1
|
Carpinelli S, Ahlert J, Rubin M, Aratani A, Smith E, Floyd D, Potter RM, Al-Nakkash L. Deleterious impacts of Western diet on jejunum function and health are reversible. Am J Physiol Gastrointest Liver Physiol 2025; 328:G83-G93. [PMID: 39711223 DOI: 10.1152/ajpgi.00160.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/22/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024]
Abstract
The goal of this study was to determine whether the influence of a high-fat high-sugar diet (Western diet) on intestinal function and health was reversible. We measured transepithelial short circuit current (Isc), across freshly isolated segments of jejunum from male C57Bl/6J mice randomly assigned to one of the following groups for the study duration: high-fat high-sugar diet for 24 wk (HFHS), HFHS diet for 12 wk then switched to standard chow and water for a further 12 wk (Std), and lean controls (standard chow and water for 24 wk). At the completion of the study, segments of jejunum were frozen for Western blot determination of key proteins involved in secretory and absorptive functions, as well as senescence. Intestinal morphology was assessed. Serum and tissue assays were performed. Basal Isc was significantly decreased (by 42%, P < 0.05) in HFHS versus leans. This decrease in Isc was fully reversed by switching to Std diet. The HFHS-induced decrease in Isc was attributed to a significant loss of calcium-activated chloride channel (ClC2) expression. Changes in inflammatory state (TNF-α) and intestinal health [myeloperoxidase (MPO) activity] were associated with body weight changes. Our data suggests that the reduced basal jejunal Isc in HFHS mice is reversible. Better understanding of Western diet-mediated intestinal disturbances may permit for improved treatment options for gastrointestinal abnormalities in obese individuals.NEW & NOTEWORTHY Our data suggests that the reduced basal jejunal Isc (decreased secretory function) in Western diet-fed mice is reversible. A better understanding of Western diet-mediated intestinal disturbances may permit improved treatment options for gastrointestinal abnormalities in obese individuals.
Collapse
Affiliation(s)
- Sarah Carpinelli
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona, United States
| | - John Ahlert
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona, United States
| | - Maxwell Rubin
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona, United States
| | - Alex Aratani
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona, United States
| | - Emma Smith
- College of Veterinary Medicine, Midwestern University, Glendale, Arizona, United States
| | - Dana Floyd
- Department of Basic Medical Sciences, University of Arizona College of Medicine-Phoenix, Arizona, United States
| | - Ross M Potter
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, Arizona, United States
| | - Layla Al-Nakkash
- Arizona College of Osteopathic Medicine, Midwestern University, Glendale, Arizona, United States
- Department of Physiology, College of Graduate Studies, Midwestern University, Glendale, Arizona, United States
| |
Collapse
|
2
|
Spencer NJ, Keating DJ. Role of 5-HT in the enteric nervous system and enteroendocrine cells. Br J Pharmacol 2025; 182:471-483. [PMID: 35861711 DOI: 10.1111/bph.15930] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 11/29/2022] Open
Abstract
Since the 1950s, considerable circumstantial evidence had been presented that endogenous 5-HT (serotonin) synthesized from within the wall of the gastrointestinal (GI) tract played an important role in GI motility and transit. However, identifying the precise functional role of gut-derived 5-HT has been difficult to ascertain, for a number of reasons. Over the past decade, as recording techniques have advanced significantly and access to new genetically modified animals improved, there have been major new insights and major changes in our understanding of the functional role of endogenous 5-HT in the GI tract. Data from many different laboratories have shown that major patterns of GI motility and transit still occur with minor or no, change when all endogenous 5-HT is pharmacologically or genetically ablated from the gut. Furthermore, antagonists of 5-HT3 receptors are equally, or more potent at inhibiting GI motility in segments of intestine that are completely depleted of endogenous 5-HT. Here, the most recent findings are discussed with regard to the functional role of endogenous 5-HT in enterochromaffin cells and enteric neurons in gut motility and more broadly in some major homeostatic pathways.
