251
|
Uchida Y, Tsunekawa C, Sato I. Systemic acyl-ghrelin increases tail skin temperature in rats without affecting their thermoregulatory behavior in a cold environment. Neurosci Lett 2020; 737:135306. [PMID: 32822766 DOI: 10.1016/j.neulet.2020.135306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/06/2020] [Accepted: 08/12/2020] [Indexed: 10/23/2022]
Abstract
Fasting increases ghrelin that is a peptide hormone with two circulating isoforms, acyl and des-acyl ghrelin. We reported that fasting or des-acyl ghrelin facilitates behavioral thermoregulation in the cold in rats assessed by tail-hiding behavior that was the indicator of rats' thermoregulatory behavior in the cold; however, the effect of acyl-ghrelin on the same process remains to be elucidated. We investigated the effect of acyl-ghrelin on thermoregulatory behavior in the cold in rats. The animals received an intraperitoneal saline or 24 μg acyl-ghrelin injection and were exposed to 27 °C or 15 °C for 2 h, while their body temperature, tail skin temperature, and tail-hiding behavior were constantly monitored. cFos immunoreactive (cFos-IR) cells in the median preoptic area, medial preoptic area, paraventricular nucleus (PVN), and arcuate nucleus were counted. Body temperature and the duration of thermoregulatory behavior did not show a significant difference between the acyl-ghrelin-treated and control groups at 15 °C; however, tail skin temperature in the acyl-ghrelin-treated group was higher than that in the control group. The number of cFos-IR cells in the PVN was greater in the control group than that in the acyl-ghrelin-treated group at 27 °C. These results indicate that acyl-ghrelin did not affect behavioral thermoregulation but might affect tail skin temperature in rats in the cold.
Collapse
Affiliation(s)
- Yuki Uchida
- Women's Environmental Science Laboratory, Department of Health Sciences, Faculty of Human Life and Environment, Nara Women's University, Nara, Japan.
| | - Chinami Tsunekawa
- Women's Environmental Science Laboratory, Department of Health Sciences, Faculty of Human Life and Environment, Nara Women's University, Nara, Japan
| | - Izumi Sato
- Women's Environmental Science Laboratory, Department of Health Sciences, Faculty of Human Life and Environment, Nara Women's University, Nara, Japan
| |
Collapse
|
252
|
Gupta S, Mitra A. Heal the heart through gut (hormone) ghrelin: a potential player to combat heart failure. Heart Fail Rev 2020; 26:417-435. [PMID: 33025414 DOI: 10.1007/s10741-020-10032-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Ghrelin, a small peptide hormone (28 aa), secreted mainly by X/A-like cells of gastric mucosa, is also locally produced in cardiomyocytes. Being an orexigenic factor (appetite stimulant), it promotes release of growth hormone (GH) and exerts diverse physiological functions, viz. regulation of energy balance, glucose, and/or fat metabolism for body weight maintenance. Interestingly, administration of exogenous ghrelin significantly improves cardiac functions in CVD patients as well as experimental animal models of heart failure. Ghrelin ameliorates pathophysiological condition of the heart in myocardial infarction, cardiac hypertrophy, fibrosis, cachexia, and ischemia reperfusion injury. This peptide also exerts significant impact at the level of vasculature leading to lowering high blood pressure and reversal of endothelial dysfunction and atherosclerosis. However, the molecular mechanism of actions elucidating the healing effects of ghrelin on the cardiovascular system is still a matter of conjecture. Some experimental data indicate its beneficial effects via complex cellular cross talks between autonomic nervous system and cardiovascular cells, some other suggest more direct receptor-mediated molecular actions via autophagy or ionotropic regulation and interfering with apoptotic and inflammatory pathways of cardiomyocytes and vascular endothelial cells. Here, in this review, we summarise available recent data to encourage more research to find the missing links of unknown ghrelin receptor-mediated pathways as we see ghrelin as a future novel therapy in cardiovascular protection.
Collapse
Affiliation(s)
- Shreyasi Gupta
- Department of Zoology, Triveni Devi Bhalotia College, Raniganj, Paschim Bardhaman, 713347, India
| | - Arkadeep Mitra
- Department of Zoology, City College , 102/1, Raja Rammohan Sarani, Kolkata, 700009, India.
| |
Collapse
|
253
|
Su M, Yan M, Yao J, Fang Y, Jin H, Gong Y. Unacylated Ghrelin Regulates Glucose-Sensitive Neurons Activity and Glycolipid Metabolism via Orexin-A Neurons in the Lateral Hypothalamic Area. Horm Metab Res 2020; 52:747-754. [PMID: 32731263 DOI: 10.1055/a-1207-1212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The objective of the study was to investigate the regulatory actions of unacylated ghrelin (UAG) on glucose-sensitive (GS) neurons and glycolipid metabolism in the lateral hypothalamus area (LHA) and its involvement with orexin-A-immunopositive neurons. The effects of UAG administered into the LHA on GS neurons discharges and glycolipid metabolism were detected by single neuron discharge recording, biochemical index analysis and quantitative real-time PCR; the level of c-fos protein in orexin-A-immunopositive neurons was observed using immunofluorescence staining. UAG microinjected into the LHA activated glucose-inhibited neurons, which were partially blocked by pre-administration of anti-orexin-A antibody in the LHA. Furthermore, UAG microinjected into the LHA significantly reduced serum triglycerides (TG), total cholesterol, low-density lipoprotein cholesterol, blood glucose, insulin and hepatic TG levels, while elevated serum high-density lipoprotein cholesterol levels. UAG elevated the mRNA expression of carnitine palmitoyltransferase-1 and reduced the mRNA expression of acetyl-CoA carboxylase-1 in the liver. The above-mentioned effects of UAG were partially blocked by pre-administration of anti-orexin-A antibody. The expressions of orexin-A and c-fos were observed in the LHA. After UAG injection into the LHA, some neurons showed double labeling, and the percentage of double-labeled orexin-A/c-fos neurons in orexin-A-immunopositive neurons increased significantly. UAG in the LHA regulates glycolipid metabolism by activating orexin-A-immunopositive neurons in the LHA.
Collapse
Affiliation(s)
- Manqing Su
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Meixing Yan
- Qingdao Women and Children's Hospital, Qingdao, China
| | - Jiatong Yao
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yanpeng Fang
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Hong Jin
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| | - Yanling Gong
- Department of Pharmacy, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, China
| |
Collapse
|
254
|
Shang L, Wang Y, Ren Y, Ai T, Zhou P, Hu L, Wang L, Li J, Li B. In vitro gastric emptying characteristics of konjac glucomannan with different viscosity and its effects on appetite regulation. Food Funct 2020; 11:7596-7610. [PMID: 32869813 DOI: 10.1039/d0fo01104e] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Konjac glucomannan (KGM) is associated with the satiety-enhancing property by imparting the food matrix with high viscosity. In the present study, rheology tests on KGM sol with different viscosities were conducted to understand its flow behavior as they presented in the mouth and stomach, and the in vitro gastric emptying characteristics of KGM were examined with a human gastric simulator. Then, their effects on subjective appetite, glycemia, and appetite-related hormones (insulin, GLP-1, PYY3-36, CCK-8, ghrelin) response were investigated by conducting a randomized, single-blind, crossover trial in 22 healthy adults (11 female and 11 male, mean age (years): 23.2 ± 2.0, BMI (kg m-2): 20.6 ± 2.1). The blood samples and ratings for subjective appetite were collected at regular time intervals after the subjects were fed with four test breakfasts (one control treatment and three experimental treatments) on four different days. An ad libitum lunch was provided to the subjects once they consumed the breakfasts and their food intake was recorded. As the viscosity increased, the gastric emptying rate was delayed despite a large part of the chyme viscosity lost during digestion. The satiating capacity of the test breakfast was significantly enhanced as its viscosity increased, the and subjects' sensation for hunger, fullness, desire-to-eat, and prospective food consumption differed significantly (p = 0.006, 0.000, 0.002, and 0.001, respectively) between the treatments. The secretion of glycemia and satiety-related hormones were beneficially modulated by the increased viscosity of the test meal but a small decrease in the ad libitum food intake was observed after the intervention of the viscous test breakfasts. Overall, elevating the meal viscosity moderately by using KGM could contribute to combating the challenge of hunger for people in the bodyweight management.
Collapse
Affiliation(s)
- Longchen Shang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Yi Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Yanyan Ren
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Tingyang Ai
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Peiyuan Zhou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Ling Hu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Ling Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Jing Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China. and Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, China
| |
Collapse
|
255
|
Cornejo MP, Barrile F, Cassano D, Aguggia JP, García Romero G, Reynaldo M, Andreoli MF, De Francesco PN, Perello M. Growth hormone secretagogue receptor in dopamine neurons controls appetitive and consummatory behaviors towards high-fat diet in ad-libitum fed mice. Psychoneuroendocrinology 2020; 119:104718. [PMID: 32535402 DOI: 10.1016/j.psyneuen.2020.104718] [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: 03/18/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 01/10/2023]
Abstract
Growth hormone secretagogue receptor (GHSR), the receptor for ghrelin, is expressed in key brain nuclei that regulate food intake. The dopamine (DA) pathways have long been recognized to play key roles mediating GHSR effects on feeding behaviors. Here, we aimed to determine the role of GHSR in DA neurons controlling appetitive and consummatory behaviors towards high fat (HF) diet. For this purpose, we crossed reactivable GHSR-deficient mice with DA transporter (DAT)-Cre mice, which express Cre recombinase under the DAT promoter that is active exclusively in DA neurons, to generate mice with GHSR expression limited to DA neurons (DAT-GHSR mice). We found that DAT-GHSR mice show an increase of c-Fos levels in brain areas containing DA neurons after ghrelin treatment, in a similar fashion as seen in wild-type mice; however, they did not increase food intake or locomotor activity in response to systemically- or centrally-administered ghrelin. In addition, we found that satiated DAT-GHSR mice displayed both anticipatory activity to scheduled HF diet exposure and HF intake in a binge-like eating protocol similar to those in wild-type mice, whereas GHSR-deficient mice displayed impaired responses. We conclude that GHSR expression in DA neurons is sufficient to both mediate increased anticipatory activity to a scheduled HF diet exposure and fully orchestrate binge-like HF intake, but it is insufficient to restore the acute orexigenic or locomotor effects of ghrelin treatment. Thus, GHSR in DA neurons affects appetitive and consummatory behaviors towards HF diet that take place in the absence of caloric needs.
Collapse
Affiliation(s)
- María Paula Cornejo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Franco Barrile
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Daniela Cassano
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Julieta Paola Aguggia
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Guadalupe García Romero
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Mirta Reynaldo
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - María Florencia Andreoli
- Laboratory of Experimental Neurodevelopment, Institute of Development and Pediatric Research (IDIP), La Plata Children's Hospital and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)], La Plata, Buenos Aires, Argentina
| | - Pablo Nicolás De Francesco
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina
| | - Mario Perello
- Laboratory of Neurophysiology of the Multidisciplinary Institute of Cell Biology [IMBICE, Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), National University of La Plata], 1900 La Plata, Buenos Aires, Argentina.
| |
Collapse
|
256
|
Koliaki C, Liatis S, Dalamaga M, Kokkinos A. The Implication of Gut Hormones in the Regulation of Energy Homeostasis and Their Role in the Pathophysiology of Obesity. Curr Obes Rep 2020; 9:255-271. [PMID: 32647952 DOI: 10.1007/s13679-020-00396-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW This review provides an update on the role of gut hormones and their interactions in the regulation of energy homeostasis, describes gut hormone adaptations in obesity and in response to weight loss, and summarizes the current evidence on the role of gut hormone-based therapies for obesity treatment. RECENT FINDINGS Gut hormones play a key role in regulating eating behaviour, energy and glucose homeostasis. Dysregulated gut hormone responses have been proposed to be pathogenetically involved in the development and perpetuation of obesity. Summarizing the major gut hormone changes in obesity, obese individuals are characterized by blunted postprandial ghrelin suppression, loss of premeal ghrelin peaks, impaired diurnal ghrelin variability and reduced fasting and postprandial levels of anorexigenic peptides. Adaptive alterations of gut hormone levels are implicated in weight regain, thus complicating hypocaloric dietary interventions, and can further explain the profound weight loss and metabolic improvement following bariatric surgery. A plethora of compounds mimicking gut hormone changes after bariatric surgery are currently under investigation, introducing a new era in the pharmacotherapy of obesity. The current trend is to combine different gut hormone receptor agonists and target multiple systems simultaneously, in order to replicate as closely as possible the gut hormone milieu after bariatric surgery and circumvent the counter-regulatory adaptive changes associated with dietary energy restriction. An increasing number of preclinical and early-phase clinical trials reveal the additive benefits obtained with dual or triple gut peptide receptor agonists in reducing body weight and improving glycaemia. Gut hormones act as potent regulators of energy and glucose homeostasis. Therapeutic strategies targeting their levels or receptors emerge as a promising approach to treat patients with obesity and hyperglycaemia.
