and the neuronal populations responsible for their integration, exploring their potential in the development of weight loss treatments for obesity and type 2 diabetes. This article is the first in a series of Perspectives that report on research funded by the American Diabetes Association Pathway to Stop Diabetes program. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Céline E. Riera
- Center for Neural Science and Medicine, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
5
|
Simon JJ, Lang PM, Rommerskirchen L, Bendszus M, Friederich HC. Hypothalamic Reactivity and Connectivity following Intravenous Glucose Administration. Int J Mol Sci 2023; 24:ijms24087370. [PMID: 37108533 PMCID: PMC10139105 DOI: 10.3390/ijms24087370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 04/29/2023] Open
Abstract
Dysfunctional glucose sensing in homeostatic brain regions such as the hypothalamus is interlinked with the pathogenesis of obesity and type 2 diabetes mellitus. However, the physiology and pathophysiology of glucose sensing and neuronal homeostatic regulation remain insufficiently understood. To provide a better understanding of glucose signaling to the brain, we assessed the responsivity of the hypothalamus (i.e., the core region of homeostatic control) and its interaction with mesocorticolimbic brain regions in 31 normal-weight, healthy participants. We employed a single-blind, randomized, crossover design of the intravenous infusion of glucose and saline during fMRI. This approach allows to investigate glucose signaling independent of digestive processes. Hypothalamic reactivity and connectivity were assessed using a pseudo-pharmacological design and a glycemia-dependent functional connectivity analysis, respectively. In line with previous studies, we observed a hypothalamic response to glucose infusion which was negatively related to fasting insulin levels. The observed effect size was smaller than in previous studies employing oral or intragastric administration of glucose, demonstrating the important role of the digestive process in homeostatic signaling. Finally, we were able to observe hypothalamic connectivity with reward-related brain regions. Given the small amount of glucose employed, this points toward a high responsiveness of these regions to even a small energy stimulus in healthy individuals. Our study highlights the intricate relationship between homeostatic and reward-related systems and their pronounced sensitivity to subtle changes in glycemia.
Collapse
Affiliation(s)
- Joe J Simon
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Pia M Lang
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Lena Rommerskirchen
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Martin Bendszus
- Department of Neuroradiology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Hans-Christoph Friederich
- Department of General Internal Medicine and Psychosomatics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
6
|
dSec16 Acting in Insulin-like Peptide Producing Cells Controls Energy Homeostasis in Drosophila. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010081. [PMID: 36676030 PMCID: PMC9862641 DOI: 10.3390/life13010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/29/2022] [Accepted: 12/08/2022] [Indexed: 12/29/2022]
Abstract
Many studies show that genetics play a major contribution to the onset of obesity. Human genome-wide association studies (GWASs) have identified hundreds of genes that are associated with obesity. However, the majority of them have not been functionally validated. SEC16B has been identified in multiple obesity GWASs but its physiological role in energy homeostasis remains unknown. Here, we use Drosophila to determine the physiological functions of dSec16 in energy metabolism. Our results showed that global RNAi of dSec16 increased food intake and triglyceride (TAG) levels. Furthermore, this TAG increase was observed in flies with a specific RNAi of dSec16 in insulin-like peptide producing cells (IPCs) with an alteration of endocrine peptides. Together, our study demonstrates that dSec16 acting in IPCs controls energy balance and advances the molecular understanding of obesity.
Collapse
|
7
|
Chan K, Wong FS, Pearson JA. Circadian rhythms and pancreas physiology: A review. Front Endocrinol (Lausanne) 2022; 13:920261. [PMID: 36034454 PMCID: PMC9399605 DOI: 10.3389/fendo.2022.920261] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/21/2022] [Indexed: 11/29/2022] Open
Abstract
Type 2 diabetes mellitus, obesity and metabolic syndrome are becoming more prevalent worldwide and will present an increasingly challenging burden on healthcare systems. These interlinked metabolic abnormalities predispose affected individuals to a plethora of complications and comorbidities. Furthermore, diabetes is estimated by the World Health Organization to have caused 1.5 million deaths in 2019, with this figure projected to rise in coming years. This highlights the need for further research into the management of metabolic diseases and their complications. Studies on circadian rhythms, referring to physiological and behavioral changes which repeat approximately every 24 hours, may provide important insight into managing metabolic disease. Epidemiological studies show that populations who are at risk of circadian disruption such as night shift workers and regular long-haul flyers are also at an elevated risk of metabolic abnormalities such as insulin resistance and obesity. Aberrant expression of circadian genes appears to contribute to the dysregulation of metabolic functions such as insulin secretion, glucose homeostasis and energy expenditure. The potential clinical implications of these findings have been highlighted in animal studies and pilot studies in humans giving rise to the development of circadian interventions strategies including chronotherapy (time-specific therapy), time-restricted feeding, and circadian molecule stabilizers/analogues. Research into these areas will provide insights into the future of circadian medicine in metabolic diseases. In this review, we discuss the physiology of metabolism and the role of circadian timing in regulating these metabolic functions. Also, we review the clinical aspects of circadian physiology and the impact that ongoing and future research may have on the management of metabolic disease.
Collapse
Affiliation(s)
- Karl Chan
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - F. Susan Wong
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Systems Immunity Research Institute, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - James Alexander Pearson
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
8
|
Alcantara IC, Tapia APM, Aponte Y, Krashes MJ. Acts of appetite: neural circuits governing the appetitive, consummatory, and terminating phases of feeding. Nat Metab 2022; 4:836-847. [PMID: 35879462 PMCID: PMC10852214 DOI: 10.1038/s42255-022-00611-y] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/16/2022] [Indexed: 12/11/2022]
Abstract
The overconsumption of highly caloric and palatable foods has caused a surge in obesity rates in the past half century, thereby posing a healthcare challenge due to the array of comorbidities linked to heightened body fat accrual. Developing treatments to manage body weight requires a grasp of the neurobiological basis of appetite. In this Review, we discuss advances in neuroscience that have identified brain regions and neural circuits that coordinate distinct phases of eating: food procurement, food consumption, and meal termination. While pioneering work identified several hypothalamic nuclei to be involved in feeding, more recent studies have explored how neuronal populations beyond the hypothalamus, such as the mesolimbic pathway and nodes in the hindbrain, interconnect to modulate appetite. We also examine how long-term exposure to a calorically dense diet rewires feeding circuits and alters the response of motivational systems to food. Understanding how the nervous system regulates eating behaviour will bolster the development of medical strategies that will help individuals to maintain a healthy body weight.
Collapse
Affiliation(s)
- Ivan C Alcantara
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA
- Department of Neuroscience, Brown University, Providence, RI, USA
| | | | - Yeka Aponte
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Michael J Krashes
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, Bethesda, MD, USA.
- National Institute on Drug Abuse (NIDA), National Institutes of Health, Baltimore, MD, USA.
| |
Collapse
|
9
|
Kovac M, Vladimirovna Ippolitova T, Pozyabin S, Aliev R, Lobanova V, Drakul N, S. Rutland C. Equine Stress: Neuroendocrine Physiology and Pathophysiology. Vet Med Sci 2022. [DOI: 10.5772/intechopen.105045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
This review presents new aspects to understanding the neuroendocrine regulation of equine stress responses, and their influences on the physiological, pathophysiological, and behavioral processes. Horse management, in essence, is more frequently confirmed by external and internal stress factors, than in other domestic animals. Regardless of the nature of the stimulus, the equine stress response is an effective and highly conservative set of interconnected relationships designed to maintain physiological integrity even in the most challenging circumstances (e.g., orthopedic injuries, abdominal pain, transport, competitions, weaning, surgery, and inflammation). The equine stress response is commonly a complementary homeostatic mechanism that provides protection (not an adaptation) when the body is disturbed or threatened. It activates numerous neural and hormonal networks to optimize metabolic, cardiovascular, musculoskeletal, and immunological functions. This review looks into the various mechanisms involved in stress responses, stress-related diseases, and assessment, prevention or control, and management of these diseases and stress. Stress-related diseases can not only be identified and assessed better, given the latest research and techniques but also prevented or controlled.
Collapse
|
10
|
Stępniowska A, Tutaj K, Juśkiewicz J, Ognik K. Effect of a high-fat diet and chromium on hormones level and Cr retention in rats. J Endocrinol Invest 2022; 45:527-535. [PMID: 34550535 PMCID: PMC8850218 DOI: 10.1007/s40618-021-01677-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/12/2021] [Indexed: 12/17/2022]
Abstract
AIMS The aim of the study was to determine how the administration of a high-fat diet supplemented with various forms of chromium to rats affects accumulation of this element in the tissues and levels of leptin, ghrelin, insulin, glucagon, serotonin, noradrenaline and histamine, as well as selected mineral elements. METHODS The experiment was conducted on 56 male Wistar rats, which were divided into 8 experimental groups. The rats received standard diet or high fat diet (HFD) with addition of 0.3 mg/kg body weight of chromium(III) picolinate (Cr-Pic), chromium(III)-methioninate (Cr-Met), or chromium nanoparticles (Cr-NP). RESULTS Chromium in organic forms was found to be better retained in the body of rats than Cr in nanoparticles form. However, Cr-Pic was the only form that increased the insulin level, which indicates its beneficial effect on carbohydrate metabolism. In blood plasma of rats fed a high-fat diet noted an increased level of serotonin and a reduced level of noradrenaline. The addition of Cr to the diet, irrespective of its form, also increased the serotonin level, which should be considered a beneficial effect. Rats fed a high-fat diet had an unfavourable reduction in the plasma concentrations of Ca, P, Mg and Zn. The reduction of P in the plasma induced by supplementation with Cr in the form of Cr-Pic or Cr-NP may exacerbate the adverse effect of a high-fat diet on the level of this element. CONCLUSION A high-fat diet was shown to negatively affect the level of hormones regulating carbohydrate metabolism (increasing leptin levels and decreasing levels of ghrelin and insulin).
Collapse
Affiliation(s)
- A Stępniowska
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland.
| | - K Tutaj
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| | - J Juśkiewicz
- Division of Food Science, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - K Ognik
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| |
Collapse
|
11
|
Martins-Oliveira M, Tavares I, Goadsby PJ. Was it something I ate? Understanding the bidirectional interaction of migraine and appetite neural circuits. Brain Res 2021; 1770:147629. [PMID: 34428465 DOI: 10.1016/j.brainres.2021.147629] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/18/2022]
Abstract
Migraine attacks can involve changes of appetite: while fasting or skipping meals are often reported triggers in susceptible individuals, hunger or food craving are reported in the premonitory phase. Over the last decade, there has been a growing interest and recognition of the importance of studying these overlapping fields of neuroscience, which has led to novel findings. The data suggest additional studies are needed to unravel key neurobiological mechanisms underlying the bidirectional interaction between migraine and appetite. Herein, we review information about the metabolic migraine phenotype and explore migraine therapeutic targets that have a strong input on appetite neuronal circuits, including the calcitonin gene-related peptide (CGRP), the pituitary adenylate cyclase-activating polypeptide (PACAP) and the orexins. Furthermore, we focus on potential therapeutic peptide targets that are involved in regulation of feeding and play a role in migraine pathophysiology, such as neuropeptide Y, insulin, glucagon and leptin. We then examine the orexigenic - anorexigenic circuit feedback loop and explore glucose metabolism disturbances. Additionally, it is proposed a different perspective on the most reported feeding-related trigger - skipping meals - as well as a link between contrasting feeding behaviors (skipping meals vs food craving). Our review aims to increase awareness of migraine through the lens of appetite neurobiology in order to improve our understanding of the earlier phase of migraine, encourage better studies and cross-disciplinary collaborations, and provide novel migraine-specific therapeutic opportunities.