Collapse
Affiliation(s)
- Chrysi Koliaki
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece.
| | - Stavros Liatis
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| | - Maria Dalamaga
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexander Kokkinos
- First Department of Propaedeutic Internal Medicine, Medical School, Laiko General Hospital, National Kapodistrian University of Athens, 17 Agiou Thoma Street, 11527, Athens, Greece
| |
Collapse
|
257
|
Al-Rawi N, Madkour M, Jahrami H, Salahat D, Alhasan F, BaHammam A, Al-Islam Faris M. Effect of diurnal intermittent fasting during Ramadan on ghrelin, leptin, melatonin, and cortisol levels among overweight and obese subjects: A prospective observational study. PLoS One 2020; 15:e0237922. [PMID: 32845924 PMCID: PMC7449475 DOI: 10.1371/journal.pone.0237922] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 08/06/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Levels of cortisol, melatonin, ghrelin, and leptin are highly correlated with circadian rhythmicity. The levels of these hormones are affected by sleep, feeding, and general behaviors, and fluctuate with light and dark cycles. During the fasting month of Ramadan, a shift to nighttime eating is expected to affect circadian rhythm hormones and, subsequently, the levels of melatonin, cortisol, ghrelin, and leptin. The present study aimed to examine the effect of diurnal intermittent fasting (DIF) during Ramadan on daytime levels of ghrelin, leptin, melatonin, and cortisol hormones in a group of overweight and obese subjects, and to determine how anthropometric, dietary, and lifestyle changes during the month of Ramadan correlate with these hormonal changes. METHODS Fifty-seven overweight and obese male (40) and female (17) subjects were enrolled in this study. Anthropometric measurements, dietary intake, sleep duration, and hormonal levels of serum ghrelin, leptin, melatonin, and salivary cortisol were assessed one week before the start of Ramadan fasting and after 28 days of fasting at fixed times of the day (11:00 am-1:00 pm). RESULTS At the end of Ramadan, serum levels of ghrelin, melatonin, and leptin significantly (P<0.001) decreased, while salivary cortisol did not change compared to the levels assessed in the pre-fasting state. CONCLUSIONS DIF during Ramadan significantly altered serum levels of ghrelin, melatonin, and serum leptin. Further, male sex and anthropometric variables were the most impacting factors on the tested four hormones. Further studies are needed to assess DIF's impact on the circadian rhythmicity of overweight and obese fasting people.
Collapse
Affiliation(s)
- Natheer Al-Rawi
- Department of Oral and Craniofacial Health Sciences, College of Dental Medicine/Research Institute of Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, UAE
| | - Mohamed Madkour
- Department of Medical Laboratory Sciences, College of Health Sciences/Research Institute of Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, UAE
| | - Haitham Jahrami
- Rehabilitation Services, Periphery Hospitals, Ministry of Health, Manama, Bahrain
- College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Dana Salahat
- Department of Medical Laboratory Sciences, College of Health Sciences/Research Institute of Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, UAE
| | - Fatima Alhasan
- Department of Medical Laboratory Sciences, College of Health Sciences/Research Institute of Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, UAE
| | - Ahmed BaHammam
- Department of Medicine, College of Medicine, University Sleep Disorders Center, King Saud University, Riyadh, Saudi Arabia
- The Strategic Technologies Program of the National Plan for Sciences and Technology and Innovation in the Kingdom of Saudi Arabia, Riyadh, Saudi Arabia
| | - Mo'ez Al-Islam Faris
- Department of Clinical Nutrition and Dietetics, College of Health Sciences/Research Institute of Medical and Health Sciences (RIMHS), University of Sharjah, Sharjah, UAE
| |
Collapse
|
258
|
Ewbank SN, Campos CA, Chen JY, Bowen AJ, Padilla SL, Dempsey JL, Cui JY, Palmiter RD. Chronic G q signaling in AgRP neurons does not cause obesity. Proc Natl Acad Sci U S A 2020; 117:20874-20880. [PMID: 32764144 PMCID: PMC7456117 DOI: 10.1073/pnas.2004941117] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Maintaining energy homeostasis requires coordinating physiology and behavior both on an acute timescale to adapt to rapid fluctuations in caloric intake and on a chronic timescale to regulate body composition. Hypothalamic agouti-related peptide (AgRP)-expressing neurons are acutely activated by caloric need, and this acute activation promotes increased food intake and decreased energy expenditure. On a longer timescale, AgRP neurons exhibit chronic hyperactivity under conditions of obesity and high dietary fat consumption, likely due to leptin resistance; however, the behavioral and metabolic effects of chronic AgRP neuronal hyperactivity remain unexplored. Here, we use chemogenetics to manipulate Gq signaling in AgRP neurons in mice to explore the hypothesis that chronic activation of AgRP neurons promotes obesity. Inducing chronic Gq signaling in AgRP neurons initially increased food intake and caused dramatic weight gain, in agreement with published data; however, food intake returned to baseline levels within 1 wk, and body weight returned to baseline levels within 60 d. Additionally, we found that, when mice had elevated body weight due to chronic Gq signaling in AgRP neurons, energy expenditure was not altered but adiposity and lipid metabolism were both increased, even under caloric restriction. These findings reveal that the metabolic and behavioral effects of chronic Gq signaling in AgRP neurons are distinct from the previously reported effects of acute Gq signaling and also of leptin insensitivity.
Collapse
Affiliation(s)
- Sedona N Ewbank
- Department of Biochemistry, University of Washington, Seattle, WA 98195;
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Carlos A Campos
- Department of Biochemistry, University of Washington, Seattle, WA 98195;
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Jane Y Chen
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Anna J Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Stephanie L Padilla
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - Joseph L Dempsey
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195
| | - Richard D Palmiter
- Department of Biochemistry, University of Washington, Seattle, WA 98195;
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| |
Collapse
|
259
|
Quaresma PGF, Teixeira PDS, Wasinski F, Campos AMP, List EO, Kopchick JJ, Donato J. Cholinergic neurons in the hypothalamus and dorsal motor nucleus of the vagus are directly responsive to growth hormone. Life Sci 2020; 259:118229. [PMID: 32781065 DOI: 10.1016/j.lfs.2020.118229] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 02/07/2023]
Abstract
AIMS Cholinergic neurons are distributed in brain areas containing growth hormone (GH)-responsive cells. We determined if cholinergic neurons are directly responsive to GH and the metabolic consequences of deleting the GH receptor (GHR) specifically in choline acetyltransferase (ChAT)-expressing cells. MAIN METHODS Mice received an acute injection of GH to detect neurons co-expressing ChAT and phosphorylated STAT5 (pSTAT5), a well-established marker of GH-responsive cells. For the physiological studies, mice carrying ablation of GHR exclusively in ChAT-expressing cells were produced and possible changes in energy and glucose homeostasis were determined when consuming regular chow or high-fat diet (HFD). KEY FINDINGS The majority of cholinergic neurons in the arcuate nucleus (60%) and dorsomedial nucleus (84%) of the hypothalamus are directly responsive to GH. Approximately 34% of pre-ganglionic parasympathetic neurons in the dorsal motor nucleus of the vagus also exhibited GH-induced pSTAT5. GH-induced pSTAT5 in these ChAT neurons was absent in GHR ChAT knockout mice. Mice carrying ChAT-specific GHR deletion, either in chow or HFD, did not exhibit significant changes in body weight, body adiposity, lean body mass, food intake, energy expenditure, respiratory quotient, ambulatory activity, serum leptin levels, glucose tolerance, insulin sensitivity and metabolic responses to 2-deoxy-d-glucose. However, GHR deletion in ChAT neurons caused decreased hypothalamic Pomc mRNA levels in HFD mice. SIGNIFICANCE Cholinergic neurons that regulate the metabolism are directly responsive to GH, although GHR signaling in these cells is not required for energy and glucose homeostasis. Thus, the physiological importance of GH action on cholinergic neurons still needs to be identified.
Collapse
Affiliation(s)
- Paula G F Quaresma
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Pryscila D S Teixeira
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Frederick Wasinski
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Ana M P Campos
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Jose Donato
- Universidade de Sao Paulo, Instituto de Ciencias Biomedicas, Departamento de Fisiologia e Biofisica, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
260
|
Sun S, Corbeels K, Desmet L, Segers A, Wang Q, Van Der Schueren B, Depoortere I. Involvement of the GHSR in the developmental programming and metabolic disturbances induced by maternal undernutrition. J Nutr Biochem 2020; 85:108468. [PMID: 32750410 DOI: 10.1016/j.jnutbio.2020.108468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
The mismatch between maternal undernutrition and adequate nutrition after birth increases the risk of developing metabolic diseases. We aimed to investigate whether the hyperghrelinemia during maternal undernourishment rewires the hypothalamic development of the offspring and contributes to the conversion to an obese phenotype when fed a high-fat diet (HFD). Pregnant C57BL/6 J, wild type (WT) and ghrelin receptor (GHSR)-/- mice were assigned to either a normal nourished (NN) group, or an undernutrition (UN) (30% food restricted) group. All pups were fostered by NN Swiss mice. After weaning, pups were fed a normal diet, followed by a HFD from week 9. Plasma ghrelin levels peaked at postnatal day 15 (P15) in both C57BL/6 J UN and NN pups. Hypothalamic Ghsr mRNA expression was upregulated at P15 in UN pups compared to NN pups and inhibited agouti-related peptide (AgRP) projections. Adequate lactation increased body weight of UN WT but not of GHSR-/- pups compared to NN littermates. After weaning with a HFD, body weight and food intake was higher in WT UN pups but lower in GHSR-/- UN pups than in NN controls. The GHSR prevented a decrease in ambulatory activity and oxygen consumption in UN offspring during ad libitum feeding. Maternal undernutrition triggers developmental changes in the hypothalamus in utero which were further affected by adequate feeding after birth during the postnatal period by affecting GHSR signaling. The GHSR contributes to the hyperphagia and the increase in body weight when maternal undernutrition is followed by an obesity prone life environment.
Collapse
Affiliation(s)
- Shu Sun
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Katrien Corbeels
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven,3000 Leuven, Belgium
| | - Louis Desmet
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Anneleen Segers
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Qiaoling Wang
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Bart Van Der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven,3000 Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium.
| |
Collapse
|
261
|
Qualls-Creekmore E, Marlatt KL, Aarts E, Bruce-Keller A, Church TS, Clément K, Fisher JO, Gordon-Larsen P, Morrison CD, Raybould HE, Ryan DH, Schauer PR, Spector AC, Spetter MS, Stuber GD, Berthoud HR, Ravussin E. What Should I Eat and Why? The Environmental, Genetic, and Behavioral Determinants of Food Choice: Summary from a Pennington Scientific Symposium. Obesity (Silver Spring) 2020; 28:1386-1396. [PMID: 32520444 PMCID: PMC7501251 DOI: 10.1002/oby.22806] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/29/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
This review details the proceedings of a Pennington Biomedical scientific symposium titled, "What Should I Eat and Why? The Environmental, Genetic, and Behavioral Determinants of Food Choice." The symposium was designed to review the literature about energy homeostasis, particularly related to food choice and feeding behaviors, from psychology to physiology. This review discusses the intrinsic determinants of food choice, including biological mechanisms (genetics), peripheral and central signals, brain correlates, and the potential role of the microbiome. This review also address the extrinsic determinants (environment) of food choice within our physical and social environments. Finally, this review reports the current treatment practices for the clinical management of eating-induced overweight and obesity. An improved understanding of these determinants will inform best practices for the clinical treatment and prevention of obesity. Strategies paired with systemic shifts in our public health policies and changes in our "obesogenic" environment will be most effective at attenuating the obesity epidemic.
Collapse
Affiliation(s)
- Emily Qualls-Creekmore
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Kara L. Marlatt
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Esther Aarts
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, The Netherlands
| | - Annadora Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Tim S. Church
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
- ACAP Health, Dallas, TX, USA
| | - Karine Clément
- Sorbonne Université, INSERM, Nutrition and Obesity: Systemic Approaches (NutriOmics) Research Unit, Paris, France
- Assistance Publique Hôpitaux de Paris, Nutrition e, Pitié-Salpêtrière Hospital, 47-83 bd de l’Hôpital, Paris, France
| | - Jennifer O. Fisher
- Center for Obesity Research and Education, Temple University, Philadelphia, PA, USA
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Christopher D. Morrison
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Helen E. Raybould
- Department of Anatomy, Physiology, Cell Biology, UC Davis School of Veterinary Medicine, Davis, CA, USA
| | - Donna H. Ryan
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Philip R. Schauer
- Bariatric and Metabolic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alan C. Spector
- Department of Psychology and Program in Neuroscience, Florida State University, Tallahassee, FL, USA
| | - Maartje S. Spetter
- Institute of Medical Psychology and Behavioral Neurobiology, University of Tübingen, Germany; School of Psychology, University of Birmingham, Edgbaston, Birmingham, UK
| | - Garret D. Stuber
- Departments of Anesthesiology, Pain Medicine & Pharmacology, University of Washington, Seattle, WA, USA
| | - Hans-Rudolf Berthoud
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA
| |
Collapse
|
262
|
Beutler LR, Corpuz TV, Ahn JS, Kosar S, Song W, Chen Y, Knight ZA. Obesity causes selective and long-lasting desensitization of AgRP neurons to dietary fat. eLife 2020; 9:e55909. [PMID: 32720646 PMCID: PMC7398661 DOI: 10.7554/elife.55909] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
Body weight is regulated by interoceptive neural circuits that track energy need, but how the activity of these circuits is altered in obesity remains poorly understood. Here we describe the in vivo dynamics of hunger-promoting AgRP neurons during the development of diet-induced obesity in mice. We show that high-fat diet attenuates the response of AgRP neurons to an array of nutritionally-relevant stimuli including food cues, intragastric nutrients, cholecystokinin and ghrelin. These alterations are specific to dietary fat but not carbohydrate or protein. Subsequent weight loss restores the responsiveness of AgRP neurons to exterosensory cues but fails to rescue their sensitivity to gastrointestinal hormones or nutrients. These findings reveal that obesity triggers broad dysregulation of hypothalamic hunger neurons that is incompletely reversed by weight loss and may contribute to the difficulty of maintaining a reduced weight.
Collapse
Affiliation(s)
| | | | - Jamie S Ahn
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Seher Kosar
- Howard Hughes Medical InstituteChevy ChaseUnited States
| | - Weimin Song
- Northwestern University Feinberg School of Medicine, Comprehensive Metabolic CoreChicagoUnited States
| | - Yiming Chen
- UCSF Department of PhysiologySan FranciscoUnited States
- UCSF Neuroscience Graduate ProgramSan FranciscoUnited States
| | - Zachary A Knight
- Howard Hughes Medical InstituteChevy ChaseUnited States
- UCSF Department of PhysiologySan FranciscoUnited States
- UCSF Neuroscience Graduate ProgramSan FranciscoUnited States
- Kavli Institute for Fundamental NeuroscienceSan FranciscoUnited States
| |
Collapse
|
263
|
Evolution of Inflammatory and Oxidative Stress Markers in Romanian Obese Male Patients with Type 2 Diabetes Mellitus after Laparoscopic Sleeve Gastrectomy: One Year Follow-Up. Metabolites 2020; 10:metabo10080308. [PMID: 32731443 PMCID: PMC7464585 DOI: 10.3390/metabo10080308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 12/04/2022] Open
Abstract
Geography is one of the key drivers of the significant variation in the etiopathogenic profile and prevalence of type 2 diabetes mellitus (T2DM) and obesity, therefore geographically based data are fundamental for implementing the appropriate interventions. Presently, the selection criteria of T2DM and obesity patients for laparoscopic sleeve gastrectomy (LSG) have not reached a worldwide consensus—highlighting the need for sharing experts’ guidance in the preoperative evaluation, choice of the interventional procedure, perioperative management and patient long-term care. The aim of the current study was to evaluate the impact of LSG on T2DM (T2DM) remission in Romanian obese male patients, based on a multiparametric, prospective investigation. We have conducted a randomized controlled study on 41 obese male participants with the body mass index (BMI) ≥ 30 kg/m2, aged 30–65 years, which were randomly divided in two study groups: one receiving conventional treatment and the second undergoing LSG. The clinical and anthropometrical parameters, resting metabolic rate, general biochemical status, adipocytes profile, gastrointestinal hormones levels, proinflammatory, oxidant and antioxidant profiles were determined at three time points: V1 (baseline), V2 (after six months) and V3 (after 12 months). Glycated hemoglobin (HbA1c), blood glucose levels, BMI, weight, visceral fat level, HDL-cholesterol, incretin hormones, proinflammatory and the oxidative stress status were significantly improved in the LSG versus conventional treatment group. This is the first study reporting on the evaluation of metabolic surgery impact on Romanian obese male patients with T2DM. Our results confirm that LSG could contribute to T2DM remission in patients with diabesity, but this beneficial effect seems to be critically influenced by the duration of T2DM rather than by the obesity status. Our results show that, in addition to the parameters included in the prediction algorithm, the proinsulin levels, proinsulin/insulin ratio and the visceral fat percentage could bring added value to the assessment of metabolic status.
Collapse
|
264
|
Augustine V, Lee S, Oka Y. Neural Control and Modulation of Thirst, Sodium Appetite, and Hunger. Cell 2020; 180:25-32. [PMID: 31923398 DOI: 10.1016/j.cell.2019.11.040] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/13/2019] [Accepted: 11/27/2019] [Indexed: 01/01/2023]
Abstract
The function of central appetite neurons is instructing animals to ingest specific nutrient factors that the body needs. Emerging evidence suggests that individual appetite circuits for major nutrients-water, sodium, and food-operate on unique driving and quenching mechanisms. This review focuses on two aspects of appetite regulation. First, we describe the temporal relationship between appetite neuron activity and consumption behaviors. Second, we summarize ingestion-related satiation signals that differentially quench individual appetite circuits. We further discuss how distinct appetite and satiation systems for each factor may contribute to nutrient homeostasis from the functional and evolutional perspectives.
Collapse
Affiliation(s)
- Vineet Augustine
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA; Department of Neuroscience, The Scripps Research Institute, La Jolla, California, USA
| | - Sangjun Lee
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Yuki Oka
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA.
| |
Collapse
|
265
|
Goswami C, Dezaki K, Wang L, Inui A, Seino Y, Yada T. Ninjin'yoeito Targets Distinct Ca 2+ Channels to Activate Ghrelin-Responsive vs. Unresponsive NPY Neurons in the Arcuate Nucleus. Front Nutr 2020; 7:104. [PMID: 32766273 PMCID: PMC7379896 DOI: 10.3389/fnut.2020.00104] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/08/2020] [Indexed: 12/22/2022] Open
Abstract
Appetite loss or anorexia substantially deteriorates quality of life in various diseases, and stand upstream of frailty. Neuropeptide Y (NPY) in the hypothalamic arcuate nucleus (ARC) and ghrelin released from stomach are potent inducers of appetite. We previously reported that Ninjin'yoeito, a Japanese kampo medicine comprising twelve herbs, restores food intake, and body weight in cisplatin-treated anorectic mice. Furthermore, Ninjin'yoeito increased cytosolic Ca2+ concentration ([Ca2+]i) in not only ghrelin-responsive but ghrelin-unresponsive NPY neurons in ARC. The cellular lineage/differentiation of ghrelin-unresponsive neuron is less defined but might alter along with aging and diet. This study examined the occupancy of ghrelin-unresponsive neurons among ARC NPY neurons in adult mice fed normal chow, and explored the mechanisms underlying Ninjin'yoeito-induced [Ca2+]i increases in ghrelin-unresponsive vs. ghrelin-responsive NPY neurons. Single ARC neurons were subjected to [Ca2+]i measurement and subsequent immunostaining for NPY. Ghrelin failed to increase [Ca2+]i in 42% of ARC NPY neurons. Ninjin'yoeito (10 μg/ml)-induced increases in [Ca2+]i were abolished in Ca2+ free condition in ghrelin-responsive and ghrelin-unresponsive ARC NPY neurons. Ninjin'yoeito-induced [Ca2+]i increases were inhibited by N-type Ca2+ channel blocker ω-conotoxin in the majority (17 of 20), while by L-type Ca2+ channel blocker nitrendipine in the minority (2 of 23), of ghrelin-responsive neurons. In contrast, Ninjin'yoeito-induced [Ca2+]i increases were inhibited by nitrendipine in the majority (14 of 17), while by ω-conotoxin in the minority (8 of 24), of ghrelin-unresponsive neurons. These results indicate that ghrelin-unresponsive neurons occur substantially among NPY neurons of ARC in adult mice fed normal chow. Ninjin'yoeito preferentially target N-type and L-type Ca2+ channels in the majority of ghrelin-responsive and ghrelin-unresponsive neurons, respectively, to increase [Ca2+]i. We suggest ARC N- and L-type Ca2+ channels as potential targets for activating, respectively, ghrelin-responsive, and unresponsive NPY neurons to treat anorexia.
Collapse
Affiliation(s)
- Chayon Goswami
- Division of Integrative Physiology, Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kobe, Japan.,Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan.,Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Katsuya Dezaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan.,Faculty of Pharmacy, Iryo Sosei University, Iwaki, Japan
| | - Lei Wang
- Division of Integrative Physiology, Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kobe, Japan.,Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Akio Inui
- Pharmacological Department of Herbal Medicine, Kagoshima University Graduate School of Medical & Dental Sciences, Kagoshima, Japan
| | - Yutaka Seino
- Division of Integrative Physiology, Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kobe, Japan.,Center for Diabetes Research, Division of Diabetes and Endocrinology, Kansai Electric Power Medical Research Institute, Kobe, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Center for Integrative Physiology, Kansai Electric Power Medical Research Institute, Kobe, Japan.,Division of Diabetes, Metabolism and Endocrinology, Kobe University Graduate School of Medicine, Kobe, Japan.,Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, Tochigi, Japan.,Pharmacological Department of Herbal Medicine, Kagoshima University Graduate School of Medical & Dental Sciences, Kagoshima, Japan
| |
Collapse
|
266
|
Role of TRPV1/TRPV3 channels in olanzapine-induced metabolic alteration: Possible involvement in hypothalamic energy-sensing, appetite regulation, inflammation and mesolimbic pathway. Toxicol Appl Pharmacol 2020; 402:115124. [PMID: 32652086 DOI: 10.1016/j.taap.2020.115124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 06/15/2020] [Accepted: 06/19/2020] [Indexed: 12/30/2022]
Abstract
Atypical antipsychotics (AAPs) have the tendency of inducing severe metabolic alterations like obesity, diabetes mellitus, insulin resistance, dyslipidemia and cardiovascular complications. These alterations have been attributed to altered hypothalamic appetite regulation, energy sensing, insulin/leptin signaling, inflammatory reactions and active reward anticipation. Line of evidence suggests that transient receptor potential vanilloid type 1 and 3 (TRPV1 and TRPV3) channels are emerging targets in treatment of obesity, diabetes mellitus and could modulate feed intake. The present study was aimed to investigate the putative role TRPV1/TRPV3 in olanzapine-induced metabolic alterations in mice. Female BALB/c mice were treated with olanzapine for six weeks to induce metabolic alterations. Non-selective TRPV1/TRPV3 antagonist (ruthenium red) and selective TRPV1 (capsazepine) and TRPV3 antagonists (2,2-diphenyltetrahydrofuran or DPTHF) were used to investigate the involvement of TRPV1/TRPV3 in chronic olanzapine-induced metabolic alterations. These metabolic alterations were differentially reversed by ruthenium red and capsazepine, while DPTHF didn't show any significant effect. Olanzapine treatment also altered the mRNA expression of hypothalamic appetite-regulating and nutrient-sensing factors, inflammatory genes and TRPV1/TRPV3, which were reversed with ruthenium red and capsazepine treatment. Furthermore, olanzapine treatment also increased expression of TRPV1/TRPV3 in nucleus accumbens (NAc), TRPV3 expression in ventral tegmental area (VTA), which were reversed by the respective antagonists. However, DPTHF treatment showed reduced feed intake in olanzapine treated mice, which might be due to TRPV3 specific antagonism and reduced hedonic feed intake. In conclusion, our results suggested the putative role TRPV1 in hypothalamic dysregulations and TRPV3 in the mesolimbic pathway; both regulate feeding in olanzapine treated mice.
Collapse
|
267
|
Dailey RE, Smith K, Fontaine C, Jia Y, Avery JP. Response of metabolic hormones and blood metabolites to realimentation in rehabilitated harbor seal (Phoca vitulina) pups. J Comp Physiol B 2020; 190:629-640. [PMID: 32617718 DOI: 10.1007/s00360-020-01290-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 05/13/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022]
Abstract
Mammals with increased requirements for adipose tissue stores, such as marine mammals, have altered nutrient allocation priorities compared to many terrestrial mammals and thus the physiological response to undernutrition (low nutritional status) and realimentation (refeeding) may differ. Key regulators of nutrient allocation and tissue specific growth include metabolic hormones of the somatotropic axis, growth hormone (GH) and insulin-like growth factor (IGF)-I, as well as satiety and adipose promoting ghrelin and the stress hormone cortisol. Longitudinal measurements of metabolic hormones, blood metabolites, and morphometrics were collected over a 10-week period in twelve (male n = 3, female n = 9) harbor seal pups (< 6 weeks of age). Blood metabolites were used to indicate metabolic response during realimentation while morphometrics estimated tissue specific growth priorities. Harbor seal pups undergoing refeeding after nutritional deprivation show a preference for protein sparing despite severe malnutrition. Both BUN and total protein were negatively associated with GH and positively associated with IGF-I and ghrelin highlighting the importance of these metabolic hormones in the regulation of protein metabolism. While the response of the somatotropic axis to realimentation was typical of the mammalian pattern, the surprising increase of ghrelin across the study period suggests the priority of adipose accretion in addition to a possible mechanism regulating compensatory growth of vital adipose stores in a species, which prioritizes adipose accretion for survival.