Collapse
Affiliation(s)
- Margarida Martins-Oliveira
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK; Nutrition and Metabolism Department, NOVA Medical School, Faculdade de Ciências Médicas de Lisboa, Universidade Nova de Lisboa, Campo Mártires da Pátria 130, 1169-056 Lisbon, Portugal.
| | - Isaura Tavares
- Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; Institute of Investigation and Innovation in Health (i3S), University of Porto, Portugal.
| | - Peter J Goadsby
- Headache Group, Wolfson Centre for Age-Related Disease, Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK; Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Wang L, Liu Y, Stratigopoulos G, Panigrahi S, Sui L, Zhang Y, Leduc CA, Glover HJ, De Rosa MC, Burnett LC, Williams DJ, Shang L, Goland R, Tsang SH, Wardlaw S, Egli D, Zheng D, Doege CA, Leibel RL. Bardet-Biedl syndrome proteins regulate intracellular signaling and neuronal function in patient-specific iPSC-derived neurons. J Clin Invest 2021; 131:146287. [PMID: 33630762 DOI: 10.1172/jci146287] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/23/2021] [Indexed: 12/11/2022] Open
Abstract
Bardet-Biedl syndrome (BBS) is a rare autosomal recessive disorder caused by mutations in genes encoding components of the primary cilium and is characterized by hyperphagic obesity. To investigate the molecular basis of obesity in human BBS, we developed a cellular model of BBS using induced pluripotent stem cell-derived (iPSC-derived) hypothalamic arcuate-like neurons. BBS mutations BBS1M390R and BBS10C91fsX95 did not affect neuronal differentiation efficiency but caused morphological defects, including impaired neurite outgrowth and longer primary cilia. Single-cell RNA sequencing of BBS1M390R hypothalamic neurons identified several downregulated pathways, including insulin and cAMP signaling and axon guidance. Additional studies demonstrated that BBS1M390R and BBS10C91fsX95 mutations impaired insulin signaling in both human fibroblasts and iPSC-derived neurons. Overexpression of intact BBS10 fully restored insulin signaling by restoring insulin receptor tyrosine phosphorylation in BBS10C91fsX95 neurons. Moreover, mutations in BBS1 and BBS10 impaired leptin-mediated p-STAT3 activation in iPSC-derived hypothalamic neurons. Correction of the BBS mutation by CRISPR rescued leptin signaling. POMC expression and neuropeptide production were decreased in BBS1M390R and BBS10C91fsX95 iPSC-derived hypothalamic neurons. In the aggregate, these data provide insights into the anatomic and functional mechanisms by which components of the BBSome in CNS primary cilia mediate effects on energy homeostasis.
Collapse
Affiliation(s)
- Liheng Wang
- Naomi Berrie Diabetes Center and.,Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Yang Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - George Stratigopoulos
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Sunil Panigrahi
- Naomi Berrie Diabetes Center and.,Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Lina Sui
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Yiying Zhang
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Charles A Leduc
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Hannah J Glover
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Maria Caterina De Rosa
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| | - Lisa C Burnett
- Naomi Berrie Diabetes Center and.,Levo Therapeutics, Skokie, Illinois, USA
| | - Damian J Williams
- Institute for Genomic Medicine, Columbia University, New York, New York, USA
| | - Linshan Shang
- Naomi Berrie Diabetes Center and.,Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Stephen H Tsang
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology, Columbia University, New York, New York, USA.,Columbia Stem Cell Initiative and.,Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Sharon Wardlaw
- Naomi Berrie Diabetes Center and.,Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Dieter Egli
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA.,New York Stem Cell Foundation Research Institute, New York, New York, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Neurology and Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Claudia A Doege
- Naomi Berrie Diabetes Center and.,Columbia Stem Cell Initiative and.,Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Rudolph L Leibel
- Naomi Berrie Diabetes Center and.,Division of Molecular Genetics, Department of Pediatrics, Columbia University, College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
13
|
Angiopoietin-Like Growth Factor Involved in Leptin Signaling in the Hypothalamus. Int J Mol Sci 2021; 22:ijms22073443. [PMID: 33810547 PMCID: PMC8037945 DOI: 10.3390/ijms22073443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/20/2021] [Accepted: 03/24/2021] [Indexed: 11/26/2022] Open
Abstract
The hypothalamic regulation of appetite governs whole-body energy balance. Satiety is regulated by endocrine factors including leptin, and impaired leptin signaling is associated with obesity. Despite the anorectic effect of leptin through the regulation of the hypothalamic feeding circuit, a distinct downstream mediator of leptin signaling in neuron remains unclear. Angiopoietin-like growth factor (AGF) is a peripheral activator of energy expenditure and antagonizes obesity. However, the regulation of AGF expression in brain and localization to mediate anorectic signaling is unknown. Here, we demonstrated that AGF is expressed in proopiomelanocortin (POMC)-expressing neurons located in the arcuate nucleus (ARC) of the hypothalamus. Unlike other brain regions, hypothalamic AGF expression is stimulated by leptin-induced signal transducers and activators of transcription 3 (STAT3) phosphorylation. In addition, leptin treatment to hypothalamic N1 cells significantly enhanced the promoter activity of AGF. This induction was abolished by the pretreatment of ruxolitinib, a leptin signaling inhibitor. These results indicate that hypothalamic AGF expression is induced by leptin and colocalized to POMC neurons.
Collapse
|
14
|
Increased plasma asprosin levels in patients with drug-naive anorexia nervosa. Eat Weight Disord 2021; 26:313-321. [PMID: 32026376 DOI: 10.1007/s40519-020-00845-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 01/07/2020] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Asprosin is a centrally acting appetite-promoting hormone and promotes glucose production in the liver. This study is the first to investigate the difference in asprosin in the plasma between anorexia nervosa (AN) and healthy controls, and to explore the relationship between asprosin changes and plasma glucose levels and AN symptoms. METHODS Plasma asprosin and glucose concentrations were detected in AN patients (n = 46) and healthy control subjects (n = 47). Eating Disorder Inventory-2 (EDI-2) was used to assess subjects' eating disorder symptoms and related personality traits. The patient's concomitant levels of depression and anxiety were also measured using the beck depression inventory and beck anxiety inventory, respectively. RESULTS Results indicate that AN patients had a higher asprosin concentration in their plasma compared to healthy controls (p = 0.033). Among AN patients, plasma asprosin levels correlated positively with EDI-2 interoceptive awareness subscale score (p = 0.030) and negatively with duration of illness (p = 0.036). Multiple linear regression analyses showed that increases in asprosin levels (p = 0.029), glucose levels (p = 0.024) and body mass index (p = 0.003) were associated with an increase of the score of EDI-2 bulimia subscale. CONCLUSIONS Our findings suggest that the increase in plasma asprosin concentration in patients with AN may be a compensation for the body's energy shortage, and asprosin may be involved in the development of bulimia and lack of interoceptive awareness in AN patients. LEVEL OF EVIDENCE Level III, case-control analytic study.
Collapse
|
15
|
Synchronous neuronal interactions in rat hypothalamic culture: a novel model for the study of network dynamics in metabolic disorders. Exp Brain Res 2021; 239:755-764. [PMID: 33388905 DOI: 10.1007/s00221-020-05977-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/29/2020] [Indexed: 12/23/2022]
Abstract
Synchronous neural activity is a feature of normal brain function, and altered synchronization is observed in several neurological diseases. Dysfunction in hypothalamic pathways leads to obesity, suggesting that hypothalamic neural synchrony is critical for energy homeostasis. The lateral hypothalamic orexin neurons are extensively interconnected with other brain structures and are important for energy balance. Earlier studies show that rats with higher orexin sensitivity are obesity resistant. Similarly, topiramate, an anti-epileptic drug, has been shown to reduce weight in humans. Since orexin enhances neuronal excitation, we hypothesized that obesity-resistant rats with higher orexin sensitivity may exhibit enhanced hypothalamic synchronization. We further hypothesized that anti-obesity agents such as orexin and topiramate will enhance hypothalamic synchronization. To test this, we examined neural synchronicity in primary embryonic hypothalamic cell cultures, obtained from embryonic day 18 (E18) obesity-susceptible Sprague-Dawley (SD) and obesity-resistant rats. Hypothalamic tissue was cultured in multielectrode array (MEA), and recordings were performed twice weekly, from 4th to 32nd day in vitro (DIV). Next, we tested the effects of orexin and topiramate application on neural synchronicity of hypothalamic cultures obtained from SD rat embryos. Signals were analyzed for synchronization using cross correlation. Our results showed that (1) obesity-resistant hypothalamus exhibits significantly higher synchronization compared to obesity-sensitive hypothalamus; and (2) orexin and topiramate enhance hypothalamic synchronization. These results support that enhanced orexin sensitivity is associated with greater neural synchronization, and that anti-obesity treatments enhance network synchronization, thus constrain variability in hypothalamic output signals, to extrahypothalamic structures involved in energy homeostasis.
Collapse
|
16
|
Ahmed Mustafa Z, Hamed Ali R, Rostum Ali D, Abdulkarimi R, Abdulkareem NK, Akbari A. The combination of ginger powder and zinc supplement improves the fructose-induced metabolic syndrome in rats by modulating the hepatic expression of NF-κB, mTORC1, PPAR-α SREBP-1c, and Nrf2. J Food Biochem 2021; 45:e13546. [PMID: 33145794 DOI: 10.1111/jfbc.13546] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/02/2020] [Accepted: 10/12/2020] [Indexed: 12/14/2022]
Abstract
Although studies have shown that ginger, as an herbal remedy and zinc are able to improve inflammation, oxidative stress, autophagy, and metabolism of lipid and glucose, their molecular mechanisms are unknown. Therefore, this study was aimed to examine the therapeutic effects of ginger with zinc supplement for eight weeks on fructose-induced metabolic syndrome (MS). Ninety-six adult male Sprague Dawley rats (220 g ± 20) were randomly assigned to twelve controlled and treated groups. After the last treatment session, the level of lipid profiles, glucose, insulin, and leptin as metabolic factors and liver enzymes as biomarkers to evaluate liver function in serum were measured. The level of antioxidant enzymes and lipid peroxidation to evaluate the oxidative status and the TNF-α level as a biomarker to assess the state of inflammation in liver were also measured. The level of zinc along with the expression of NF-κB, mTORC1, PPAR-α, SREBP-1c, and Nrf2 in liver was also evaluated. The level of metabolic factors and liver enzymes in serum along with lipid peroxidation and TNF-α in liver increased; zinc and antioxidant enzymes levels decreased in rats with MS compared to control rats (p < .05). The hepatic expression of SREBP-1c, NF-κB and mTORC1 were upregulated and the expression of PPAR-α and Nrf2 were downregulated in rats with MS compared to control rats (p < .05). Treatment with different doses of ginger, zinc, and the combination of them could improve metabolic, inflammatory oxidative stress factors, and expression of the above genes in rats with MS compared to the MS group (p < .05). It can be concluded that ginger, zinc, and the combination of them could improve oxidative damage, inflammation, and autophagy induced by fructose and could adjust the glucose and lipid metabolism and the homeostasis of zinc in rats with MS. PRACTICAL APPLICATIONS: Due to the increasing prevalence of metabolic diseases, the use of plant compounds such as ginger has attracted widespread attention. Ginger as an herbal remedy with predominant pharmacological properties due to its availability, cheapness, and lack of side effects is also very popular for the treatment of metabolic disorders in folk medicine. Moreover, enhancing its medicinal properties with supplements such as zinc can be widely welcomed. This study was actually performed with the aim of investigating the effects of ginger + zinc supplement on MS. The results showed that the ginger + zinc supplement could improve oxidative damage, inflammation, and autophagy caused by fructose and adjust the glucose and lipid metabolism and the homeostasis of zinc in rats with MS. The results of this study support the hypothesis that ginger can be used as a very suitable option for the production of medicinal supplements to maintain human health.