Collapse
Affiliation(s)
- Rachael E Dailey
- Department of Biological Sciences, University of North Florida, Jacksonville, FL, USA
| | - Kacie Smith
- Department of Biological Sciences, University of North Florida, Jacksonville, FL, USA
| | | | - Yisu Jia
- Department of Mathematics and Statistics, University of North Florida, Jacksonville, FL, USA
| | - Julie P Avery
- Department of Biological Sciences, University of North Florida, Jacksonville, FL, USA. .,Water and Environmental Research Center, University of Alaska Fairbanks, 1764 Tanana Loop, PO Box 75 5910, Fairbanks, AK, 99775-5910, USA.
| |
Collapse
|
268
|
Bernardoni F, Bernhardt N, Pooseh S, King JA, Geisler D, Ritschel F, Boehm I, Seidel M, Roessner V, Smolka MN, Ehrlich S. Metabolic state and value-based decision-making in acute and recovered female patients with anorexia nervosa. J Psychiatry Neurosci 2020; 45:253-261. [PMID: 32129584 PMCID: PMC7828930 DOI: 10.1503/jpn.190031] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Patients with anorexia nervosa forgo eating despite emaciation and severe health consequences. Such dysfunctional decision-making might be explained by an excessive level of self-control, alterations in homeostatic and hedonic regulation, or an interplay between these processes. We aimed to understand value-based decision-making in anorexia nervosa and its association with the gut hormone ghrelin. Besides its homeostatic function, ghrelin has been implicated in the hedonic regulation of appetite and reward via the modulation of phasic dopamine signalling. METHODS In a cross-sectional design, we studied acutely underweight (n = 94) and recovered (n = 37) patients with anorexia nervosa of the restrictive subtype, as well as healthy control participants (n = 119). We assessed plasma concentrations of desacyl ghrelin and parameters of delay discounting, probability discounting for gains and losses, and loss aversion. RESULTS Recovered patients displayed higher risk aversion for gains, but we observed no group differences for the remaining decision-making parameters. Desacyl ghrelin was higher in acutely underweight and recovered participants with anorexia nervosa relative to healthy controls. Moreover, we found a significant group × desacyl ghrelin interaction in delay discounting, indicating that in contrast to healthy controls, acutely underweight patients with anorexia nervosa who had high desacyl ghrelin concentrations preferably chose the delayed reward option. LIMITATIONS We probed decision-making using monetary rewards, but patients with anorexia nervosa may react differently to disorder-relevant stimuli. Furthermore, in contrast to acyl ghrelin, the functions of desacyl ghrelin are unclear. Therefore, the interpretation of the results is preliminary. CONCLUSION The propensity for risk aversion as found in recovered patients with anorexia nervosa could help them successfully complete therapy, or it could reflect sequelae of the disorder. Conversely, ghrelin findings might be related to a mechanism contributing to disease maintenance; that is, in acutely underweight anorexia nervosa, a hungry state may facilitate the ability to forgo an immediate reward to achieve a (dysfunctional) long-term goal.
Collapse
Affiliation(s)
- Fabio Bernardoni
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Nadine Bernhardt
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Shakoor Pooseh
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Joseph A. King
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Daniel Geisler
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Franziska Ritschel
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Ilka Boehm
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Maria Seidel
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Veit Roessner
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Michael N. Smolka
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| | - Stefan Ehrlich
- From the Division of Psychological and Social Medicine and Developmental Neuroscience, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Bernardoni, King, Geisler, Ritschel, Boehm, Seidel, Ehrlich); the Department of Psychiatry and Psychotherapy, Technische Universität Dresden, Dresden, Germany (Bernhardt, Pooseh, Smolka); the Freiburg Center for Data Analysis and Modeling, Albert-Ludwigs-Universität Freiburg, Germany (Pooseh); and the Translational Developmental Neuroscience Section, Eating Disorder Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany (Roessner, Ehrlich)
| |
Collapse
|
269
|
Moose JE, Leets KA, Mate NA, Chisholm JD, Hougland JL. An overview of ghrelin O-acyltransferase inhibitors: a literature and patent review for 2010-2019. Expert Opin Ther Pat 2020; 30:581-593. [PMID: 32564644 DOI: 10.1080/13543776.2020.1776263] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The peptide hormone ghrelin regulates physiological processes associated with energy homeostasis such as appetite, insulin signaling, glucose metabolism, and adiposity. Ghrelin has also been implicated in a growing number of neurological pathways involved in stress response and addiction behavior. For ghrelin to bind the growth hormone secretagogue receptor 1a (GHS-R1a) and activate signaling, the hormone must first be octanoylated on a specific serine side chain. This key transformation is performed by the enzyme ghrelin O-acyltransferase (GOAT), and therefore GOAT inhibitors may be useful in treating disorders related to ghrelin signaling such as diabetes, obesity, and related metabolic syndromes. AREAS COVERED This report covers ghrelin and GOAT as potential therapeutic targets and summarizes work on GOAT inhibitors through the end of 2019, highlighting recent successes with both peptidomimetics and small molecule GOAT inhibitors as potent modulators of GOAT-catalyzed ghrelin octanoylation. EXPERT OPINION A growing body of biochemical and structural knowledge regarding the ghrelin/GOAT system now enables multiple avenues for identifying and optimizing GOAT inhibitors. We are at the beginning of a new era with increased opportunities for leveraging ghrelin and GOAT in the understanding and treatment of multiple health conditions including diabetes, obesity, and addiction.
Collapse
Affiliation(s)
- Jacob E Moose
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - Katelyn A Leets
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - Nilamber A Mate
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - John D Chisholm
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| | - James L Hougland
- Department of Chemistry and BioInspired Syracuse, Syracuse University , Syracuse, NY, USA
| |
Collapse
|
270
|
Ghrayeb H, Elias M, Nashashibi J, Youssef A, Manal M, Mahagna L, Refaat M, Schwartz N, Elias A. Appetite and ghrelin levels in iron deficiency anemia and the effect of parenteral iron therapy: A longitudinal study. PLoS One 2020; 15:e0234209. [PMID: 32497136 PMCID: PMC7272047 DOI: 10.1371/journal.pone.0234209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 05/20/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Iron deficiency anemia (IDA) is associated with decreased appetite. The ghrelin hormone is one of the major regulators of appetite. OBJECTIVES To evaluate appetite and ghrelin levels in patients with IDA, and to investigate the change in appetite and ghrelin following intravenous iron therapy. METHODS A total of 56 IDA patients and 51 controls were included in the study. Both appetite and ghrelin were assessed at baseline and following intravenous iron therapy. These were assessed at corresponding time intervals in the control group. Appetite was assessed by the SNAQ score (Simplified Nutritional Appetite Questionnaire) and fasting ghrelin levels were assessed by acylated ghrelin (AG), unacylated ghrelin (UAG) and their respective ratio AG/UAG. RESULTS IDA patients had significantly lower SNAQ scores, yet higher AG levels and higher AG/UAG ratios compared to healthy controls; the mean SNAQ scores were 12.56 ± 3.45 and 16.1 ± 2, respectively (P<0.01); the median AG levels were 57.5 pg/ml and 43 pg/ml respectively (P = 0.007); and the median AG/UAG ratios were 0.48 and 0.25 respectively (P = 0.04). On multivariate linear regression analysis, IDA remained independently associated with decreased SNAQ score (β = -0.524, P<0.001) and increased acylated ghrelin (β = 0.289, P = 0.013). After IDA was treated, SNAQ scores increased significantly by a mean of 2 points. AG and AG/UAG ratios decreased significantly by a mean of -18.44 pg/ml and -0.2 respectively. The control group showed no significant change in SNAQ scores or ghrelin at corresponding time intervals. CONCLUSIONS IDA patients have a reduced appetite and paradoxically elevated ghrelin hormone activity compared to healthy controls. Treating IDA enhances appetite and lowers ghrelin levels. Future studies are needed to explore the mechanism of this paradoxical ghrelin activity.
Collapse
Affiliation(s)
- Hanin Ghrayeb
- Department of Internal Medicine C, Emek Medical Center, Afula, Israel
- Rapaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| | - Mazen Elias
- Department of Internal Medicine C, Emek Medical Center, Afula, Israel
- Rapaport Faculty of Medicine, Technion Institute of Technology, Haifa, Israel
| | - Jeries Nashashibi
- Department of Internal Medicine D, Rambam Health Care Campus, Haifa, Israel
| | - Awni Youssef
- Bar-Ilan University Faculty of Medicine, Azrieli Faculty of medicine, Safed, Israel
| | - Mari Manal
- Nazareth Tower Out Patients Clinic Ambulatory, Clalit, Nazareth, Israel
| | - Liala Mahagna
- Endocrine laboratory, HaEmek Medical Center, Afula, Israel
| | - Masalha Refaat
- Endocrine laboratory, HaEmek Medical Center, Afula, Israel
| | - Naama Schwartz
- Clinical Research Unit, HaEmek Medical Center, Afula, Israel
| | - Adi Elias
- Department of Internal Medicine B, Rambam Health Care Campus, Haifa, Israel
- * E-mail:
| |
Collapse
|
271
|
Pietraszko W, Furgala A, Gorecka-Mazur A, Kwinta B, Kaszuba-Zwoinska J, Polak J, Fiszer U, Gil K, Krygowska-Wajs A. Assessments of plasma acyl-ghrelin levels in Parkinson's disease patients treated with deep brain stimulation. Peptides 2020; 128:170299. [PMID: 32305796 DOI: 10.1016/j.peptides.2020.170299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/26/2020] [Accepted: 03/12/2020] [Indexed: 12/26/2022]
Abstract
Gastrointestinal dysfunction is the most common non-motor symptom in Parkinson's disease (PD) with rates rising as the disease progresses. Deep brain stimulation of subthalamic nucleus (STN DBS) improves motor functions in advanced PD. However, the effect of STN DBS on ghrelin concentration and consequently on motility disturbances as well as body weight is unclear. The objective of this study was to assess acyl-ghrelin levels in comparison to weight in advanced PD patients treated with STN DBS. Plasma concentrations of acyl-ghrelin was measured in 29 PD patients in the fasting state and at 30, 60, 120, and 180 min after a standard meal preoperatively and 3 months after surgery. The level of acyl-ghrelin in PD patients were compared with 30 age and sex-matched healthy controls. We reported that mean plasma acyl-ghrelin levels were decreased in PD patients before STN DBS in fasting (p = 0.0003) and in 30 min postprandial phase (p = 0.04) compared with healthy controls. The plasma acyl-ghrelin levels after STN DBS increased in pre-prandial and postprandial phase in PD patients at the investigated time points. Body weight gained on average 2.33 kg during the first 3 months after surgery. There was no correlation between the acyl-ghrelin plasma levels and BMI. After STN DBS in fasting and postprandial phase plasma acyl-ghrelin levels were increased. The results showed that STN DBS therapy elicited a modification of ghrelin levels, increasing its concentration in pre- and postprandial state. In addition, body weight was increased during 3 months after surgery.
Collapse
Affiliation(s)
- Wojciech Pietraszko
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Agata Furgala
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Agnieszka Gorecka-Mazur
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Borys Kwinta
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Jolanta Kaszuba-Zwoinska
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Jaroslaw Polak
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Urszula Fiszer
- Department of Neurology and Epileptology, Centre of Postgraduate Medical Education, Warsaw, Czerniakowska 231, Poland
| | - Krzysztof Gil
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Anna Krygowska-Wajs
- Department of Neurology, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland.
| |
Collapse
|
272
|
Abstract
PURPOSE OF REVIEW There is substantial inter-individual variability in body weight change, which is not fully accounted by differences in daily energy intake and physical activity levels. The metabolic responses to short-term perturbations in energy intake can explain part of this variability by quantifying the degree of metabolic "thriftiness" that confers more susceptibility to weight gain and more resistance to weight loss. It is unclear which metabolic factors and pathways determine this human "thrifty" phenotype. This review will investigate and summarize emerging research in the field of energy metabolism and highlight important metabolic mechanisms implicated in body weight regulation in humans. RECENT FINDINGS Dysfunctional adipose tissue lipolysis, reduced brown adipose tissue activity, blunted fibroblast growth factor 21 secretion in response to low-protein hypercaloric diets, and impaired sympathetic nervous system activity might constitute important metabolic factors characterizing "thriftiness" and favoring weight gain in humans. The individual propensity to weight gain in the current obesogenic environment could be ascertained by measuring specific metabolic factors which might open up new pathways to prevent and treat human obesity.
Collapse
Affiliation(s)
- Tim Hollstein
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ, USA.
- Department of Information Engineering, University of Pisa, Pisa, Italy.
| |
Collapse
|
273
|
Brojeni MS, Nasseri F, Haghparast A, Eliassi A. Paraventricular nucleus-microinjected glucose increases food intake in 18 h food-deprived rats: A central regulatory mechanism on serum ghrelin and leptin levels. Eur J Pharmacol 2020; 876:173073. [DOI: 10.1016/j.ejphar.2020.173073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 03/13/2020] [Indexed: 01/29/2023]
|
274
|
Shinsyu A, Bamba S, Kurihara M, Matsumoto H, Sonoda A, Inatomi O, Andoh A, Takebayashi K, Kojima M, Iida H, Tani M, Sasaki M. Inflammatory cytokines, appetite-regulating hormones, and energy metabolism in patients with gastrointestinal cancer. Oncol Lett 2020; 20:1469-1479. [PMID: 32724390 DOI: 10.3892/ol.2020.11662] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 12/17/2019] [Indexed: 12/18/2022] Open
Abstract
This study investigated energy metabolism and its association with inflammatory cytokines and appetite- regulating hormones in patients with gastrointestinal cancer. Subjects were inpatients scheduled to undergo therapeutic intervention for diagnosed gastrointestinal cancer. Nutritional status on admission was assessed based on anthropometric measurements, nutrition screening results, food intake rate (energy intake/energy provided in hospital food), and biochemical test results. Fat-free mass (FFM) was measured using the bioelectrical impedance analysis. Resting energy expenditure (REE) and respiratory quotient were measured with indirect calorimetry, and basal energy expenditure (BEE) was calculated using the Harris-Benedict equation. A total 51 patients with gastrointestinal cancer were enrolled (17 with esophageal cancer, 15 with gastric cancer, and 19 with colorectal cancer); 16 had stage I disease, 11 had stage II, 13 had stage III, and 11 had stage IV. The levels of inflammatory cytokines such as interleukin (IL)-6 and tumor necrosis factor (TNF)-α increased significantly with cancer stage progression (P<0.001; Jonckheere-Terpstra trend test). The REE/body weight and the REE/FFM tended to increase with cancer stage progression (P=0.064 and P=0.053, respectively; Jonckheere-Terpstra trend test). FFM showed a significant negative correlation with the level of TNF-α (P=0.008; Spearman's correlation coefficient). Also, food intake rate showed a significant negative correlation with levels of IL-6 and TNF-α (P<0.001). The level of active ghrelin was positively correlated with that of IL-6 and energy metabolism (P=0.004 and 0.016, respectively) and negatively correlated with food intake rate (P=0.035), which suggests a state of ghrelin resistance. In conclusion, this study confirmed increases in the levels of inflammatory cytokines with the progression of gastrointestinal cancer and suggested the possible association of such increases with decreased FFM and the increased energy metabolism. However, the increased levels of active ghrelin failed to compensate for cachexia in cancer patients.
Collapse
Affiliation(s)
- Ayaka Shinsyu
- Division of Clinical Nutrition, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Shigeki Bamba
- Division of Clinical Nutrition, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Mika Kurihara
- Division of Clinical Nutrition, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Hiroshi Matsumoto
- Division of Gastroenterology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Ayano Sonoda
- Division of Gastroenterology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Osamu Inatomi
- Division of Gastroenterology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Akira Andoh
- Division of Gastroenterology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Katsushi Takebayashi
- Division of Gastrointestinal Surgery, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Masatsugu Kojima
- Division of Gastrointestinal Surgery, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Hiroya Iida
- Division of Gastrointestinal Surgery, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Masaji Tani
- Division of Gastrointestinal Surgery, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Masaya Sasaki
- Division of Clinical Nutrition, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| |
Collapse
|
275
|
Influence of obestatin on the histological development of the small intestine in piglets during the first week of postnatal life. Animal 2020; 14:2129-2137. [PMID: 32398171 DOI: 10.1017/s1751731120000919] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Obestatin is a gastrointestinal peptide having wide-ranging effects on cell proliferation; however, its mechanism of action remains poorly understood. Thus, the aim of the study was to elucidate the effect of exogenous obestatin on the postnatal structural development of the small intestine. Seven-day-old piglets with an average BW of 1.56 ± 0.23 kg were divided into four groups (n = 10) that received intragastrically obestatin (2, 10 or 15 μg/kg BW) or vehicle. After a 6-day experimental period, morphological analysis of gastrointestinal tract and small intestine wall (mitosis and apoptosis indexes, histomorphometry of mucosa and muscularis layers) was performed. The study revealed a seemingly incoherent pattern of the histological structure of the small intestine among the experimental groups, suggesting that the effect of obestatin is both intestinal segment specific and dose dependent. Histomorphometric analysis of the small intestine showed that higher doses of obestatin seem to promote the structural development of the duodenum while simultaneously hindering the maturation of more distal parts of the intestine. Intragastric administration of obestatin increased the crypt mitotic index in all segments of the small intestine with the strongest pro-mitotic activity following the administration of obestatin at a dose of 10 and 15 μg/kg BW. The significant differences in the number of apoptotic cells in the intestinal villi among the groups were observed only in proximal jejunum and ileum. In conclusion, it seems that obestatin shows a broad-spectrum of activity in the gastrointestinal tract of newborn piglets, being able to accelerate its structural development. However, the varied effect depending on the intestinal segment or the concentration of exogenous obestatin causes that further research is needed to clarify the exact mechanism of this phenomenon.
Collapse
|
276
|
Mani BK, Puzziferri N, He Z, Rodriguez JA, Osborne-Lawrence S, Metzger NP, Chhina N, Gaylinn B, Thorner MO, Thomas EL, Bell JD, Williams KW, Goldstone AP, Zigman JM. LEAP2 changes with body mass and food intake in humans and mice. J Clin Invest 2020; 129:3909-3923. [PMID: 31424424 DOI: 10.1172/jci125332] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 06/11/2019] [Indexed: 12/11/2022] Open
Abstract
Acyl-ghrelin administration increases food intake, body weight, and blood glucose. In contrast, mice lacking ghrelin or ghrelin receptors (GHSRs) exhibit life-threatening hypoglycemia during starvation-like conditions, but do not consistently exhibit overt metabolic phenotypes when given ad libitum food access. These results, and findings of ghrelin resistance in obese states, imply nutritional state dependence of ghrelin's metabolic actions. Here, we hypothesized that liver-enriched antimicrobial peptide-2 (LEAP2), a recently characterized endogenous GHSR antagonist, blunts ghrelin action during obese states and postprandially. To test this hypothesis, we determined changes in plasma LEAP2 and acyl-ghrelin due to fasting, eating, obesity, Roux-en-Y gastric bypass (RYGB), vertical sleeve gastrectomy (VSG), oral glucose administration, and type 1 diabetes mellitus (T1DM) using humans and/or mice. Our results suggest that plasma LEAP2 is regulated by metabolic status: its levels increased with body mass and blood glucose and decreased with fasting, RYGB, and in postprandial states following VSG. These changes were mostly opposite of those of acyl-ghrelin. Furthermore, using electrophysiology, we showed that LEAP2 both hyperpolarizes and prevents acyl-ghrelin from activating arcuate NPY neurons. We predict that the plasma LEAP2/acyl-ghrelin molar ratio may be a key determinant modulating acyl-ghrelin activity in response to body mass, feeding status, and blood glucose.
Collapse
Affiliation(s)
- Bharath K Mani
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Nancy Puzziferri
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas, USA.,Department of Surgery, Veterans Administration North Texas Heath Care System, Dallas, Texas, USA
| | | | - Juan A Rodriguez
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Sherri Osborne-Lawrence
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Nathan P Metzger
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| | - Navpreet Chhina
- PsychoNeuroEndocrinology Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, and.,Computational, Cognitive and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Bruce Gaylinn
- Department of Endocrinology, University of Virginia, Charlottesville, Virginia, USA
| | - Michael O Thorner
- Department of Endocrinology, University of Virginia, Charlottesville, Virginia, USA
| | - E Louise Thomas
- Research Centre for Optimal Health, University of Westminster, London, United Kingdom
| | - Jimmy D Bell
- Research Centre for Optimal Health, University of Westminster, London, United Kingdom
| | | | - Anthony P Goldstone
- PsychoNeuroEndocrinology Research Group, Neuropsychopharmacology Unit, Centre for Psychiatry, and.,Computational, Cognitive and Clinical Neuroimaging Laboratory, Division of Brain Sciences, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Jeffrey M Zigman
- Division of Hypothalamic Research.,Division of Endocrinology & Metabolism, Department of Internal Medicine.,Department of Psychiatry, and
| |
Collapse
|
277
|
Abstract
Ghrelin is a key signal driving energy seeking and storage in order to reverse energy deficit. In line with this view, the metabolic status of an organism predicts sensitivity to ghrelin, with fasting increasing and obesity decreasing ghrelin sensitivity. However, the mechanism responsible for controlling this sensitivity is unknown. In this issue of the JCI, Mani and colleagues show that plasma levels of plasma liver-enriched antimicrobial peptide-2 (LEAP2), a recently identified hormone that antagonizes the ghrelin receptor, are inversely correlated with those of plasma acyl-ghrelin under conditions of both energy deficit and energy surplus in mice and humans. Their results show that a fall in plasma LEAP2 during energy deficit facilitates the actions of acyl-ghrelin, whereas increased LEAP2 in obesity suppresses the actions of acyl-ghrelin. This important discovery helps reshape our understanding of ghrelin function and may provide a new approach to aiding weight maintenance after diet-induced weight loss.
Collapse
|
278
|
Zhou CX, Xie SC, Li MY, Huang CQ, Zhou HY, Cong H, Zhu XQ, Cong W. Analysis of the serum peptidome associated with Toxoplasma gondii infection. J Proteomics 2020; 222:103805. [PMID: 32387797 DOI: 10.1016/j.jprot.2020.103805] [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: 12/27/2019] [Revised: 04/26/2020] [Accepted: 05/01/2020] [Indexed: 01/27/2023]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that can cause severe disease in immunocompromised individuals and congenitally infected neonates. In order to determine whether serum peptide profile could reveal disease markers or allow determination of toxoplasmosis aggressiveness, mouse sera were collected from acutely infected, chronically infected and control subjects, and analyzed by a quantitative label-free pepdomics approach (LC-MS/MS). Six hundred and seven endogenous peptides were identified among all samples, with peptide profiling of difference that readily distinguished between acutely infected samples and other samples. Among these peptides detected in this study, 81 and 68 differentially expressed peptides (DEPs) were found in the acute and chronic infection stages, respectively. Through Gene Ontology analysis, most of the precursor proteins of these DEPs were associated with biological regulation and binding activity. These findings in this study will help in the search of peptide targets with a key role in disease diagnosis and create new opportunities for the development of better means for the prevention and control of toxoplasmosis. SIGNIFICANCE: Toxoplasma gondii is an unicellular parasite which infects humans and a wide range of warm-blooded animals. The serum peptidome contains a large set of low molecular weight endogenous peptides derived from secretion, protease activity and PTMs. In the present study we quantified the effects of T. gondii infection on the serum peptidome to identify novel disease regulated secretory factors. We developed an optimized label-free LC-MS/MS method to analyze endogenous peptides during toxoplasmosis progression. This resulted in quantification of 607 unique peptides at both acute and chronic infection stages. Collectively, our deep peptidomic analysis of serum revealed that peptide variations were affected by disease development, and peptidomics is an ideal method for quantifying changes in circulating factors on a global scale in response to pathophysiological perturbations such as T. gondii infection.
Collapse
Affiliation(s)
- Chun-Xue Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, PR China
| | - Shi-Chen Xie
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China
| | - Man-Yao Li
- Marine College, Shandong University, Weihai, Shandong Province 264209, PR China
| | - Cui-Qin Huang
- College of Life Sciences and Fujian Provincial Key Laboratory for the Prevention and Control of Animal Infectious Diseases and Biotechnology, Longyan University, Longyan, Fujian Province 364012, PR China
| | - Huai-Yu Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, PR China
| | - Hua Cong
- Department of Pathogen Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province 250012, PR China
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu Province 730046, PR China.
| | - Wei Cong
- Marine College, Shandong University, Weihai, Shandong Province 264209, PR China.
| |
Collapse
|
279
|
Aldawudi I, Katwal PC, Jirjees S, Htun ZM, Khan S. Future of Bariatric Embolization: A Review of Up-to-date Clinical Trials. Cureus 2020; 12:e7958. [PMID: 32509483 PMCID: PMC7270878 DOI: 10.7759/cureus.7958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 04/29/2020] [Indexed: 11/05/2022] Open
Abstract
Obesity is a significant health issue with an overall rise in mortality; it has multiple risk factors, including hormonal effects, which play a significant role in the balance of food intake and weight gain. Ghrelin is an anabolic hormone secreted from stomach fundus and plays a significant role in this regulation. Management of obesity involves multiple interventions, including lifestyle adjustment, pharmacotherapy, and bariatric surgery. Bariatric embolization is a relatively new procedure; several animal studies show that embolization of the left gastric artery reduces serum ghrelin and induces weight loss. Also, several clinical studies were conducted in the past ten years which have shown bariatric embolization's effectiveness in inducing weight loss: a meta-analysis of 47 patients included in six different clinical studies of left gastric artery embolization resulted in 8% total weight loss from baseline body weight. Many studies also show this procedure's effect on lowering the HgA1C level and lipid profile. Clinical studies mostly reported minor adverse effects such as transient abdominal discomfort, nausea and vomiting, gastric ulcers, and major adverse effects were uncommon, suggesting the procedure is well tolerated. It may be an alternative line of management in patients who are not suitable candidates for bariatric surgery. Although future clinical studies will provide an answer to several questions like the exact effects of the procedure on diabetes and metabolic syndrome, future studies are also needed to establish particular guidelines to match different patient characteristics with their optimal procedural techniques and pre- and post-procedure evaluation tests.
Collapse
Affiliation(s)
- Israa Aldawudi
- Radiology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Prakash C Katwal
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Srood Jirjees
- Neurology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Zin Mar Htun
- Internal Medicine, California Institute of Behavioral Neuroscience and Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
280
|
Sovetkina A, Nadir R, Fung JNM, Nadjarpour A, Beddoe B. The Physiological Role of Ghrelin in the Regulation of Energy and Glucose Homeostasis. Cureus 2020; 12:e7941. [PMID: 32499981 PMCID: PMC7266561 DOI: 10.7759/cureus.7941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Ghrelin is a peptide hormone that is primarily released from the stomach. It is best known for its role in appetite initiation. However, evidence also suggests that ghrelin may play a much wider role in energy homeostasis and glucose metabolism. It is known that exogenous ghrelin exerts an orexigenic signal via growth hormone secretagogue receptors in the arcuate nucleus of the hypothalamus. However, blocking ghrelin signalling in the arcuate nucleus does not decrease feeding. Evidence now proposes that an alternative pathway for ghrelin’s action is via the vagus nerve. Furthermore, it has been suggested that ghrelin signalling is an important physiological regulator of body adiposity and energy storage. Ghrelin also seems to be important in controlling glucose metabolism through action in the pancreatic islets of Langerhans, representing a promising novel therapeutic target in diabetes treatment. Despite these findings, further research in humans is required before ghrelin can be indicated as a therapeutic target in obesity or diabetes. This review summarises the current literature concerning ghrelin’s physiological roles in energy and glucose homeostasis.