Collapse
Affiliation(s)
- Zana Ahmed Mustafa
- Department of Pharmacy, Medical Technical Institute, Erbil Polytechnic University, Erbil, Iraq
| | - Rojgar Hamed Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Dler Rostum Ali
- Basic Science Department, College of Medicine, Hawler Medical University, Erbil, Iraq
| | - Rahim Abdulkarimi
- Independent Scholar, Department of Environment, Boukan, Iran
- Department of Animal Science, Faculty of Agriculture, University of Zanjan, Zanjan, Iran
| | - Nashwan K Abdulkareem
- Biophysics Unit, Department of Basic Science, College of Medicine, Hawler Medical University, Erbil, Iraq
| | - Abolfazl Akbari
- Department of Physiology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
17
|
Stolzenbach F, Valdivia S, Ojeda-Provoste P, Toledo F, Sobrevia L, Kerr B. DNA methylation changes in genes coding for leptin and insulin receptors during metabolic-altered pregnancies. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165465. [DOI: 10.1016/j.bbadis.2019.05.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 03/19/2019] [Accepted: 05/02/2019] [Indexed: 01/07/2023]
|
18
|
Skinner JA, Garg ML, Dayas CV, Burrows TL. Is weight status associated with peripheral levels of oxytocin? A pilot study in healthy women. Physiol Behav 2019; 212:112684. [PMID: 31629767 DOI: 10.1016/j.physbeh.2019.112684] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 09/09/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
The neuropeptide oxytocin is best known for its role during parturition and the milk-let down reflex. Recent evidence identifies a role for oxytocin in eating behaviour. After oxytocin administration, caloric intake is reduced with stronger inhibitory effects in individuals with obesity. Whether the experience of visual food cues affects secretion or circulating levels of oxytocin is unknown. This pilot study had three aims: 1) to measure fasting appetite hormones with a focus on plasma oxytocin concentrations; 2) determine whether healthy vs. hyperpalatable visual food cues differentially altered plasma oxytocin; and 3) assess whether appetite hormone responses to healthy vs. hyperpalatable food images depended on weight or food addiction status. Eighteen healthy women of varying weight status, with/without self-reported food addiction were recruited. Study participants completed a set of standardised questionnaires, including Yale Food Addiction Scale, and attended a one-off experimental session. Blood was collected before and after viewing two sets of food images (healthy and hyperpalatable foods). Participants were randomly allocated in a crossover design to view either healthy images or hyperpalatable foods first. A positive correlation between BMI and plasma oxytocin was found (r2 = 0.32, p = 0.021) at baseline. Oxytocin levels were higher, and cholecystokinin levels lower, in food addicted (n = 6) vs. non-food addicted females (p = 0.015 and p<0.001, respectively). There were no significant changes (p>0.05) in plasma oxytocin levels in response to either healthy or hyperpalatable food images. Given that endogenous oxytocin administration tends to suppress eating behaviour; these data indicate that oxytocin receptor desensitization or oxytocin resistance may be important factors in the pathogenesis of obesity and food addiction. However, further studies in larger samples are needed to determine if peripheral oxytocin is responsive to visual food cues.
Collapse
Affiliation(s)
- Janelle A Skinner
- Nutrition and Dietetics, School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan NSW 2308, Australia; Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan NSW 2308, Australia
| | - Manohar L Garg
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan NSW 2308, Australia
| | - Christopher V Dayas
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan NSW 2308, Australia
| | - Tracy L Burrows
- Nutrition and Dietetics, School of Health Sciences, Faculty of Health and Medicine, University of Newcastle, Callaghan NSW 2308, Australia; Priority Research Centre for Physical Activity and Nutrition, University of Newcastle, Callaghan NSW 2308, Australia.
| |
Collapse
|
19
|
Chaskiel L, Bristow AD, Bluthé RM, Dantzer R, Blomqvist A, Konsman JP. Interleukin-1 reduces food intake and body weight in rat by acting in the arcuate hypothalamus. Brain Behav Immun 2019; 81:560-573. [PMID: 31310797 DOI: 10.1016/j.bbi.2019.07.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 07/12/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022] Open
Abstract
A reduction in food intake is commonly observed after bacterial infection, a phenomenon that can be reproduced by peripheral administration of Gram-negative bacterial lipopolysaccharide (LPS) or interleukin-1beta (IL-1β), a pro-inflammatory cytokine released by LPS-activated macrophages. The arcuate nucleus of the hypothalamus (ARH) plays a major role in food intake regulation and expresses IL-1 type 1 receptor (IL-1R1) mRNA. In the present work, we tested the hypothesis that IL-1R1 expressing cells in the ARH mediate IL-1β and/or LPS-induced hypophagia in the rat. To do so, we developed an IL-1β-saporin conjugate, which eliminated IL-R1-expressing neurons in the hippocampus, and micro-injected it into the ARH prior to systemic IL-1β and LPS administration. ARH IL-1β-saporin injection resulted in loss of neuropeptide Y-containing cells and attenuated hypophagia and weight loss after intraperitoneal IL-1β, but not LPS, administration. In conclusion, the present study shows that ARH NPY-containing neurons express functional IL-1R1s that mediate peripheral IL-1β-, but not LPS-, induced hypophagia. Our present and previous findings indicate that the reduction of food intake after IL-1β and LPS are mediated by different neural pathways.
Collapse
Affiliation(s)
- Léa Chaskiel
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Adrian D Bristow
- National Institute for Biological Standards and Control, Blanche Lane, South Mimms, Potters Bar, Hertfordshire EN6 3QG, UK
| | - Rose-Marie Bluthé
- Psychoneuroimmunology, Nutrition and Genetics, UMR CNRS 5226-INRA 1286, University of Bordeaux, 33076 Bordeaux, France
| | - Robert Dantzer
- Department of Symptom Research, MD Anderson Cancer Center, The University of Texas, Houston, TX 770030, USA
| | - Anders Blomqvist
- Division of Neurobiology, Department of Clinical and Experimental Medicine, Faculty of Medicine and Health Sciences, Linköping University, S-581 85 Linköping, Sweden
| | - Jan Pieter Konsman
- UMR CNRS 5287 Aquitaine Institute for Integrative and Cognitive Neuroscience, University of Bordeaux, 33076 Bordeaux, France.
| |
Collapse
|
20
|
Gougoula C, Bielfeld AP, Pour SJ, Sager M, Krüssel JS, Benten WPM, Baston-Büst DM. Metabolic and behavioral parameters of mice with reduced expression of Syndecan-1. PLoS One 2019; 14:e0219604. [PMID: 31299063 PMCID: PMC6625734 DOI: 10.1371/journal.pone.0219604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/27/2019] [Indexed: 11/19/2022] Open
Abstract
Energy balance is essential for all species. Ligand-receptor interactions mediate processes that regulate body activities like reproduction and metabolism based on the energy status. Such receptors are the heparan sulfate proteoglycans and specifically the family of syndecans. Therefore we investigated the differences of metabolic parameters of heterozygous Syndecan 1 mice (Sdc1+/-) with reduced expression of Sdc1 and the corresponding wild type mice. Sdc1+/- mice have a reduced body weight although they show increased leptin and decreased corticosterone levels. Furthermore, their food and water intake is increased. This is accompanied with less adipose tissue, smaller adipocytes and thus an increased density of adipocytes. For the detailed analysis of the metabolism the automated PhenoMaster system has been used, which allowed continuous and undisturbed recording of food and water intake, energy expenditure and movement. The reason for the lower body weight was the higher energy expenditure of these animals compared to controls. Additionally, female Sdc1+/- mice showed an increased locomotor activity. Referring to organs, the intestine in Sdc1+/- mice was heavier and longer, but no differences at the cellular level could be observed. These findings were independent of normal mating or vice versa embryo transfers of Sdc1+/- and wild type embryos in recipient females of the other genotype. Herein we showed that the reduced expression of Sdc1 led to an altered metabolism on fetal as well as on maternal side, which may play a role in the growth restriction observed in human pregnancy pathologies and in mice lacking Sdc1.
Collapse
Affiliation(s)
- Christina Gougoula
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT) of the Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | | | - Sarah Jean Pour
- Düsseldorf University Hospital, Department of OB/GYN and REI (UniKiD), Düsseldorf, Germany
| | - Martin Sager
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT) of the Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - Jan-Steffen Krüssel
- Düsseldorf University Hospital, Department of OB/GYN and REI (UniKiD), Düsseldorf, Germany
| | - Wilhelm Peter M. Benten
- Central Unit for Animal Research and Animal Welfare Affairs (ZETT) of the Heinrich-Heine-University of Düsseldorf, Düsseldorf, Germany
| | - Dunja Maria Baston-Büst
- Düsseldorf University Hospital, Department of OB/GYN and REI (UniKiD), Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
21
|
Yang H, Liu CN, Wolf RM, Ralle M, Dev S, Pierson H, Askin F, Steele KE, Magnuson TH, Schweitzer MA, Wong GW, Lutsenko S. Obesity is associated with copper elevation in serum and tissues. Metallomics 2019; 11:1363-1371. [PMID: 31249997 DOI: 10.1039/c9mt00148d] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Copper misbalance has been linked to fat accumulation in animals and experimental systems; however, information about copper homeostasis in human obesity is limited. In this study, the copper status of obese individuals was evaluated by measuring their levels of copper and cuproproteins in serum, adipose and hepatic tissues. The analysis of serum trace elements showed significant positive and element-specific correlation between copper and BMI after controlling for gender, age, and ethnicity. Serum copper also positively correlated with leptin, insulin, and the leptin/BMI ratio. When compared to lean controls, obese patients had elevated circulating cuproproteins, such as semucarbazide-sensitive amine oxidase (SSAO) and ceruloplasmin, and higher SSAO activity and copper levels in visceral fat. Although hepatic steatosis reduces copper levels in the liver, obese patients with no or mild steatosis have higher copper content in the liver compared to lean controls. In conclusion, obese patients evaluated in this study had altered copper status. Strong positive correlations of copper levels with BMI and leptin suggest that copper and/or cuproproteins may be functionally linked to fat accumulation.
Collapse
Affiliation(s)
- Haojun Yang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Chin-Nung Liu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Risa M Wolf
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martina Ralle
- Department of Genetics, Oregon Health & Science University, Portland, Oregon, USA
| | - Som Dev
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Hannah Pierson
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Frederic Askin
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kimberley E Steele
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas H Magnuson
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Schweitzer
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - G William Wong
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. and Center for Metabolism and Obesity Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Svetlana Lutsenko
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
22
|
Menting MD, Mintjens S, van de Beek C, Frick CJ, Ozanne SE, Limpens J, Roseboom TJ, Hooijmans CR, van Deutekom AW, Painter RC. Maternal obesity in pregnancy impacts offspring cardiometabolic health: Systematic review and meta-analysis of animal studies. Obes Rev 2019; 20:675-685. [PMID: 30633422 PMCID: PMC6849816 DOI: 10.1111/obr.12817] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/19/2018] [Accepted: 11/06/2018] [Indexed: 12/24/2022]
Abstract
Obesity before and during pregnancy leads to reduced offspring cardiometabolic health. Here, we systematically reviewed animal experimental evidence of maternal obesity before and during pregnancy and offspring anthropometry and cardiometabolic health. We systematically searched Embase and Medline from inception until January 2018. Eligible publications compared offspring of mothers with obesity to mothers with a normal weight. We performed meta-analyses and subgroup analyses. We also examined methodological quality and publication bias. We screened 2543 publications and included 145 publications (N = 21 048 animals, five species). Essential methodological details were not reported in the majority of studies. We found evidence of publication bias for birth weight. Offspring of mothers with obesity had higher body weight (standardized mean difference (SMD) 0.76 [95% CI 0.60;0.93]), fat percentage (0.99 [0.64;1.35]), systolic blood pressure (1.33 [0.75;1.91]), triglycerides (0.64 [0.42;0.86], total cholesterol (0.46 [0.18;0.73]), glucose level (0.43 [0.24;0.63]), and insulin level (0.81 [0.61;1.02]) than offspring of control mothers, but similar birth weight. Sex, age, or species did not influence the effect of maternal obesity on offspring's cardiometabolic health. Obesity before and during pregnancy reduces offspring cardiometabolic health in animals. Future intervention studies should investigate whether reducing obesity prior to conception could prevent these detrimental programming effects and improve cardiometabolic health of future generations.