Collapse
Affiliation(s)
| | - Rans Nadir
- Faculty of Medicine, Imperial College London, London, GBR
| | | | | | | |
Collapse
|
281
|
Nagoya T, Kamimura K, Inoue R, Ko M, Owaki T, Niwa Y, Sakai N, Setsu T, Sakamaki A, Yokoo T, Kamimura H, Nakamura Y, Ueno M, Terai S. Ghrelin-insulin-like growth factor-1 axis is activated via autonomic neural circuits in the non-alcoholic fatty liver disease. Neurogastroenterol Motil 2020; 32:e13799. [PMID: 31984635 DOI: 10.1111/nmo.13799] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/23/2019] [Accepted: 12/23/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND The correlation of the growth hormone (GH) and insulin-like growth factor-1 (IGF-1) with non-alcoholic fatty liver disease (NAFLD) has been reported in epidemiological studies. However, the mechanisms of molecular and inter-organ systems that render these factors to influence on NAFLD have not been elucidated. In this study, we examined the induction of ghrelin which is the GH-releasing hormone and IGF-1, and involvement of autonomic neural circuits, in the pathogenesis of NAFLD. METHODS The expression of gastric and hypothalamic ghrelin, neural activation in the brain, and serum IGF-1 were examined in NAFLD models of choline-deficient defined l-amino-acid diet-fed, melanocortin 4 receptor knockout mice, and partial hepatectomy mice with or without the blockades of autonomic nerves to test the contribution of neural circuits connecting the brain, liver, and stomach. KEY RESULTS The fatty changes in the liver increased the expression of gastric ghrelin through the autonomic pathways which sends the neural signals to the arcuate nucleus in the hypothalamus through the afferent vagal nerve which reached the pituitary gland to release GH and then stimulate the IGF-1 release from the liver. In addition, high levels of ghrelin expression in the arcuate nucleus were correlated with NAFLD progression regardless of the circuits. CONCLUSIONS Our study demonstrated that the fatty liver stimulates the autonomic nervous signal circuits which suppress the progression of the disease by activating the gastric ghrelin expression, the neural signal transduction in the brain, and the release of IGF-1 from the liver.
Collapse
Affiliation(s)
- Takuro Nagoya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Ryosuke Inoue
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masayoshi Ko
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takashi Owaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yusuke Niwa
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Norihiro Sakai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Toru Setsu
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Akira Sakamaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hiroteru Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| |
Collapse
|
282
|
Fernandez DC, Komal R, Langel J, Ma J, Duy PQ, Penzo MA, Zhao H, Hattar S. Retinal innervation tunes circuits that drive nonphotic entrainment to food. Nature 2020; 581:194-198. [PMID: 32404998 PMCID: PMC7291822 DOI: 10.1038/s41586-020-2204-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 02/21/2020] [Indexed: 12/14/2022]
Abstract
Daily changes in light and food availability are major time cues that influence circadian timing1. However, little is known about the circuits that integrate these time cues to drive a coherent circadian output1-3. Here we investigate whether retinal inputs modulate entrainment to nonphotic cues such as time-restricted feeding. Photic information is relayed to the suprachiasmatic nucleus (SCN)-the central circadian pacemaker-and the intergeniculate leaflet (IGL) through intrinsically photosensitive retinal ganglion cells (ipRGCs)4. We show that adult mice that lack ipRGCs from the early postnatal stages have impaired entrainment to time-restricted feeding, whereas ablation of ipRGCs at later stages had no effect. Innervation of ipRGCs at early postnatal stages influences IGL neurons that express neuropeptide Y (NPY) (hereafter, IGLNPY neurons), guiding the assembly of a functional IGLNPY-SCN circuit. Moreover, silencing IGLNPY neurons in adult mice mimicked the deficits that were induced by ablation of ipRGCs in the early postnatal stages, and acute inhibition of IGLNPY terminals in the SCN decreased food-anticipatory activity. Thus, innervation of ipRGCs in the early postnatal period tunes the IGLNPY-SCN circuit to allow entrainment to time-restricted feeding.
Collapse
Affiliation(s)
- Diego Carlos Fernandez
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Ruchi Komal
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jennifer Langel
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jun Ma
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Phan Q Duy
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA.,MSTP, Yale University, New Haven, CT, USA
| | - Mario A Penzo
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Haiqing Zhao
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Samer Hattar
- National Institute of Mental Health (NIMH), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
283
|
Fibrillin-1 and fibrillin-1-derived asprosin in adipose tissue function and metabolic disorders. J Cell Commun Signal 2020; 14:159-173. [PMID: 32279186 DOI: 10.1007/s12079-020-00566-3] [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: 01/23/2020] [Revised: 03/28/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022] Open
Abstract
The extracellular matrix microenvironment of adipose tissue is of critical importance for the differentiation, remodeling and function of adipocytes. Fibrillin-1 is one of the main components of microfibrils and a key player in this process. Furin processing of profibrillin-1 results in mature fibrillin-1 and releases the C-terminal propeptide as a circulating hunger hormone, asprosin. Mutations in the fibrillin-1 gene lead to adipose tissue dysfunction and causes Marfan syndrome, marfanoid progeroid lipodystrophy syndrome, and neonatal progeroid syndrome. Increased TGF-β signaling, altered mechanical properties and impaired adipogenesis are potential causes of adipose tissue dysfunction, mediated through deficient microfibrils. Circulating asprosin on the other hand is secreted primarily by white adipose tissue under fasting conditions and in obesity. It increases hepatic glucose production and drives insulin secretion and appetite stimulation through inter-organ cross talk. This review discusses the metabolic consequences of fibrillin-1 and fibrillin-1-derived asprosin in pathological conditions. Understanding the dynamic role of fibrillin-1 in the adipose tissue milieu and of circulating asprosin in the body can provide novel mechanistic insights into how fibrillin-1 may contribute to metabolic syndrome. This could lead to new management regimens of patients with metabolic disease.
Collapse
|
284
|
Hoffmann JG, Xie W, Chopra AR. Energy Regulation Mechanism and Therapeutic Potential of Asprosin. Diabetes 2020; 69:559-566. [PMID: 32198197 PMCID: PMC7085243 DOI: 10.2337/dbi19-0009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
Genetic studies of patients with neonatal progeroid syndrome led to the discovery of the novel fasting-induced, glucogenic, and orexigenic hormone named asprosin, the C-terminal cleavage product of profibrillin. Upon secretion, asprosin travels to the liver, where it exerts a glucogenic effect through OR4M1, an olfactory G-protein-coupled receptor. It also crosses the blood-brain barrier to stimulate appetite-modulating neurons in the arcuate nucleus of the hypothalamus, exerting an orexigenic effect via an as yet unidentified receptor. Specifically, it stimulates appetite by activating orexigenic AgRP neurons and inhibiting anorexigenic POMC neurons. Studies have also focused on the therapeutic potential of inhibiting asprosin for treatment of obesity and type 2 diabetes, both of which are characterized by high levels of circulating asprosin. It has been shown that anti-asprosin monoclonal antibodies reduce blood glucose, appetite, and body weight, validating asprosin as a therapeutic target. Current work aims to uncover key features of the asprosin biology such as the identification of its neuronal receptor, identification of the secretion mechanism from adipose tissue, and development of anti-asprosin monoclonal antibodies as diabetes and obesity therapies.
Collapse
Affiliation(s)
| | - Wei Xie
- Harrington Discovery Institute at University Hospitals, Cleveland, OH
| | - Atul R Chopra
- Harrington Discovery Institute at University Hospitals, Cleveland, OH
- Department of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH
| |
Collapse
|
285
|
Hernández Morante JJ, Díaz Soler I, Muñoz JSG, Sánchez HP, Barberá Ortega MDC, Martínez CM, Morillas Ruiz JM. Moderate Weight Loss Modifies Leptin and Ghrelin Synthesis Rhythms but Not the Subjective Sensations of Appetite in Obesity Patients. Nutrients 2020; 12:E916. [PMID: 32230732 PMCID: PMC7230904 DOI: 10.3390/nu12040916] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022] Open
Abstract
Obesity is characterized by a resistance to appetite-regulating hormones, leading to a misalignment between the physiological signals and the perceived hunger/satiety signal. A disruption of the synthesis rhythm may explain this situation. The aim of this study was to evaluate the effect of dietary-induced weight loss on the daily rhythms of leptin and ghrelin and its influence on the daily variability of the appetite sensations of patients with obesity. Twenty subjects with obesity underwent a hypocaloric dietary intervention for 12 weeks. Plasma leptin and ghrelin were analyzed at baseline and at the end of the intervention and in 13 normal-weight controls. Appetite ratings were analyzed. Weight loss decreased leptin synthesis (pauc < 0.001) but not the rhythm characteristics, except the mean variability value (pmesor = 0.020). By contrast, the mean ghrelin level increased after weight loss. The rhythm characteristics were also modified until a rhythm similar to the normal-weight subjects was reached. The amount of variability of leptin and ghrelin was correlated with the effectiveness of the dietary intervention (p < 0.020 and p < 0.001, respectively). Losing weight partially restores the daily rhythms of leptin and modifies the ghrelin rhythms, but appetite sensations are barely modified, thus confirming that these hormones cannot exercise their physiological function properly.
Collapse
Affiliation(s)
- Juan José Hernández Morante
- Eating Disorders Research Unit, Catholic University of Murcia (UCAM), 30107 Murcia, Spain; (I.D.S.); (M.d.C.B.O.)
| | - Inmaculada Díaz Soler
- Eating Disorders Research Unit, Catholic University of Murcia (UCAM), 30107 Murcia, Spain; (I.D.S.); (M.d.C.B.O.)
| | | | - Horacio Pérez Sánchez
- Bioinformatics and High Performance Computing Research Group (BIO-HPC), Computer Engineering Department, Catholic University of Murcia (UCAM), 30107 Murcia, Spain;
| | - Mª del Carmen Barberá Ortega
- Eating Disorders Research Unit, Catholic University of Murcia (UCAM), 30107 Murcia, Spain; (I.D.S.); (M.d.C.B.O.)
| | | | - Juana Mª Morillas Ruiz
- Food Technology & Nutrition Dept., Catholic University of Murcia (UCAM), 30107 Murcia, Spain;
| |
Collapse
|
286
|
Estradiol Replacement Improves High-Fat Diet-Induced Obesity by Suppressing the Action of Ghrelin in Ovariectomized Rats. Nutrients 2020; 12:nu12040907. [PMID: 32224927 PMCID: PMC7230822 DOI: 10.3390/nu12040907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 12/16/2022] Open
Abstract
This study aims to investigate the effects of estradiol replacement on the orexigenic action of ghrelin in ovariectomized (OVX) obese rats fed with a high-fat diet (HFD). Four weeks after OVX at 9 weeks of age, Wistar rats were subcutaneously implanted with either 17β-estradiol (E2) or placebo (Pla) pellets and started on HFD feeding. After 4 weeks, growth hormone-releasing peptide (GHRP)-6, a growth hormone secretagogue receptor (GHSR) agonist injected intraperitoneally, induced changes in HFD intake, and c-Fos-positive neurons in the hypothalamic arcuate nucleus (ARC) were measured in both groups. The ghrelin protein and mRNA levels, as well as GHSR protein in stomach, were analyzed by Western blotting and real-time PCR. HFD increased energy intake and body weight in the Pla group, while it temporarily reduced these in the E2 group. GHRP-6 enhanced HFD intake and activated neurons in the ARC only in the Pla group. Furthermore, gastric ghrelin and GHSR protein levels were lower in the E2 group than in the Pla group, but plasma acyl ghrelin levels were similar in both groups. Our results suggest that E2 replacement improves obesity by inhibiting the orexigenic action of ghrelin via downregulation of ghrelin and its receptor in stomach in HFD-fed OVX rats.
Collapse
|
287
|
Ghrelin infused into the dorsomedial hypothalamus of male mice increases food intake and adiposity. Physiol Behav 2020; 220:112882. [PMID: 32205145 DOI: 10.1016/j.physbeh.2020.112882] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/14/2020] [Accepted: 03/16/2020] [Indexed: 02/07/2023]
Abstract
Ghrelin is a 28 amino acid peptide hormone that targets the brain to promote feeding and adiposity. The ghrelin receptor, the GHSR1a, is expressed within most hypothalamic nuclei, including the DMH, but the role of GHSR1a in this region on energy balance is unknown. In order to investigate whether GHSR1a within the DMH modulate energy balance, we implanted osmotic minipumps filled with saline, ghrelin, or the GHSR1a antagonist JMV2959, and connected it to a cannula aimed unilaterally at the DMH of adult male C57BLJ6 mice and assessed their metabolic profile. We found that chronic infusion of ghrelin in the DMH promoted an increase in caloric intake as well as a decrease in energy expenditure. This translated to an overall increase in weight gain, primarily in the form of adipose tissue in ghrelin treated animals. Further, chronic ghrelin unilateral infusion into the DMH slowed glucose clearance. These results suggest that GHSR in the DMH significantly contribute to the metabolic effects produced by ghrelin.
Collapse
|
288
|
Hay RE, Edwards A, Klein M, Hyland L, MacDonald D, Karatsoreos I, Hill MN, Abizaid A. Ghrelin Receptor Signaling Is Not Required for Glucocorticoid-Induced Obesity in Male Mice. Endocrinology 2020; 161:5636885. [PMID: 31748785 PMCID: PMC7445420 DOI: 10.1210/endocr/bqz023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/19/2019] [Indexed: 12/11/2022]
Abstract
Chronically elevated levels of glucocorticoids increase food intake, weight gain, and adiposity. Similarly, ghrelin, a gut-secreted hormone, is also associated with weight gain, adiposity, and increased feeding. Here we sought to determine if corticosterone-induced metabolic and behavioral changes require functional ghrelin receptors (GHSR). To do this, we treated male C57BL mice with chronic corticosterone (CORT) mixed in their drinking water for 28 days. Half of these mice received the GHSR antagonist JMV2959 via osmotic minipumps while treated with CORT. In a second experiment, we gave the same CORT protocol to mice with a targeted mutation to the GHSR or their wild-type littermates. As expected, CORT treatment increased food intake, weight gain, and adiposity, but contrary to expectations, mice treated with a GHSR receptor antagonist or GHSR knockout (KO) mice did not show attenuated food intake, weight gain, or adiposity in response to CORT. Similarly, the effects of CORT on the liver were the same or more pronounced in GHSR antagonist-treated and GHSR KO mice. Treatment with JMV2959 did attenuate the effects of chronic CORT on glycemic regulation as determined by the glucose tolerance test. Finally, disruption of GHSR signaling resulted in behavioral responses associated with social withdrawal, potentially due to neuroprotective effects of GHSR activation. In all, we propose that blocking GHSR signaling helps to moderate glucose concentrations when CORT levels are high, but blocking GHSR signaling does not prevent increased food intake, weight gain, or increased adiposity produced by chronic CORT.
Collapse
Affiliation(s)
- Rebecca E Hay
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Alex Edwards
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Marianne Klein
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Lindsay Hyland
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - David MacDonald
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - Ilia Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, US
| | - Matthew N Hill
- Hotchkiss Brain Institute, Department of Cell Biology and Anatomy, University of Calgary, Calgary, AB, Canada
| | - Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
- Correspondence: Alfonso Abizaid, Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S5B6, Canada. E-mail:
| |
Collapse
|
289
|
Miyano K, Ohshima K, Suzuki N, Furuya S, Yoshida Y, Nonaka M, Higami Y, Yoshizawa K, Fujii H, Uezono Y. Japanese Herbal Medicine Ninjinyoeito Mediates Its Orexigenic Properties Partially by Activating Orexin 1 Receptors. Front Nutr 2020; 7:5. [PMID: 32175325 PMCID: PMC7056666 DOI: 10.3389/fnut.2020.00005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/10/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer cachexia is highly prevalent in patients with progressive cancer and is characterized by decreased food consumption, and body weight. Japanese herbal medicine Ninjinyoeito (NYT), composed of 12 herbal crude drugs, is prescribed in Asian countries to improve several symptoms such as anorexia and fatigue, which are commonly observed in patients with cancer cachexia. However, the action mechanisms of NYT in improving anorexia or fatigue in patients with cancer are not clear. Therefore, in the present study, we examined the effects of NYT on the activities of several G-protein-coupled receptors (GPCRs), which activate hyperphagia signaling in the central nervous system, using an in vitro assay with the CellKey™ system, which detects the activation of GPCRs as a change in intracellular impedance (ΔZ). NYT increased the ΔZ of human embryonic kidney 293 (HEK293) cells expressing orexin 1 receptor (OX1R) and those expressing neuropeptide Y1 receptor (NPY1R) in a dose-dependent manner. On the contrary, NYT did not significantly increase the ΔZ of HEK293A cells expressing growth hormone secretagogue receptor (GHSR) and those expressing NPY5R. The selective OX1R antagonist SB674042 significantly decreased the NYT-induced increase in ΔZ in OX1R-expressing cells. Contrarily, the selective NPY1R antagonist BIBO3340 failed to inhibit the NPY-induced increase in ΔZ in NPY1R-expressing cells. Additionally, we prepared modified NYT excluding each one of the 12 herbal crude drugs in NYT and investigated the effects on the activity of OX1R. Among the 12 modified NYT formulations, the one without citrus unshiu peel failed to activate OX1R. A screening of each of the 12 herbal crude drugs showed that citrus unshiu peel significantly activated OX1R, which was significantly suppressed by SB674042. These finding suggest that NYT and citrus unshiu peel could increase food intake via activation of orexigenic OX1R-expressing neurons in the hypothalamus. This study provides scientific evidence to support the potential of NYT for cancer patients with anorexia.
Collapse
Affiliation(s)
- Kanako Miyano
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kaori Ohshima
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan.,Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Nozomi Suzuki
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan.,Department of Medicinal Chemistry, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Saho Furuya
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan.,Department of Medicinal Chemistry, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Yuki Yoshida
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan.,Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Miki Nonaka
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Kazumi Yoshizawa
- Laboratory of Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Hideaki Fujii
- Department of Medicinal Chemistry, School of Pharmacy, Kitasato University, Tokyo, Japan
| | - Yasuhito Uezono
- Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan.,Division of Supportive Care Research, National Cancer Center Exploratory Oncology Research and Clinical Trial Center, Tokyo, Japan.,Innovation Center for Supportive, Palliative and Psychosocial Care, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
290
|
Abstract
MODY (Maturity Onset Diabetes of the Young) is a type of diabetes resulting from a pathogenic effect of gene mutations. Up to date, 13 MODY genes are known. Gene HNF1A is one of the most common causes of MODY diabetes (HNF1A-MODY; MODY3). This gene is polymorphic and more than 1200 pathogenic and non-pathogenic HNF1A variants were described in its UTRs, exons and introns. For HNF1A-MODY, not just gene but also phenotype heterogeneity is typical. Although there are some clinical instructions, HNF1A-MODY patients often do not meet every diagnostic criteria or they are still misdiagnosed as type 1 and type 2 diabetics. There is a constant effort to find suitable biomarkers to help with in distinguishing of MODY3 from Type 1 Diabetes (T1D) and Type 2 Diabetes (T2D). DNA sequencing is still necessary for unambiguous confirmation of clinical suspicion of MODY. NGS (Next Generation Sequencing) methods brought discoveries of multiple new gene variants and new instructions for their pathogenicity classification were required. The most actual problem is classification of variants with uncertain significance (VUS) which is a stumbling-block for clinical interpretation. Since MODY is a hereditary disease, DNA analysis of family members is helpful or even crucial. This review is updated summary about HNF1A-MODY genetics, pathophysiology, clinics functional studies and variant classification.
Collapse
|
291
|
Wang Q, Liszt KI, Depoortere I. Extra-oral bitter taste receptors: New targets against obesity? Peptides 2020; 127:170284. [PMID: 32092303 DOI: 10.1016/j.peptides.2020.170284] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/10/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022]
Abstract
Taste perception on the tongue is essential to help us to identify nutritious or potential toxic food substances. Emerging evidence has demonstrated the expression and function of bitter taste receptors (TAS2Rs) in a wide range of extra-oral tissues. In particular, TAS2Rs in gastrointestinal enteroendocrine cells control the secretion of appetite regulating gut hormones and influence hunger and food intake. Furthermore, these effects may be reinforced by the presence of TAS2Rs on intestinal smooth muscle cells, adipocytes and the brain. This review summarises how activation of extra-oral TAS2Rs can influence appetite and body weight control and how obesity impacts the expression and function of TAS2Rs. Region-selective targeting of bitter taste receptors may be promising targets for the treatment of obesity.
Collapse
Affiliation(s)
- Qiaoling Wang
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Kathrin I Liszt
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium.
| |
Collapse
|
292
|
Singh R, Bansal Y, Sodhi RK, Singh DP, Bishnoi M, Kondepudi KK, Medhi B, Kuhad A. Berberine attenuated olanzapine-induced metabolic alterations in mice: Targeting transient receptor potential vanilloid type 1 and 3 channels. Life Sci 2020; 247:117442. [PMID: 32081663 DOI: 10.1016/j.lfs.2020.117442] [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: 12/26/2019] [Accepted: 02/14/2020] [Indexed: 02/08/2023]
Abstract
Transient receptor potential vanilloid type 1 (TRPV1) channels are emerging therapeutic targets for metabolic disorders. Berberine, which is a modulator of TRPV1, has proven antiobesity and antidiabetic potentials. The present study was aimed to investigate the protective effects of berberine in olanzapine-induced alterations in hypothalamic appetite control, inflammation and metabolic aberrations in mice targeting TRPV1 channels. Female BALB/c mice (18-23 g) were treated with olanzapine (6 mg/kg, p.o.) for six weeks to induce metabolic alterations, while berberine (100 and 200 mg/kg, p.o.) and metformin (100 mg/kg, p.o) were used as test and standard interventions respectively. Weekly assessment of feed-water intake, body temperature and body weight was done, while locomotion was measured at the end of week 1 and 6. Serum glucose and lipid profile were assessed by biochemical methods, while other serum biomarkers were assessed by ELISA. qPCR was used to quantify the mRNA expression in the hypothalamus. Olanzapine treatment significantly increased the feed intake, weight gain, adiposity index, while reduced body temperature and locomotor activity which were reversed by berberine treatment. Berberine treatment reduced serum ghrelin and leptin levels as well decrease in hypothalamic mRNA expression of orexigenic neuropeptides, inflammatory markers and ghrelin receptor in olanzapine-treated mice. Olanzapine treatment increased expression of TRPV1/TRPV3 in the hypothalamus which was significantly decreased by berberine treatment. Our results suggest that berberine, by TRPV1/TRPV3 modulation, attenuated the olanzapine-induced metabolic alterations in mice. Hence berberine supplementation in psychiatric patients could be a preventive approach to reduce the metabolic adverse effects of antipsychotics.
Collapse
Affiliation(s)
- Raghunath Singh
- Pharmacology Research Lab, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Yashika Bansal
- Pharmacology Research Lab, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Rupinder Kaur Sodhi
- Pharmacology Research Lab, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Dhirendra Pratap Singh
- Pharmacology Research Lab, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India; Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS Nagar, Punjab 140306, India; ICMR-National Institute of Occupational Health (NIOH), Ahmedabad 380016, India
| | - Mahendra Bishnoi
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS Nagar, Punjab 140306, India
| | - Kanthi Kiran Kondepudi
- Food and Nutritional Biotechnology Division, National Agri-Food Biotechnology Institute (NABI), SAS Nagar, Punjab 140306, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India
| | - Anurag Kuhad
- Pharmacology Research Lab, University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India.
| |
Collapse
|
293
|
Behavioural characterization of ghrelin ligands, anamorelin and HM01: Appetite and reward-motivated effects in rodents. Neuropharmacology 2020; 168:108011. [PMID: 32067989 DOI: 10.1016/j.neuropharm.2020.108011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 02/12/2020] [Accepted: 02/13/2020] [Indexed: 12/14/2022]
Abstract
The ghrelinergic system has been steadily investigated as a therapeutic target in the treatment of metabolic disorders and modulation of appetite. While endogenous ghrelin activates the full complement of the growth hormone secretagogue receptor (GHSR-1a) pathways, synthetic GHSR-1a ligands display biased signalling and functional selectivity, which have a significant impact on the intended and indeed, unintended, therapeutic effects. The widespread expression of the GHSR-1a receptor in vivo also necessitates an imperative consideration of the biodistribution of GHSR-1a ligands. Here, we investigate anamorelin and HM01, two recently described synthetic GHSR-1a ligands which have shown promising effects on food intake in preclinical and clinical studies. We compare the downstream signalling pathways in cellular in vitro assays, including calcium mobilization, IP-one, internalization and β-arrestin recruitment assays. We describe a novel divergent activation of central reward circuitry by anamorelin and HM01 using c-Fos immunostaining as well as behavioural effects in food intake and reward paradigms. Interestingly, we found a paradoxical reduction in reward-related behaviour for anamorelin and HM01 treated animals in our chosen paradigms. The work highlights the critical importance to consider signalling bias in relation to future ghrelin-based therapies. In addition, central access of GHSR-1a ligands, particularly to reward areas of the brain, remains a crucial factor in eliciting potent appetite-stimulating effects. The precise characterization of downstream ghrelinergic signalling and biodistribution of novel GHSR-1a ligands will be decisive in their successful development and will allow predictive modelling and design of future synthetic ligands to combat metabolic and appetite disorders involving the ghrelinergic system. This article is part of the special issue on 'Neuropeptides'.