Collapse
Affiliation(s)
- M D Menting
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - S Mintjens
- Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pediatrics, Department of Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam, The Netherlands
| | - C van de Beek
- Department of Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - C J Frick
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - S E Ozanne
- MRC Metabolic Diseases Unit and Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - J Limpens
- Department of Research Support-Medical Library, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - T J Roseboom
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Obstetrics and Gynecology, Amsterdam Public Health Research Institute, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - C R Hooijmans
- Department for Health Evidence Unit SYRCLE, Department of Anesthesiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - A W van Deutekom
- Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Pediatric Cardiology, Amsterdam, The Netherlands
| | - R C Painter
- Department of Obstetrics and Gynecology, Amsterdam Reproduction and Development Research Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Chapelot D, Charlot K. Physiology of energy homeostasis: Models, actors, challenges and the glucoadipostatic loop. Metabolism 2019; 92:11-25. [PMID: 30500561 DOI: 10.1016/j.metabol.2018.11.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/25/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022]
Abstract
The aim of this review is to discuss the physiology of energy homeostasis (EH), which is a debated concept. Thus, we will see that the set-point theory is highly challenged and that other models integrating an anticipative component, such as energy allostasis, seem more relevant to experimental reports and life preservation. Moreover, the current obesity epidemic suggests that EH is poorly efficient in the modern human dietary environment. Non-homeostatic phenomena linked to hedonism and reward seem to profoundly impair EH. In this review, the apparent failed homeostatic responses to energy challenges such as exercise, cafeteria diet, overfeeding and diet-induced weight loss, as well as their putative determinants, are analyzed to highlight the mechanisms of EH. Then, the hormonal, neuronal, and metabolic factors of energy intake or energy expenditure are briefly presented. Last, this review focuses on the contributions of two of the most pivotal and often overlooked determinants of EH: the availability of endogenous energy and the pattern of energy intake. A glucoadipostatic loop model is finally proposed to link energy stored in adipose tissue to EH through changes in eating behavior via leptin and sympathetic nervous system activity.
Collapse
Affiliation(s)
- Didier Chapelot
- Université Paris 13, Centre de Recherche en Epidémiologie et Statistique, Equipe de Recherche en Epidémiologie Nutritionnelle (EREN), Inserm (U1153), Inra (U1125), Cnam, Bobigny, France.
| | - Keyne Charlot
- Institut de Recherche Biomédicale des Armées, Unité de Physiologie des Exercices et Activités en Conditions Extrêmes, Département Environnements Opérationnels, Brétigny-sur-Orge, France
| |
Collapse
|
24
|
Habeeballah H, Alsuhaymi N, Stebbing MJ, Badoer E. Central Administration of Insulin Combined With Resistin Reduces Renal Sympathetic Nerve Activity in Rats Fed a High Fat Diet. Front Physiol 2019; 10:93. [PMID: 30804811 PMCID: PMC6378567 DOI: 10.3389/fphys.2019.00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/25/2019] [Indexed: 11/13/2022] Open
Abstract
Insulin receptors are widely distributed in the central nervous system and their activation by insulin elicits renal sympatho-excitatory effects. Resistin, an adipokine, promotes resistance to the metabolic effects of insulin. Resistin also induces increases in renal sympathetic nerve activity (RSNA) by acting in the brain, but whether it can influence insulin’s actions on RSNA is unknown. In the present study we investigated, in male Sprague-Dawley rats (7–8 weeks of age), the effects of central administration of insulin combined with resistin on RSNA following a normal diet (ND) and a high fat diet (HFD) (22% fat), since HFD can reportedly attenuate insulin’s actions. RSNA, mean arterial pressure (MAP) and heart rate (HR) responses were monitored and recorded before and for 180 min after intracerebroventricular injection of saline (control) (n = 5 HFD and ND), resistin (7 μg; n = 4 ND, n = 5 HFD), insulin (500 mU; n = 6 ND, n = 5 HFD), and the combination of both resistin and insulin (n = 7 ND, n = 5 HFD). The key finding of the present study was that when resistin and insulin were combined there was no increase in RSNA induced in rats fed a normal diet or the high fat diet. This contrasted with the sympatho-excitatory RSNA effects of the hormones when each was administered alone in rats fed the ND and the HFD.
Collapse
|
25
|
Yang S, Li S, Li XJ. MANF: A New Player in the Control of Energy Homeostasis, and Beyond. Front Physiol 2018; 9:1725. [PMID: 30555354 PMCID: PMC6282101 DOI: 10.3389/fphys.2018.01725] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/15/2018] [Indexed: 01/06/2023] Open
Abstract
All human behaviors, including the control of energy homeostasis, are ultimately mediated by neuronal activities in the brain. Neurotrophic factors represent a protein family that plays important roles in regulating neuronal development, function, and survival. It has been well established that canonical neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and ciliary neurotrophic factor (CNTF), play important roles in the central regulation of energy homeostasis. Recently, a class of non-canonical neurotrophic factors, represented by mesencephalic astrocyte-derived neurotrophic factor (MANF), has been discovered. MANF is structurally and functionally distinct from those canonical neurotrophic factors, hence raising the issue of MANF being non-canonical. Nonetheless, emerging evidence suggests that MANF is critically involved in many neuronal activities. Here, we review our current understanding about the functions of MANF in the brain, with a primary focus on the control of energy homeostasis.
Collapse
Affiliation(s)
- Su Yang
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| | - Shihua Li
- GHM Institute of CNS Regeneration, Jinan University Guangzhou, China
| | - Xiao-Jiang Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
26
|
Abstract
The hypothalamus is the brain region responsible for the maintenance of energetic homeostasis. The regulation of this process arises from the ability of the hypothalamus to orchestrate complex physiological responses such as food intake and energy expenditure, circadian rhythm, stress response, and fertility. Metabolic alterations such as obesity can compromise these hypothalamic regulatory functions. Alterations in circadian rhythm, stress response, and fertility further contribute to aggravate the metabolic dysfunction of obesity and contribute to the development of chronic disorders such as depression and infertility.At cellular level, obesity caused by overnutrition can damage the hypothalamus promoting inflammation and impairing hypothalamic neurogenesis. Furthermore, hypothalamic neurons suffer apoptosis and impairment in synaptic plasticity that can compromise the proper functioning of the hypothalamus. Several factors contribute to these phenomena such as ER stress, oxidative stress, and impairments in autophagy. All these observations occur at the same time and it is still difficult to discern whether inflammatory processes are the main drivers of these cellular dysfunctions or if the hypothalamic hormone resistance (insulin, leptin, and ghrelin) can be pinpointed as the source of several of these events.Understanding the mechanisms that underlie the pathophysiology of obesity in the hypothalamus is crucial for the development of strategies that can prevent or attenuate the deleterious effects of obesity.
Collapse
|
27
|
Cuvelier E, Méquinion M, Leghay C, Sibran W, Stievenard A, Sarchione A, Bonte MA, Vanbesien-Mailliot C, Viltart O, Saitoski K, Caron E, Labarthe A, Comptdaer T, Semaille P, Carrié H, Mutez E, Gressier B, Destée A, Chartier-Harlin MC, Belarbi K. Overexpression of Wild-Type Human Alpha-Synuclein Causes Metabolism Abnormalities in Thy1-aSYN Transgenic Mice. Front Mol Neurosci 2018; 11:321. [PMID: 30333721 PMCID: PMC6176013 DOI: 10.3389/fnmol.2018.00321] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/17/2018] [Indexed: 12/20/2022] Open
Abstract
Parkinson’s disease is a progressive neurodegenerative disorder characterized by loss of dopaminergic neurons, pathological accumulation of alpha-synuclein and motor symptoms, but also by non-motor symptoms. Metabolic abnormalities including body weight loss have been reported in patients and could precede by several years the emergence of classical motor manifestations. However, our understanding of the pathophysiological mechanisms underlying body weight loss in PD is limited. The present study investigated the links between alpha-synuclein accumulation and energy metabolism in transgenic mice overexpressing Human wild-type (WT) alpha-synuclein under the Thy1 promoter (Thy1-aSYN mice). Results showed that Thy1-aSYN mice gained less body weight throughout life than WT mice, with significant difference observed from 3 months of age. Body composition analysis of 6-month-old transgenic animals showed that body mass loss was due to lower adiposity. Thy1-aSYN mice displayed lower food consumption, increased spontaneous activity, as well as a reduced energy expenditure compared to control mice. While no significant change in glucose or insulin responses were observed, Thy1-aSYN mice had significantly lower plasmatic levels of insulin and leptin than control animals. Moreover, the pathological accumulation of alpha-synuclein in the hypothalamus of 6-month-old Thy1-aSYN mice was associated with a down-regulation of the phosphorylated active form of the signal transducer and activator of transcription 3 (STAT3) and of Rictor (the mTORC2 signaling pathway), known to couple hormonal signals with the maintenance of metabolic and energy homeostasis. Collectively, our results suggest that (i) metabolic alterations are an important phenotype of alpha-synuclein overexpression in mice and that (ii) impaired STAT3 activation and mTORC2 levels in the hypothalamus may underlie the disruption of feeding regulation and energy metabolism in Thy1-aSYN mice.
Collapse
Affiliation(s)
- Elodie Cuvelier
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Mathieu Méquinion
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Coline Leghay
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - William Sibran
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Aliçia Stievenard
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Alessia Sarchione
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Marie-Amandine Bonte
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Christel Vanbesien-Mailliot
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Odile Viltart
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Kevin Saitoski
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Emilie Caron
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Alexandra Labarthe
- UMR 894, Centre de Psychiatrie et Neurosciences, Inserm, Université Paris Descartes, Paris, France
| | - Thomas Comptdaer
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Pierre Semaille
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Hélène Carrié
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Eugénie Mutez
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Bernard Gressier
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Alain Destée
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Marie-Christine Chartier-Harlin
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| | - Karim Belarbi
- UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer, Inserm, Centre Hospitalier Régional Universitaire de Lille, Université de Lille, Lille, France
| |
Collapse
|
28
|
Patel JC, Stouffer MA, Mancini M, Nicholson C, Carr KD, Rice ME. Interactions between insulin and diet on striatal dopamine uptake kinetics in rodent brain slices. Eur J Neurosci 2018; 49:794-804. [PMID: 29791756 DOI: 10.1111/ejn.13958] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/21/2018] [Accepted: 04/25/2018] [Indexed: 12/28/2022]
Abstract
Diet influences dopamine transmission in motor- and reward-related basal ganglia circuitry. In part, this reflects diet-dependent regulation of circulating and brain insulin levels. Activation of striatal insulin receptors amplifies axonal dopamine release in brain slices, and regulates food preference in vivo. The effect of insulin on dopamine release is indirect, and requires striatal cholinergic interneurons that express insulin receptors. However, insulin also acts directly on dopamine axons to increase dopamine uptake by promoting dopamine transporter (DAT) surface expression, counteracting enhanced dopamine release. Here, we determined the functional consequences of acute insulin exposure and chronic diet-induced changes in insulin on DAT activity after evoked dopamine release in striatal slices from adult ad-libitum fed (AL) rats and mice, and food-restricted (FR) or high-fat/high-sugar obesogenic (OB) diet rats. Uptake kinetics were assessed by fitting evoked dopamine transients to the Michaelis-Menten equation and extracting Cpeak and Vmax . Insulin (30 nm) increased both parameters in the caudate putamen and nucleus accumbens core of AL rats in an insulin receptor- and PI3-kinase-dependent manner. A pure effect of insulin on uptake was unmasked using mice lacking striatal acetylcholine, in which increased Vmax caused a decrease in Cpeak . Diet also influenced Vmax , which was lower in FR vs. AL. The effects of insulin on Cpeak and Vmax were amplified by FR but blunted by OB, consistent with opposite consequences of these diets on insulin levels and insulin receptor sensitivity. Overall, these data reveal acute and chronic effects of insulin and diet on dopamine release and uptake that will influence brain reward pathways.
Collapse
Affiliation(s)
- Jyoti C Patel
- Department of Neurosurgery, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Melissa A Stouffer
- Department of Neurosurgery, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA.,Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA
| | - Maria Mancini
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.,NYU Marlene and Paolo Fresco Institute on Parkinson's Disease and Movement Disorders, New York University School of Medicine, New York, NY, USA
| | - Charles Nicholson
- Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.,NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| | - Kenneth D Carr
- NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA.,Psychiatry, New York University School of Medicine, New York, NY, USA.,Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, USA
| | - Margaret E Rice
- Department of Neurosurgery, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA.,Department of Neuroscience and Physiology, New York University School of Medicine, New York, NY, USA.,NYU Marlene and Paolo Fresco Institute on Parkinson's Disease and Movement Disorders, New York University School of Medicine, New York, NY, USA.,NYU Neuroscience Institute, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
29
|
Street ME, Angelini S, Bernasconi S, Burgio E, Cassio A, Catellani C, Cirillo F, Deodati A, Fabbrizi E, Fanos V, Gargano G, Grossi E, Iughetti L, Lazzeroni P, Mantovani A, Migliore L, Palanza P, Panzica G, Papini AM, Parmigiani S, Predieri B, Sartori C, Tridenti G, Amarri S. Current Knowledge on Endocrine Disrupting Chemicals (EDCs) from Animal Biology to Humans, from Pregnancy to Adulthood: Highlights from a National Italian Meeting. Int J Mol Sci 2018; 19:1647. [PMID: 29865233 PMCID: PMC6032228 DOI: 10.3390/ijms19061647] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/23/2018] [Accepted: 05/31/2018] [Indexed: 02/07/2023] Open
Abstract
Wildlife has often presented and suggested the effects of endocrine disrupting chemicals (EDCs). Animal studies have given us an important opportunity to understand the mechanisms of action of many chemicals on the endocrine system and on neurodevelopment and behaviour, and to evaluate the effects of doses, time and duration of exposure. Although results are sometimes conflicting because of confounding factors, epidemiological studies in humans suggest effects of EDCs on prenatal growth, thyroid function, glucose metabolism and obesity, puberty, fertility, and on carcinogenesis mainly through epigenetic mechanisms. This manuscript reviews the reports of a multidisciplinary national meeting on this topic.
Collapse
Affiliation(s)
- Maria Elisabeth Street
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Sabrina Angelini
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Sergio Bernasconi
- Former Department of Medicine, University of Parma, Via A. Catalani 10, 43123 Parma, Italy.
| | - Ernesto Burgio
- ECERI European Cancer and Environment Research Institute, Square de Meeus, 38-40, 1000 Bruxelles, Belgium.
| | - Alessandra Cassio
- Pediatric Endocrinology Programme, Pediatrics Unit, Department of Woman, Child Health and Urologic Diseases, AOU S. Orsola-Malpighi, Via Massarenti, 11, 40138 Bologna, Italy.
| | - Cecilia Catellani
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Francesca Cirillo
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Annalisa Deodati
- Department of Pediatrics (DPUO), Bambino Gesù Children's Hospital, Tor Vergata University, Piazza S. Onofrio 4, 00165 Rome, Italy.
| | - Enrica Fabbrizi
- Department of Pediatrics and Neonatology, Augusto Murri Hospital, Via Augusto Murri, 17, 63900 Fermo, Itlay.
| | - Vassilios Fanos
- Neonatal Intensive Care Unit, Neonatal Pathology and Neonatal Section, AOU and University of Cagliari, via Ospedale, 54, 09124 Cagliari, Italy.
| | - Giancarlo Gargano
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Enzo Grossi
- Villa Santa Maria Institute, Neuropsychiatric Rehabilitation Center, Via IV Novembre 15, 22038 Tavernerio (Como), Italy.
| | - Lorenzo Iughetti
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, Pediatrics Unit, University of Modena and Reggio Emilia, via del Pozzo, 71, 41124 Modena, Italy.
| | - Pietro Lazzeroni
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Alberto Mantovani
- Department of Veterinary Public Health and Food Safety, Food and Veterinary Toxicology Unit ISS⁻National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Lucia Migliore
- Department of Traslational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55, 56123 Pisa, Italy.
| | - Paola Palanza
- Unit of Neuroscience, Department of Medicine and Surgery, University of Parma, Via Gramsci, 14, 43126 Parma, Italy.
| | - Giancarlo Panzica
- Laboratory of Neuroendocrinology, Department of Neuroscience Rita Levi Montalcini, University of Turin, Via Cherasco 15, 10126 Turin, Italy.
- Neuroscience Institute Cavalieri-Ottolenghi (NICO), Regione Gonzole, 10, 10043 Orbassano (Turin), Italy.
| | - Anna Maria Papini
- Department of Chemistry 'Ugo Schiff', University of Florence, Via della Lastruccia, 3-13, 50019 Sesto Fiorentino, Florence, Italy.
| | - Stefano Parmigiani
- Unit of Evolutionary and Functional Biology-Department of Chemistry, Life Sciences and Environmental Sustainability (SCVSA)-University of Parma⁻11/a, 43124 Parma, Italy.
| | - Barbara Predieri
- Department of Medical and Surgical Sciences of the Mother, Children and Adults, Pediatrics Unit, University of Modena and Reggio Emilia, via del Pozzo, 71, 41124 Modena, Italy.
| | - Chiara Sartori
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Gabriele Tridenti
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| | - Sergio Amarri
- Department of Obstetrics, Gynaecology and Paediatrics, Azienda USL-IRCCS, Viale Risorgimento 80, 42123 Reggio Emilia, Italy.
| |
Collapse
|
30
|
Wolf T, Tsenkova V, Ryff CD, Davidson RJ, Willette AA. Neural, Hormonal, and Cognitive Correlates of Metabolic Dysfunction and Emotional Reactivity. Psychosom Med 2018; 80:452-459. [PMID: 29595709 PMCID: PMC5976543 DOI: 10.1097/psy.0000000000000582] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
OBJECTIVE Prediabetes and type 2 diabetes (i.e., hyperglycemia) are characterized by insulin resistance. These problems with energy metabolism may exacerbate emotional reactivity to negatively valenced stimuli and related phenomena such as predisposition toward negative affect, as well as cognitive deficits. Higher emotional reactivity is seen with hyperglycemia and insulin resistance. However, it is largely unknown how metabolic dysfunction correlates with related neural, hormonal, and cognitive outcomes. METHODS Among 331 adults from the Midlife in the United States study, eye-blink response (EBR) we cross sectionally examined to gauge reactivity to negative, positive, or neutrally valenced pictures from international affect picture system stimuli proximal to an acoustic startle probe. Increased EBR to negative stimuli was considered an index of stress reactivity. Frontal alpha asymmetry, a biomarker of negative affect predisposition, was determined using resting electroencephalography. Baseline urinary cortisol output was collected. Cognitive performance was gauged using the Brief Test of Adult Cognition by telephone. Fasting glucose and insulin characterized hyperglycemia or the homeostatic model assessment of insulin resistance. RESULTS Higher homeostatic model assessment of insulin resistance corresponded to an increased startle response, measured by EBR magnitude, for negative versus positive stimuli (R = 0.218, F(1,457) = 5.48, p = .020, euglycemia: M(SD) = .092(.776), hyperglycemia: M(SD) = .120(.881)). Participants with hyperglycemia versus euglycemia showed greater right frontal alpha asymmetry (F(1,307) = 6.62, p = .011, euglycemia: M(SD) = .018(.167), hyperglycemia: M(SD) = -.029(.160)), and worse Brief Test of Adult Cognition by telephone arithmetic performance (F(1,284) = 4.25, p = .040, euglycemia: M(SD) = 2.390(1.526), hyperglycemia: M(SD) = 1.920(1.462)). Baseline urinary cortisol (log10 μg/12 hours) was also dysregulated in individuals with hyperglycemia (F(1,324) = 5.09, p = .025, euglycemia: M(SD) = 1.052 ± .332, hyperglycemia: M(SD) = .961 (.362)). CONCLUSIONS These results suggest that dysmetabolism is associated with increased emotional reactivity, predisposition toward negative affect, and specific cognitive deficits.
Collapse
Affiliation(s)
- Tovah Wolf
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
| | - Vera Tsenkova
- Institute on Aging, University of Wisconsin-Madison, Madison, WI, United States
| | - Carol D. Ryff
- Institute on Aging, University of Wisconsin-Madison, Madison, WI, United States
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, United States
| | - Richard J. Davidson
- Institute on Aging, University of Wisconsin-Madison, Madison, WI, United States
- Department of Psychology, University of Wisconsin-Madison, Madison, WI, United States
- Center for Healthy Minds, University of Wisconsin-Madison, Madison, WI, United States
- Waisman Laboratory for Brain Imaging and Behavior, University of Wisconsin-Madison, Madison, WI, United States
| | - Auriel A. Willette
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA, United States
- Department of Psychology, Iowa State University, Ames, IA, United States
- Department of Biomedical Sciences, Iowa State University, Ames, IA, United States
- Department of Neurology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
31
|
Chaiyasut C, Woraharn S, Sivamaruthi BS, Lailerd N, Kesika P, Peerajan S. Lactobacillus fermentum HP3-Mediated Fermented Hericium erinaceus Juice as a Health Promoting Food Supplement to Manage Diabetes Mellitus. J Evid Based Integr Med 2018; 23:2515690X18765699. [PMID: 29619846 PMCID: PMC5894895 DOI: 10.1177/2515690x18765699] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The current study investigated the antidiabetic property of Lactobacillus fermentum HP3–mediated fermented Hericium erinaceus juice (FHJ) using male Wistar rats with streptozotocin-induced diabetes mellitus (DM). FHJ was prepared using boiled mushroom juice and L. fermentum HP3. Amino acid and γ-aminobutyric acid (GABA) content of FHJ was analyzed. Streptozotocin-induced DM rats were supplemented with FHJ in a pre- and posttreatment method. The changes in plasma insulin, plasma glucose level, glycated hemoglobin (HbA1c), representative cytokines, and the antioxidant system were assessed in experimental rats using spectrophotometric methods and enzyme-linked immunosorbent assay. The supplementation of FHJ improved the body mass, insulin level, and recovery progress of hyperglycemia. HbA1c level was altered by the FHJ intervention. The inflammatory cytokines level was suppressed in FHJ supplemented group compared with control. Intervention of FHJ and insulin improved the production of interleukin-10 and transforming growth factor-–β1 in DM rat. The study suggested that fermented H erinaceus juice may be used as one of the food-based health-promoting supplement to manage DM along with medication.
Collapse
Affiliation(s)
- Chaiyavat Chaiyasut
- 1 Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Sasimar Woraharn
- 1 Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand.,2 Mahidol University Nakhon Sawan Campus, Nakhon Sawan, Thailand
| | - Bhagavathi Sundaram Sivamaruthi
- 1 Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Narissara Lailerd
- 3 Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Periyanaina Kesika
- 1 Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | | |
Collapse
|
32
|
Functional Interrogation of the AgRP Neural Circuits in Control of Appetite, Body Weight, and Behaviors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:1-16. [PMID: 30390282 DOI: 10.1007/978-981-13-1286-1_1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurons expressing agouti-related protein (AgRP), the so-called hunger neurons, protect mammals from starvation by promoting food-seeking behaviors (Trends Neurosci 36:504-512, 2013). Now an increasing amount of evidence show that these hunger-sensing neurons not only motivate animals to forage and ingest food but also help conserve energy by inhibiting innate processes that demand large amounts of energy such as growth, reproduction, and stress response. It has further been perceived that AgRP neurons transmit signals with negative valence to reward and cognitive centers so as to engage the motivational behavior toward seeking and obtaining foods (Physiol Behav 190:34-42, 2017). Recent advancement in genome editing and neurotechniques unleashed an escalated research of uniquely defined neuronal populations and neural circuits underlying the behavioral regulation of body weight and food responses (Nat Biotechnol 32:347-355, 2014; Proc Natl Acad Sci 113, 2016). In this chapter we will review literatures describing the functional organization of the AgRP circuit and its correlative signaling components that influence ingestive, foraging, motivational, and cognitive responses, a framework that reshaped our thinking toward the new hope and challenges in treatment of obesity and eating disorders.