Collapse
|
294
|
Aklan I, Sayar Atasoy N, Yavuz Y, Ates T, Coban I, Koksalar F, Filiz G, Topcu IC, Oncul M, Dilsiz P, Cebecioglu U, Alp MI, Yilmaz B, Davis DR, Hajdukiewicz K, Saito K, Konopka W, Cui H, Atasoy D. NTS Catecholamine Neurons Mediate Hypoglycemic Hunger via Medial Hypothalamic Feeding Pathways. Cell Metab 2020; 31:313-326.e5. [PMID: 31839488 PMCID: PMC9017597 DOI: 10.1016/j.cmet.2019.11.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 09/22/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023]
Abstract
Glucose is the essential energy source for the brain, whose deficit, triggered by energy deprivation or therapeutic agents, can be fatal. Increased appetite is the key behavioral defense against hypoglycemia; however, the central pathways involved are not well understood. Here, we describe a glucoprivic feeding pathway by tyrosine hydroxylase (TH)-expressing neurons from nucleus of solitary tract (NTS), which project densely to the hypothalamus and elicit feeding through bidirectional adrenergic modulation of agouti-related peptide (AgRP)- and proopiomelanocortin (POMC)-expressing neurons. Acute chemogenetic inhibition of arcuate nucleus (ARC)-projecting NTSTH neurons or their target, AgRP neurons, impaired glucoprivic feeding induced by 2-Deoxy-D-glucose (2DG) injection. Neuroanatomical tracing results suggested that ARC-projecting orexigenic NTSTH neurons are largely distinct from neighboring catecholamine neurons projecting to parabrachial nucleus (PBN) that promotes satiety. Collectively, we describe a circuit organization in which an ascending pathway from brainstem stimulates appetite through key hunger neurons in the hypothalamus in response to hypoglycemia.
Collapse
Affiliation(s)
- Iltan Aklan
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Nilufer Sayar Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Yavuz Yavuz
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA; Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Tayfun Ates
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Ilknur Coban
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Fulya Koksalar
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Gizem Filiz
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Iskalen Cansu Topcu
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Merve Oncul
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Pelin Dilsiz
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Utku Cebecioglu
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Muhammed Ikbal Alp
- Department of Physiology, School of Medicine, Regenerative and Restorative Medical Research Center (REMER), Istanbul Medipol University, Istanbul, Turkey
| | - Bayram Yilmaz
- Department of Physiology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Deborah R Davis
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Karolina Hajdukiewicz
- Laboratory of Animal Models, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Kenji Saito
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Witold Konopka
- Laboratory of Animal Models, Neurobiology Center, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Huxing Cui
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Deniz Atasoy
- Department of Neuroscience and Pharmacology, Roy J. and Lucille A. Carver College of Medicine, Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA.
| |
Collapse
|
295
|
Koubaa-Ghorbel F, Chaâbane M, Turki M, Makni-Ayadi F, El Feki A. The protective effects of Salvia officinalis essential oil compared to simvastatin against hyperlipidemia, liver, and kidney injuries in mice submitted to a high-fat diet. J Food Biochem 2020; 44:e13160. [PMID: 32010989 DOI: 10.1111/jfbc.13160] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 01/12/2020] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
The present study was undertaken to evaluate the effects of Salvia officinalis essential oil (SEO) and simvastatin in hyperlipidemic mice. Animals were randomly divided into four groups. The control group received a standard diet. The high-fat diet (HFD) group received HFD. The third and fourth groups received HFD associated either with simvastatin (2.5 mg/kg bw) or with SEO (4 mg/kg bw). All animals were sacrificed after 8 weeks of treatment. SEO and simvastatin reduced in HFD mice body weight gain, hyperlipidemia, disruption of liver and renal functions and reactive oxygen species production. In fact, total cholesterol, triglycerides, total lipids, and low-density lipoprotein cholesterol levels, as well as aspartate transaminase, alanine transaminase, gamma-glutamyltranspeptidase and lactate dehydrogenase activities were reduced, while fecal lipids increased compared to those of HFD mice. The lipid-lowering effect of SEO was more effective than that of simvastatin. PRACTICAL APPLICATIONS: High-fat diet provokes hyperlipidemia, atherosclerosis, and abnormal lipid metabolism leading to the development of hepatic and renal dysfunctions as well as perturbations of the antioxidant status in liver and kidney. The results of this research highlight the beneficial effects of SEO in the management of these disorders without inducing side effects; in fact, the plant essential oil decreased lipids and improved the antioxidant status more than did a synthetic drug.
Collapse
Affiliation(s)
- Fatma Koubaa-Ghorbel
- Animal Ecophysiology Laboratory, Sciences Faculty, University of Sfax, Sfax, Tunisia
| | - Mariem Chaâbane
- Unit of Enzymes and Bioconversion, National Engineering School, University of Sfax, Sfax, Tunisia
| | - Mouna Turki
- Biochemistry Laboratory, CHU H. Bourguiba, University of Sfax, Sfax, Tunisia
| | - Fatma Makni-Ayadi
- Biochemistry Laboratory, CHU H. Bourguiba, University of Sfax, Sfax, Tunisia
| | - Abdelfattah El Feki
- Animal Ecophysiology Laboratory, Sciences Faculty, University of Sfax, Sfax, Tunisia
| |
Collapse
|
296
|
Abizaid A, Hougland JL. Ghrelin Signaling: GOAT and GHS-R1a Take a LEAP in Complexity. Trends Endocrinol Metab 2020; 31:107-117. [PMID: 31636018 PMCID: PMC7299083 DOI: 10.1016/j.tem.2019.09.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 09/25/2019] [Accepted: 09/27/2019] [Indexed: 12/30/2022]
Abstract
Ghrelin and the growth hormone secretagogue receptor 1a (GHS-R1a) are important targets for disorders related to energy balance and metabolic regulation. Pharmacological control of ghrelin signaling is a promising avenue to address health issues involving appetite, weight gain, obesity, and related metabolic disorders, and may be an option for patients suffering from wasting conditions like cachexia. In this review, we summarize recent developments in the biochemistry of ghrelin and GHS-R1a signaling. These include unravelling the enzymatic transformations that generate active ghrelin and the discovery of multiple proteins that interact with ghrelin and GHS-R1a to regulate signaling. Furthermore, we propose that harnessing these processes will lead to highly selective treatments to address obesity, diabetes, and other metabolism-linked disorders.
Collapse
Affiliation(s)
- Alfonso Abizaid
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | - James L Hougland
- Department of Chemistry, Syracuse University, Syracuse, NY, USA.
| |
Collapse
|
297
|
Tajiri E, Yoshimura E, Hatamoto Y, Shiratsuchi H, Tanaka S, Shimoda S. Acute Sleep Curtailment Increases Sweet Taste Preference, Appetite and Food Intake in Healthy Young Adults: A Randomized Crossover Trial. Behav Sci (Basel) 2020; 10:bs10020047. [PMID: 32024073 PMCID: PMC7071396 DOI: 10.3390/bs10020047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 11/16/2022] Open
Abstract
This study aimed to examine the effect of acute sleep curtailment on sweet taste preference, appetite and food intake, and the correlation between food intake and sweet taste preference or active ghrelin using a randomized crossover design (5 h sleep curtailment vs. 8 h control). Twenty-four participants (11 men) aged 21.4 ± 1.0 years, with BMI 19.8 ± 1.7 kg/m2, who habitually slept 5 h/night or more experienced interventions lasting three consecutive nights. Participants came into the laboratory for testing on day 4. Fasting blood tests were conducted at 8:00 a.m. to measure active ghrelin and leptin levels. Sweet taste preference was assessed by presenting five different concentration sucrose solutions at 9:00 a.m. Ad libitum intake at breakfast was assessed for 30 min from 9:30 a.m. Sweet taste preference was higher following sleep curtailment than control. Active ghrelin was likewise higher following sleep curtailment than control. Leptin did not differ between conditions. Energy intake was higher following sleep curtailment than control, being derived primarily from carbohydrates. However, sweet taste preference and active ghrelin did not correlate with energy intake. These results suggest that acute consecutive sleep curtailment increases sweet taste preference, active ghrelin, and energy intake in healthy young adults.
Collapse
Affiliation(s)
- Eri Tajiri
- Graduate School of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, 3-1-100 Tsukide, Higashi-ku, Kumamoto 862-8502, Japan;
| | - Eiichi Yoshimura
- Faculty of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, 3-1-100 Tsukide, Higashi-ku, Kumamoto 862-8502, Japan; (H.S.); (S.S.)
- Correspondence: ; Tel.: +81-096-383-2929 x 454
| | - Yoichi Hatamoto
- Department of Nutrition and Metabolism, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo 162-8636, Japan; (Y.H.); (S.T.)
| | - Hideki Shiratsuchi
- Faculty of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, 3-1-100 Tsukide, Higashi-ku, Kumamoto 862-8502, Japan; (H.S.); (S.S.)
| | - Shigeho Tanaka
- Department of Nutrition and Metabolism, National Institute of Health and Nutrition, National Institutes of Biomedical Innovation, Health and Nutrition, Tokyo 162-8636, Japan; (Y.H.); (S.T.)
| | - Seiya Shimoda
- Faculty of Environmental and Symbiotic Sciences, Prefectural University of Kumamoto, 3-1-100 Tsukide, Higashi-ku, Kumamoto 862-8502, Japan; (H.S.); (S.S.)
| |
Collapse
|
298
|
Abstract
Feeding schedules entrain circadian clocks in multiple brain regions and most peripheral organs and tissues, thereby synchronizing daily rhythms of foraging behavior and physiology with times of day when food is most likely to be found. Entrainment of peripheral clocks to mealtime is accomplished by multiple feeding-related signals, including absorbed nutrients and metabolic hormones, acting in parallel or in series in a tissue-specific fashion. Less is known about the signals that synchronize circadian clocks in the brain with feeding time, some of which are presumed to generate the circadian rhythms of food-anticipatory activity that emerge when food is restricted to a fixed daily mealtime. In this commentary, I consider the possibility that food-anticipatory activity rhythms are driven or entrained by circulating ghrelin, ketone bodies or insulin. While evidence supports the potential of these signals to participate in the induction or amount of food-anticipatory behavior, it falls short of establishing either a necessary or sufficient role or accounting for circadian properties of anticipatory rhythms. The availability of multiple, circulating signals by which circadian oscillators in many brain regions might entrain to mealtime has supported a view that food-anticipatory rhythms of behavior are mediated by a broadly distributed system of clocks. The evidence, however, does not rule out the possibility that multiple peripheral and central food-entrained oscillators and feeding-related signals converge on circadian oscillators in a defined location which ultimately set the phase and gate the expression of anticipatory activity rhythms. A candidate location is the dorsal striatum, a core component of the neural system which mediates reward, motivation and action and which contains circadian oscillators entrainable by food and dopaminergic drugs. Systemic metabolic signals, such as ghrelin, ketones and insulin, may participate in circadian food anticipation to the extent that they modulate dopamine afferents to circadian clocks in this area.
Collapse
Affiliation(s)
- Ralph E Mistlberger
- Department of Psychology, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A2S6, Canada
| |
Collapse
|
299
|
Sato T, Oishi K, Ida T, Kojima M. Suppressive effect of ghrelin on nicotine-induced clock gene expression in the mouse pancreas. Endocr J 2020; 67:73-80. [PMID: 31611477 DOI: 10.1507/endocrj.ej19-0169] [Citation(s) in RCA: 1] [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] [Indexed: 11/23/2022] Open
Abstract
Those who smoke nicotine-based cigarettes have elevated plasma levels of ghrelin, a hormone secreted from the stomach. Ghrelin has various physiological functions and has recently been shown to be involved in regulating biological rhythms. Therefore, in this study, in order to clarify the significance of the plasma ghrelin increase in smokers, we sought to clarify how nicotine and ghrelin affect the expression dynamics of clock genes using a mouse model. A single dose of nicotine administered intraperitoneally increased plasma ghrelin concentrations transiently, whereas continuous administration of nicotine with an osmotic minipump did not induce any change in the plasma ghrelin concentration. Single administration of nicotine resulted in a transient increase in ghrelin gene expression in the pancreas but not in the stomach, which is the major producer of ghrelin. In addition, in the pancreas, the expression of clock genes was also increased temporarily. Therefore, in order to clarify the interaction between nicotine-induced ghrelin gene expression and clock gene expression in the pancreas, nicotine was administered to ghrelin gene-deficient mice. Administration of nicotine to ghrelin-gene deficient mice increased clock gene expression in the pancreas. However, upon nicotine administration to mice pretreated with octanoate to upregulate ghrelin activity, expression levels of nicotine-inducible clock genes in the pancreas were virtually the same as those in mice not administered nicotine. Thus, our findings indicate that pancreatic ghrelin may suppress nicotine-induced clock gene expression in the pancreas.
Collapse
Affiliation(s)
- Takahiro Sato
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Kanae Oishi
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Fukuoka 830-0011, Japan
| | - Takanori Ida
- Division for Searching and Identification of Bioactive Peptides, Department of Bioactive Peptides, Frontier Science Research Center, University of Miyazaki, Kiyotake, Miyazaki 889-1692, Japan
| | - Masayasu Kojima
- Molecular Genetics, Institute of Life Science, Kurume University, Kurume, Fukuoka 830-0011, Japan
| |
Collapse
|
300
|
Pituitary Adenylate Cyclase-Activating Polypeptide in the Ventromedial Hypothalamus Is Responsible for Food Intake Behavior by Modulating the Expression of Agouti-Related Peptide in Mice. Mol Neurobiol 2020; 57:2101-2114. [DOI: 10.1007/s12035-019-01864-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/22/2019] [Indexed: 12/18/2022]
|