Collapse
|
33
|
Shakya M, Briski KP. Rebound Feeding in the Wake of Short-Term Suspension of Food Intake Differs in the Presence of Estrous Cycle Peak versus Nadir Levels of Estradiol. Endocrinol Metab (Seoul) 2017; 32:475-484. [PMID: 29271620 PMCID: PMC5744734 DOI: 10.3803/enm.2017.32.4.475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/10/2017] [Accepted: 09/18/2017] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Short-term interruption of feeding is ordinary in modern life but negatively impacts appetite control and body weight. Estradiol (E) imposes long-term inhibitory tonus on food consumption; however, E influence on energy repletion secondary to food deprivation (FD) is unclear. This study investigated the hypothesis that E signal strength regulates hyperphagic responses to FD of varying duration. METHODS Ovariectomized female rats were implanted with E-containing silastic capsules (30 [E-30] or 300 μg [E-300]/mL) to replicate plasma concentrations at cycle nadir versus peak levels. RESULTS Data show that food intake was increased equally in E-30 and E-300 rats after 12 hours of food deprivation (FD-12); yet, FD of 18 hours (FD-18) amplified refeeding by E-300 versus E-30. Caudal fourth ventricular administration of the 5'-monophosphate-activated protein kinase (AMPK) inhibitor compound C (Cc) did not modify FD-induced hyperphagia in E-30 (regardless of FD interval) or E-300 animals exposed to FD-12, but diminished refeeding after FD-18 in E-300 rats. Cc-reversible hyperglycemia occurred in refed FD-18 groups. Serum insulin was resistant to FD-12 plus refeeding, but was elevated by AMPK-dependent mechanisms in refed E-300 FD-18 rats; equivalent Cc-insensitive decrements in circulating leptin occurred in all FD groups. CONCLUSION Current results show that estrous cycle peak, but not baseline, E levels engage hindbrain AMPK signaling to intensify hyperphagia in response to prolongation of FD. Observations of hindbrain AMPK-dependent hyperglycemia, alongside elevated insulin secretion, in refed rats exposed to FD-18 implicate this sensor in insulin resistance mechanisms of glucose partitioning in response to this metabolic imbalance.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, College of Health and Pharmaceutical Sciences, The University of Louisiana Monroe, Monroe, LA, USA
| | - Karen P Briski
- Department of Basic Pharmaceutical Sciences, School of Pharmacy, College of Health and Pharmaceutical Sciences, The University of Louisiana Monroe, Monroe, LA, USA.
| |
Collapse
|
34
|
Mitchell SE, Delville C, Konstantopedos P, Derous D, Green CL, Wang Y, Han JDJ, Promislow DEL, Douglas A, Chen L, Lusseau D, Speakman JR. The effects of graded levels of calorie restriction: V. Impact of short term calorie and protein restriction on physical activity in the C57BL/6 mouse. Oncotarget 2017; 7:19147-70. [PMID: 27007156 PMCID: PMC4991372 DOI: 10.18632/oncotarget.8158] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/28/2016] [Indexed: 12/15/2022] Open
Abstract
Calorie restriction (CR) delays the onset of age-related disease and extends lifespan in a number of species. When faced with reduced energy supply animals need to lower energy demands, which may be achieved in part by reducing physical activity (PA). We monitored changes in PA using implanted transmitters in male C57BL/6 mice in response to graded levels of CR (10 to 40%) or matched levels of graded protein restriction (PR) for 3 months. Mice were fed at lights out and ad libitum controls were limited to dark-phase feeding (12AL) or 24hr/day. Total daily PA declined in a non-linear manner over the first 30 days of CR or PR, remaining stable thereafter. Total daily PA was not related to the level of CR or PR. Total daily PA over the last 20 days of restriction was related to circulating leptin, insulin, tumor necrosis factor-α (TNF-α) and insulin-like growth factor (IGF)-1 levels, measured after 3 months. Mice under restriction showed a high level of activity in the 2hrs before feeding (food anticipatory activity: FAA). FAA followed a complex pattern, peaking around day 20, falling on ∼day 37 then increasing again. FAA was also positively related to the level of restriction and inversely to leptin, insulin, TNF-α and IGF-1. Non-FAA, in contrast, declined over the period of restriction, generally more so in mice under greater restriction, thereby offsetting to some extent the increase in FAA. Mice under PR displayed no changes in PA over time or in comparison to 12AL, and showed no increase in FAA.
Collapse
Affiliation(s)
- Sharon E Mitchell
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Camille Delville
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Penelope Konstantopedos
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Davina Derous
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Cara L Green
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Daniel E L Promislow
- Department of Pathology and Department of Biology, University of Washington, Seattle, Washington, USA
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Luonan Chen
- Key Laboratory of Systems Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - David Lusseau
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - John R Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| |
Collapse
|
35
|
MANF regulates hypothalamic control of food intake and body weight. Nat Commun 2017; 8:579. [PMID: 28924165 PMCID: PMC5603516 DOI: 10.1038/s41467-017-00750-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 07/25/2017] [Indexed: 12/30/2022] Open
Abstract
The hypothalamus has a vital role in controlling food intake and energy homeostasis; its activity is modulated by neuropeptides and endocrine factors. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a neurotrophic factor that is also localized in the endoplasmic reticulum (ER) in neurons. Here we show that MANF is highly enriched in distinct nuclei of the mouse hypothalamus, and that MANF expression in the hypothalamus is upregulated in response to fasting. Increasing or decreasing hypothalamic MANF protein levels causes hyperphagia or hypophagia, respectively. Moreover, MANF triggers hypothalamic insulin resistance by enhancing the ER localization and activity of PIP4k2b, a kinase known to regulate insulin signaling. Our findings indicate that MANF influences food intake and body weight by modulating hypothalamic insulin signaling.MANF is a neurotrophic factor that is secreted but also mediates the unfolded protein response acting intracellularly. Here, the authors show that MANF expression in the brain is influenced by nutritional cues, and hypothalamic MANF influences food intake and systemic energy homeostasis.
Collapse
|
36
|
Guarino D, Nannipieri M, Iervasi G, Taddei S, Bruno RM. The Role of the Autonomic Nervous System in the Pathophysiology of Obesity. Front Physiol 2017; 8:665. [PMID: 28966594 PMCID: PMC5606212 DOI: 10.3389/fphys.2017.00665] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/22/2017] [Indexed: 12/18/2022] Open
Abstract
Obesity is reaching epidemic proportions globally and represents a major cause of comorbidities, mostly related to cardiovascular disease. The autonomic nervous system (ANS) dysfunction has a two-way relationship with obesity. Indeed, alterations of the ANS might be involved in the pathogenesis of obesity, acting on different pathways. On the other hand, the excess weight induces ANS dysfunction, which may be involved in the haemodynamic and metabolic alterations that increase the cardiovascular risk of obese individuals, i.e., hypertension, insulin resistance and dyslipidemia. This article will review current evidence about the role of the ANS in short-term and long-term regulation of energy homeostasis. Furthermore, an increased sympathetic activity has been demonstrated in obese patients, particularly in the muscle vasculature and in the kidneys, possibily contributing to increased cardiovascular risk. Selective leptin resistance, obstructive sleep apnea syndrome, hyperinsulinemia and low ghrelin levels are possible mechanisms underlying sympathetic activation in obesity. Weight loss is able to reverse metabolic and autonomic alterations associated with obesity. Given the crucial role of autonomic dysfunction in the pathophysiology of obesity and its cardiovascular complications, vagal nerve modulation and sympathetic inhibition may serve as therapeutic targets in this condition.
Collapse
Affiliation(s)
- Daniela Guarino
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy.,Institute of Clinical Physiology of CNRPisa, Italy.,Scuola Superiore Sant'AnnaPisa, Italy
| | - Monica Nannipieri
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | | | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Rosa Maria Bruno
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
37
|
Bruce EB, de Kloet AD. The intricacies of the renin-angiotensin-system in metabolic regulation. Physiol Behav 2017; 178:157-165. [PMID: 27887998 PMCID: PMC5600901 DOI: 10.1016/j.physbeh.2016.11.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 12/15/2022]
Abstract
Over recent years, the renin-angiotensin-system (RAS), which is best-known as an endocrine system with established roles in hydromineral balance and blood pressure control, has emerged as a fundamental regulator of many additional physiological and pathophysiological processes. In this manuscript, we celebrate and honor Randall Sakai's commitment to his trainees, as well as his contribution to science. Scientifically, Randall made many notable contributions to the recognition of the RAS's roles in brain and behavior. His interests, in this regard, ranged from its traditionally-accepted roles in hydromineral balance, to its less-appreciated functions in stress responses and energy metabolism. Here we review the current understanding of the role of the RAS in the regulation of metabolism. In particular, the opposing actions of the RAS within adipose tissue vs. its actions within the brain are discussed.
Collapse
Affiliation(s)
- Erin B Bruce
- Department of Pharmacodynamics, College of Pharmacy, University of Florida, United States
| | - Annette D de Kloet
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, United States.
| |
Collapse
|
38
|
Persaud N, Ziai H, Lebovic G, Maguire JL, Khovratovich M, Simpson JAR, Adeli K, Hamilton J, McCrindle BW, Parkin PC, Birken CS. Parent reported nutritional risk and laboratory indices of cardiometabolic risk and in preschool-aged children. J Pediatr Endocrinol Metab 2017; 30:839-846. [PMID: 28723611 DOI: 10.1515/jpem-2016-0328] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/12/2017] [Indexed: 11/15/2022]
Abstract
BACKGROUND Eating habits formed during childhood may contribute to the increasing prevalence of cardiometabolic disorders. Assessing nutritional risk in young children may help to prevent later cardiometabolic disease. The objective of this study was to determine whether parent-reported nutritional risk in preschool-aged children was associated with laboratory indices of cardiometabolic risk, namely leptin and insulin. METHODS In this cross-sectional study, the relationship between nutritional risk as determined by the parent-completed NutriSTEP® questionnaire was assessed and compared to the serum leptin and insulin concentrations, hormones involved in regulation of food intake and biomarkers of adiposity and cardiometabolic risk. The community-based primary care research network for children in Toronto, Canada (TARGet Kids!) was used. The participants were children aged 3-5 years recruited from TARGet Kids! A total of 1856 children were recruited from seven primary care practices. Of these, 1086 children completed laboratory testing. Laboratory data for leptin and insulin were available for 714 and 1054 of those individuals, respectively. RESULTS The total NutriSTEP® score was significantly associated with serum leptin concentrations (p=0.003); for each unit increase in the total NutriSTEP® score, there was an increase of 0.01 ng/mL (95% confidence interval [CI] 0.004-0.018) in serum leptin concentrations after adjusting for potential confounders. The total NutriSTEP® score was not significantly associated with serum insulin concentration. CONCLUSIONS Parent reported nutritional risk is associated with serum leptin, but not insulin, concentrations in preschool-aged children. The NutriSTEP® questionnaire may be an effective tool for predicting future cardiometabolic risk in preschool-aged children.
Collapse
|
39
|
Iwakoshi-Ukena E, Shikano K, Kondo K, Taniuchi S, Furumitsu M, Ochi Y, Sasaki T, Okamoto S, Bentley GE, Kriegsfeld LJ, Minokoshi Y, Ukena K. Neurosecretory protein GL stimulates food intake, de novo lipogenesis, and onset of obesity. eLife 2017; 6. [PMID: 28799896 PMCID: PMC5553934 DOI: 10.7554/elife.28527] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 07/10/2017] [Indexed: 02/06/2023] Open
Abstract
Mechanisms underlying the central regulation of food intake and fat accumulation are not fully understood. We found that neurosecretory protein GL (NPGL), a newly-identified neuropeptide, increased food intake and white adipose tissue (WAT) in rats. NPGL-precursor gene overexpression in the hypothalamus caused increases in food intake, WAT, body mass, and circulating insulin when fed a high calorie diet. Intracerebroventricular administration of NPGL induced de novo lipogenesis in WAT, increased insulin, and it selectively induced carbohydrate intake. Neutralizing antibody administration decreased the size of lipid droplets in WAT. Npgl mRNA expression was upregulated by fasting and low insulin levels. Additionally, NPGL-producing cells were responsive to insulin. These results point to NPGL as a novel neuronal regulator that drives food intake and fat deposition through de novo lipogenesis and acts to maintain steady-state fat level in concert with insulin. Dysregulation of NPGL may be a root cause of obesity. DOI:http://dx.doi.org/10.7554/eLife.28527.001 Throughout history, our ancestors needed to accumulate fat to survive during times when food sources were scarce. However, for most people in the modern age, food is abundant and eating too much is a major cause of weight gain, obesity and diseases affecting the metabolism. Obesity in particular, can lead to diseases such as diabetes and heart disease. Hunger and appetite are regulated by proteins and other chemicals that act as messengers, for example insulin, and a region of the brain called the hypothalamus. However, the full mechanisms that regulate these sensations remain unclear. Only recently, a protein called NPGL was discovered in a part of the hypothalamus of birds and mammals. However, it was not known if NPGL plays a role in regulating eating habits and weight gain. Iwakoshi-Ukena et al. have now discovered that NPGL is found in the hypothalamus of rats and is regulated by diet and insulin. When the gene for NPGL was manipulated to produce too much of the protein, rats fed a high calorie diet started to eat more, and gained more weight and body fat. Adding additional NPGL to their brains had the same effect. When the animals were fed a normal diet, NPGL only moderately affected how much they ate, but it substantially increased how much fat they produced. Iwakoshi-Ukena et al. also observed that when animals were starved and insulin levels were low, the rats started to produce more NPGL. These results suggest that NPGL plays a role in fat storage when energy sources are limited, and can contribute to obesity when too much NPGL is produced in animals on a high calorie diet. These findings indicate that NPGL could be an additional brain chemical that regulates hunger and fat storage in mammals. A next step will be to reveal the specific mechanisms by which NPGL regulates overeating and fat accumulation. These findings will further advance the study and treatment of obesity and obesity-related diseases. DOI:http://dx.doi.org/10.7554/eLife.28527.002
Collapse
Affiliation(s)
- Eiko Iwakoshi-Ukena
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan.,Department of Integrative Biology and The Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, United States
| | - Kenshiro Shikano
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan
| | - Kunihiro Kondo
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan
| | - Shusuke Taniuchi
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan
| | - Megumi Furumitsu
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yuta Ochi
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan
| | - Tsutomu Sasaki
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Shiki Okamoto
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, Sokendai (The Graduate University for Advanced Studies), Hayama, Japan.,Second Department of Internal Medicine (Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology), Graduate School of Medicine, University of the Ryukyus, Nakagami-gun, Japan
| | - George E Bentley
- Department of Integrative Biology and The Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, United States
| | - Lance J Kriegsfeld
- Department of Psychology and The Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, United States
| | - Yasuhiko Minokoshi
- Division of Endocrinology and Metabolism, Department of Homeostatic Regulation, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Science, Sokendai (The Graduate University for Advanced Studies), Hayama, Japan
| | - Kazuyoshi Ukena
- Section of Behavioral Sciences, Graduate School of Integrated Arts and Sciences, Hiroshima University, Higashi-Hiroshima, Japan.,Department of Psychology and The Helen Wills Neuroscience Institute, University of California at Berkeley, Berkeley, United States
| |
Collapse
|
40
|
Lee AA, Owyang C. Sugars, Sweet Taste Receptors, and Brain Responses. Nutrients 2017; 9:nu9070653. [PMID: 28672790 PMCID: PMC5537773 DOI: 10.3390/nu9070653] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/20/2017] [Accepted: 06/21/2017] [Indexed: 12/25/2022] Open
Abstract
Sweet taste receptors are composed of a heterodimer of taste 1 receptor member 2 (T1R2) and taste 1 receptor member 3 (T1R3). Accumulating evidence shows that sweet taste receptors are ubiquitous throughout the body, including in the gastrointestinal tract as well as the hypothalamus. These sweet taste receptors are heavily involved in nutrient sensing, monitoring changes in energy stores, and triggering metabolic and behavioral responses to maintain energy balance. Not surprisingly, these pathways are heavily regulated by external and internal factors. Dysfunction in one or more of these pathways may be important in the pathogenesis of common diseases, such as obesity and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Allen A Lee
- 1500 East Medical Center Drive, Division of Gastroenterology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109-5362, USA.
| | - Chung Owyang
- 3912 Taubman Center, SPC 5362, Ann Arbor, MI 48109-5362, USA.
| |
Collapse
|
41
|
Tundo GR, Sbardella D, Ciaccio C, Grasso G, Gioia M, Coletta A, Polticelli F, Di Pierro D, Milardi D, Van Endert P, Marini S, Coletta M. Multiple functions of insulin-degrading enzyme: a metabolic crosslight? Crit Rev Biochem Mol Biol 2017. [PMID: 28635330 DOI: 10.1080/10409238.2017.1337707] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Insulin-degrading enzyme (IDE) is a ubiquitous zinc peptidase of the inverzincin family, which has been initially discovered as the enzyme responsible for insulin catabolism; therefore, its involvement in the onset of diabetes has been largely investigated. However, further studies on IDE unraveled its ability to degrade several other polypeptides, such as β-amyloid, amylin, and glucagon, envisaging the possible implication of IDE dys-regulation in the "aggregopathies" and, in particular, in neurodegenerative diseases. Over the last decade, a novel scenario on IDE biology has emerged, pointing out a multi-functional role of this enzyme in several basic cellular processes. In particular, latest advances indicate that IDE behaves as a heat shock protein and modulates the ubiquitin-proteasome system, suggesting a major implication in proteins turnover and cell homeostasis. In addition, recent observations have highlighted that the regulation of glucose metabolism by IDE is not merely based on its largely proposed role in the degradation of insulin in vivo. There is increasing evidence that improper IDE function, regulation, or trafficking might contribute to the etiology of metabolic diseases. In addition, the enzymatic activity of IDE is affected by metals levels, thus suggesting a role also in the metal homeostasis (metallostasis), which is thought to be tightly linked to the malfunction of the "quality control" machinery of the cell. Focusing on the physiological role of IDE, we will address a comprehensive vision of the very complex scenario in which IDE takes part, outlining its crucial role in interconnecting several relevant cellular processes.
Collapse
Affiliation(s)
- Grazia R Tundo
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Diego Sbardella
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Chiara Ciaccio
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Giuseppe Grasso
- d Department of Chemistry , University of Catania , Catania , Italy.,e CNR IBB , Catania , Italy
| | - Magda Gioia
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | - Andrea Coletta
- f Department of Chemistry , University of Aarhus , Aarhus , Denmark
| | | | - Donato Di Pierro
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy
| | | | - Peter Van Endert
- h Université Paris Descartes, INSERM, U1151, CNRS , Paris , France
| | - Stefano Marini
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| | - Massimo Coletta
- a Department of Clinical Sciences and Translation Medicine , University of Roma Tor Vergata , Roma , Italy.,b CIRCMSB , Bari , Italy.,c Center for TeleInfrastructures, University of Roma Tor Vergata , Roma , Italy
| |
Collapse
|
42
|
Russell AL, Grimes JM, Cruthirds DF, Westerfield J, Wooten L, Keil M, Weiser MJ, Landauer MR, Handa RJ, Wu TJ, Larco DO. Dietary Isoflavone-Dependent and Estradiol Replacement Effects on Body Weight in the Ovariectomized (OVX) Rat. Horm Metab Res 2017; 49:457-465. [PMID: 28482370 PMCID: PMC5820000 DOI: 10.1055/s-0043-108250] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
17β-Estradiol is known to regulate energy metabolism and body weight. Ovariectomy results in body weight gain while estradiol administration results in a reversal of weight gain. Isoflavones, found in rodent chow, can mimic estrogenic effects making it crucial to understand the role of these compounds on metabolic regulation. The goal of this study is to examine the effect of dietary isoflavones on body weight regulation in the ovariectomized rat. This study will examine how dietary isoflavones can interact with estradiol treatment to affect body weight. Consistent with previous findings, animals fed an isoflavone-rich diet had decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin levels (p<0.05) compared to animals fed an isoflavone-free diet. Estradiol replacement resulted in decreased body weight (p<0.05), abdominal fat (p<0.05), and serum leptin (p<0.05). Current literature suggests the involvement of cytokines in the inflammatory response of body weight gain. We screened a host of cytokines and chemokines that may be altered by dietary isoflavones or estradiol replacement. Serum cytokine analysis revealed significant (p<0.05) diet-dependent increases in inflammatory cytokines (keratinocyte-derived chemokine). The isoflavone-free diet in OVX rats resulted in the regulation of the following cytokines and chemokines: interleukin-10, interleukin-18, serum regulated on activation, normal T cell expressed and secreted, and monocyte chemoattractant protein-1 (p<0.05). Overall, these results reveal that estradiol treatment can have differential effects on energy metabolism and body weight regulation depending on the presence of isoflavones in rodent chow.
Collapse
Affiliation(s)
- Ashley L. Russell
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Jamie Moran Grimes
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Danette F. Cruthirds
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Joanna Westerfield
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lawren Wooten
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | - Michael J. Weiser
- DSM Nutritional Products Inc., Human Nutrition & Health, Boulder, Colorado, USA
| | - Michael R. Landauer
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert J. Handa
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - T. John Wu
- Program in Neuroscience, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Darwin O. Larco
- Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
43
|
Ren P, Yang XJ, Cui SQ, Kim JS, Menon D, Baidoo SK. Effects of different feeding levels during three short periods of gestation on gilt and litter performance, nutrient digestibility, and energy homeostasis in gilts. J Anim Sci 2017; 95:1232-1242. [PMID: 28380514 DOI: 10.2527/jas.2016.1208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The present study investigated the effects of different feeding levels during 3 short periods of gestation on gilt and litter performance, apparent total tract digestibility (ATTD) of energy and nutrients, and energy homeostasis in gilts. A total of 18 gilts were allotted to 1 of 3 dietary treatments using a completely randomized design. All gilts were fed 1 common corn-soybean meal-based diet with the amount of 1.0 × maintenance energy intake (100 × BW (BW) kcal ME/d) throughout gestation except 3 periods of 7 d when dietary treatments were imposed on d 27, d 55, and d 83 of gestation. During the 3 short periods, gilts were fed 1 of 3 different feeding levels: 0.5, 1.0, and 2.0 × maintenance energy level (0.5M, 1.0M, and 2.0M, respectively). Results showed that gilts on 2.0M feeding level had higher ( < 0.05) weight gain from d 27 to 109 of gestation (37.05 vs. 15.34 kg) and greater ( < 0.05) BW change, average daily gain, and gain to feed ratio during gestation periods 1 (d 27-34) and 3 (d 83-90) when compared with gilts on 0.5M feeding level. No differences ( > 0.10) in litter performance were observed among the 3 feeding levels. Additionally, the slopes of BW change in response to feeding levels in period 1 were 4.32 kg/0.5M change from 0.5M to 1.0M feeding level and 3.72 kg/0.5M change from 1.0M to 2.0M feeding level, respectively. There were quadratic ( < 0.05) effects of feeding levels on ATTD of dry matter and gross energy during periods 1 and 2 (d 55-62). Furthermore, fasting plasma concentrations of acyl ghrelin and nonesterified fatty acid (NEFA) in period 1 were greater ( < 0.01) in gilts on 0.5M feeding level than those on 2.0M feeding level. In conclusion, increasing feeding levels during 3 short periods increased primiparous sow performance during these short periods but did not affect litter performance. ATTD of energy and nutrients, and BW change efficiency were maximized for gilts on 1.0 M feeding level. The data also indicated that sows on the lowest feeding level were exposed to negative energy balance as evidenced by the higher plasma acyl ghrelin and NEFA concentrations.
Collapse
|
44
|
Evans MC, Anderson GM. Neuroendocrine integration of nutritional signals on reproduction. J Mol Endocrinol 2017; 58:R107-R128. [PMID: 28057770 DOI: 10.1530/jme-16-0212] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 01/05/2017] [Indexed: 12/28/2022]
Abstract
Reproductive function in mammals is energetically costly and therefore tightly regulated by nutritional status. To enable this integration of metabolic and reproductive function, information regarding peripheral nutritional status must be relayed centrally to the gonadotropin-releasing hormone (GNRH) neurons that drive reproductive function. The metabolically relevant hormones leptin, insulin and ghrelin have been identified as key mediators of this 'metabolic control of fertility'. However, the neural circuitry through which they act to exert their control over GNRH drive remains incompletely understood. With the advent of Cre-LoxP technology, it has become possible to perform targeted gene-deletion and gene-rescue experiments and thus test the functional requirement and sufficiency, respectively, of discrete hormone-neuron signaling pathways in the metabolic control of reproductive function. This review discusses the findings from these investigations, and attempts to put them in context with what is known from clinical situations and wild-type animal models. What emerges from this discussion is clear evidence that the integration of nutritional signals on reproduction is complex and highly redundant, and therefore, surprisingly difficult to perturb. Consequently, the deletion of individual hormone-neuron signaling pathways often fails to cause reproductive phenotypes, despite strong evidence that the targeted pathway plays a role under normal physiological conditions. Although transgenic studies rarely reveal a critical role for discrete signaling pathways, they nevertheless prove to be a good strategy for identifying whether a targeted pathway is absolutely required, critically involved, sufficient or dispensable in the metabolic control of fertility.
Collapse
Affiliation(s)
- Maggie C Evans
- Centre for Neuroendocrinology and Department of AnatomyUniversity of Otago School of Medical Sciences, Dunedin, New Zealand
| | - Greg M Anderson
- Centre for Neuroendocrinology and Department of AnatomyUniversity of Otago School of Medical Sciences, Dunedin, New Zealand
| |
Collapse
|
45
|
Levin BE, Lutz TA. Amylin and Leptin: Co-Regulators of Energy Homeostasis and Neuronal Development. Trends Endocrinol Metab 2017; 28:153-164. [PMID: 27938937 DOI: 10.1016/j.tem.2016.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 12/18/2022]
Abstract
While the regulation of energy homeostasis by amylin is already well-characterized, emerging data suggest that amylin is also crucial for the development of neural pathways in the hypothalamus and caudal hindbrain (area postrema, AP; nucleus tractus solitarius, NTS). Exciting new findings demonstrate crucial amylin-leptin interactions in altering the activity of specific hypothalamic and AP neurons, and a role for amylin as a novel class of 'leptin sensitizers' which enhance leptin signaling in both leptin-sensitive and -resistant individuals, in part by stimulating IL-6 production by hypothalamic microglia. This review summarizes these findings and provides a hypothetical framework for future studies to elucidate the mechanisms by which amylin and leptin act individually and as co-conspirators to alter energy homeostasis and neuronal development.
Collapse
Affiliation(s)
- Barry E Levin
- Department of Neurology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA.
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
46
|
Chen W, Balland E, Cowley MA. Hypothalamic Insulin Resistance in Obesity: Effects on Glucose Homeostasis. Neuroendocrinology 2017; 104:364-381. [PMID: 28122381 DOI: 10.1159/000455865] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/04/2017] [Indexed: 01/05/2023]
Abstract
The central link between obesity and type 2 diabetes is the development of insulin resistance. To date, it is still not clear whether hyperinsulinemia causes insulin resistance, which underlies the pathogenesis of obesity-associated type 2 diabetes, owing to the sophisticated regulatory mechanisms that exist in the periphery and in the brain. In recent years, accumulating evidence has demonstrated the existence of insulin resistance within the hypothalamus. In this review, we have integrated the recent discoveries surrounding both central and peripheral insulin resistance to provide a comprehensive overview of insulin resistance in obesity and the regulation of systemic glucose homeostasis. In particular, this review will discuss how hyperinsulinemia and hyperleptinemia in obesity impair insulin sensitivity in tissues such as the liver, skeletal muscle, adipose tissue, and the brain. In addition, this review highlights insulin transport into the brain, signaling pathways associated with hypothalamic insulin receptor expression in the regulation of hepatic glucose production, and finally the perturbation of systemic glucose homeostasis as a consequence of central insulin resistance. We also suggest future approaches to overcome both central and peripheral insulin resistance to treat obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Weiyi Chen
- Department of Physiology/Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | | |
Collapse
|
47
|
van de Pol I, Flik G, Gorissen M. Comparative Physiology of Energy Metabolism: Fishing for Endocrine Signals in the Early Vertebrate Pool. Front Endocrinol (Lausanne) 2017; 8:36. [PMID: 28303116 PMCID: PMC5332387 DOI: 10.3389/fendo.2017.00036] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/10/2017] [Indexed: 01/23/2023] Open
Abstract
Energy is the common currency of life. To guarantee a homeostatic supply of energy, multiple neuro-endocrine systems have evolved in vertebrates; systems that regulate food intake, metabolism, and distribution of energy. Even subtle (lasting) dysregulation of the delicate balance of energy intake and expenditure may result in severe pathologies. Feeding-related pathologies have fueled research on mammals, including of course the human species. The mechanisms regulating food intake and body mass are well-characterized in these vertebrates. The majority of animal life is ectothermic, only birds and mammals are endotherms. What can we learn from a (comparative) study on energy homeostasis in teleostean fishes, ectotherms, with a very different energy budget and expenditure? We present several adaptation strategies in fish. In recent years, the components that regulate food intake in fishes have been identified. Although there is homology of the major genetic machinery with mammals (i.e., there is a vertebrate blueprint), in many cases this does not imply analogy. Although both mammals and fish must gain their energy from food, the expenditure of the energy obtained is different. Mammals need to spend vast amounts of energy to maintain body temperature; fishes seem to utilize a broader metabolic range to their advantage. In this review, we briefly discuss ecto- and endothermy and their consequences for energy balance. Next, we argue that the evolution of endothermy and its (dis-)advantages may explain very different strategies in endocrine regulation of energy homeostasis among vertebrates. We follow a comparative and evolutionary line of thought: we discuss similarities and differences between fish and mammals. Moreover, given the extraordinary radiation of teleostean fishes (with an estimated number of 33,400 contemporary species, or over 50% of vertebrate life forms), we also compare strategies in energy homeostasis between teleostean species. We present recent developments in the field of (neuro)endocrine regulation of energy balance in teleosts, with a focus on leptin.
Collapse
Affiliation(s)
- Iris van de Pol
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, Netherlands
| | - Gert Flik
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, Netherlands
- *Correspondence: Gert Flik,
| | - Marnix Gorissen
- Department of Animal Ecology and Physiology, Institute for Water and Wetland Research, Radboud University, Nijmegen, Netherlands
| |
Collapse
|
48
|
Roh E, Kim MS. Brain Regulation of Energy Metabolism. Endocrinol Metab (Seoul) 2016; 31:519-524. [PMID: 28029023 PMCID: PMC5195827 DOI: 10.3803/enm.2016.31.4.519] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 09/20/2016] [Accepted: 09/30/2016] [Indexed: 11/11/2022] Open
Abstract
In healthy individuals, energy intake is in balance with energy expenditure, which helps to maintain a normal body weight. The brain's inability to control energy homeostasis underlies the pathology of hyperphagia and obesity. The brain detects body energy excess and deficit by sensing the levels of circulating metabolic hormones and nutrients and by receiving metabolic information from the periphery via the autonomic nervous system. A specialized neuronal network coordinates energy intake behavior and the metabolic processes affecting energy expenditure. Here, we briefly review neuronal mechanisms by which our body maintains energy balance.
Collapse
Affiliation(s)
- Eun Roh
- Department of Biomedical Science, University of Ulsan College of Medicine, Seoul, Korea
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Min Seon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea.
| |
Collapse
|
49
|
Belfort-DeAguiar R, Seo D, Naik S, Hwang J, Lacadie C, Schmidt C, Constable RT, Sinha R, Sherwin R. Food image-induced brain activation is not diminished by insulin infusion. Int J Obes (Lond) 2016; 40:1679-1686. [PMID: 27569684 PMCID: PMC5101182 DOI: 10.1038/ijo.2016.152] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 07/26/2016] [Accepted: 08/07/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND/OBJECTIVES The obesity epidemic appears to be driven in large part by our modern environment inundated by food cues, which may influence our desire to eat. Although insulin decreases food intake in both animals and humans, the effect of insulin on motivation for food in the presence of food cues is not known. Therefore, the aim of this study was to evaluate the effect of an intravenous insulin infusion on the brain response to visual food cues, hunger and food craving in non-obese human subjects. SUBJECTS/METHODS Thirty-four right-handed healthy non-obese subjects (19F/15M, age: 29±8 years.; BMI: 23.1±2.1 kg m-2) were divided in two groups matched by age and BMI; the insulin group (18 subjects) underwent a hyperinsulinemic-euglycemic-clamp, and the control group (16 subjects) received an intravenous saline infusion, while viewing high and low-calorie food and non-food pictures during a functional MRI scan. Motivation for food was determined via analog scales for hunger, wanting and liking ratings. RESULTS Food images induced brain responses in the hypothalamus, striatum, amygdala, insula, ventromedial prefrontal cortex (PFC), dorsolateral PFC and occipital lobe (whole brain correction, P<0.05). Wanting (P<0.001) and liking (P<0.001) ratings were significantly higher for the food than the non-food images, but not different between insulin and saline infusion groups. Hunger ratings increased throughout the MRI scan and correlated with preference for high-calorie food pictures (r=0.70; P<0.001). However, neither brain activity nor food cravings were affected by hyperinsulinemia or hormonal status (leptin and ghrelin levels) (P=NS). CONCLUSIONS Our data demonstrate that visual food cues induce a strong response in motivation/reward and cognitive-executive control brain regions in non-obese subjects, but that these responses are not diminished by hyperinsulinemia per se. These findings suggest that our modern food cue saturated environment may be sufficient to overpower homeostatic hormonal signals, and thus contribute to the current obesity epidemic.
Collapse
Affiliation(s)
| | - Dongju Seo
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Sarita Naik
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
- University College London Hospitals NHS, London, UK
| | - Janice Hwang
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Cheryl Lacadie
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Christian Schmidt
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - R. Todd Constable
- Department of Radiology, Yale University School of Medicine, New Haven, CT, USA
| | - Rajita Sinha
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Robert Sherwin
- Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
50
|
Volta F, Gerdes JM. The role of primary cilia in obesity and diabetes. Ann N Y Acad Sci 2016; 1391:71-84. [DOI: 10.1111/nyas.13216] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/19/2016] [Accepted: 08/01/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Francesco Volta
- Institute for Diabetes and Regeneration Research; Helmholtz Zentrum München; Garching Germany
| | - Jantje M. Gerdes
- Institute for Diabetes and Regeneration Research; Helmholtz Zentrum München; Garching Germany
- German Center for Diabetes Research; DZD; Munich Germany
| |
Collapse
|