Collapse
Affiliation(s)
- Nick J Spencer
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University of South Australia, Adelaide, Australia
| | - Damien J Keating
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University of South Australia, Adelaide, Australia
| |
Collapse
|
3
|
Liu Y, Wang Z, Fang L, Xu Y, Zhao B, Kang X, Zhao Y, Han J, Zhang Y, Dong E, Wang N. Deficiency of 5-HT 2B receptors alleviates atherosclerosis by regulating macrophage phenotype through inhibiting interferon signalling. Br J Pharmacol 2024. [PMID: 39232850 DOI: 10.1111/bph.17315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 06/09/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND AND PURPOSE Elevated levels of 5-HT have been correlated with coronary artery disease and cardiac events, suggesting 5-HT is a potential novel factor in the development of atherosclerotic cardiovascular disease. However, the underlying pathological mechanisms of the 5-HT system in atherosclerosis remain unclear. The 5-HT2B receptor (5-HT2BR), which establishes a positive feedback loop with 5-HT, has been identified as a contributor to pathophysiological processes in various vascular disorders. In this study, we investigated the immunological impact of 5-HT2BR in atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice. EXPERIMENTAL APPROACH Plasma levels of 5-HT were measured in mice using an ELISA kit. Atherosclerotic plaque formation, macrophage infiltration and inflammatory signalling were assessed in ApoE-/- mice by employing both pharmacological inhibition and genetic deficiency of 5-HT2BR. Inflammasome activation was elucidated using peritoneal macrophages isolated from 5-HT2BR-deficient mice. KEY RESULTS An upregulation of 5-HT2BR expression was observed in the aortas of ApoE-/- mice, exhibiting a strong correlation with the presence of macrophages in plaques. Atherosclerosis was attenuated in mice through pharmacological inhibition and genetic deficiency of 5-HT2BR. Additionally, a significant reduction in atherosclerotic plaque size was achieved through bone marrow reconstitution with 5-HT2BR-deficient cells. 5-HT2BR-deficient macrophages showed attenuated interferon (IFN) signalling, NLRP3 inflammasome activation, and interleukin-1β release. Moreover, macrophages primed with 5-HT2BR deficiency displayed an anti-inflammatory phenotype. CONCLUSION AND IMPLICATIONS These findings support the hypothesis that 5-HT2BR in macrophages plays a causal role in the development of atherosclerosis, revealing a novel perspective for potential therapeutic strategies in atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Yahan Liu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhipeng Wang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Li Fang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yaohua Xu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Beilei Zhao
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xuya Kang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yanqing Zhao
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Jintao Han
- Department of Interventional Radiology and Vascular Surgery, Peking University Third Hospital, Beijing, China
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Institute of Cardiovascular Diseases, The first affiliated Hospital of Dalian Medical University, Dalian, China
| | - Erdan Dong
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital); School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, China
| | - Nanping Wang
- Wuhu Hospital, East China Normal University (ECNU), Wuhu, China
- East China Normal University Health Science Center, Shanghai, China
| |
Collapse
|
4
|
Jiang L, Han D, Hao Y, Song Z, Sun Z, Dai Z. Linking serotonin homeostasis to gut function: Nutrition, gut microbiota and beyond. Crit Rev Food Sci Nutr 2024; 64:7291-7310. [PMID: 36861222 DOI: 10.1080/10408398.2023.2183935] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Serotonin (5-HT) produced by enterochromaffin (EC) cells in the digestive tract is crucial for maintaining gut function and homeostasis. Nutritional and non-nutritional stimuli in the gut lumen can modulate the ability of EC cells to produce 5-HT in a temporal- and spatial-specific manner that toning gut physiology and immune response. Of particular interest, the interactions between dietary factors and the gut microbiota exert distinct impacts on gut 5-HT homeostasis and signaling in metabolism and the gut immune response. However, the underlying mechanisms need to be unraveled. This review aims to summarize and discuss the importance of gut 5-HT homeostasis and its regulation in maintaining gut metabolism and immune function in health and disease with special emphasis on different types of nutrients, dietary supplements, processing, and gut microbiota. Cutting-edge discoveries in this area will provide the basis for the development of new nutritional and pharmaceutical strategies for the prevention and treatment of serotonin homeostasis-related gut and systematic disorders and diseases.
Collapse
Affiliation(s)
- Lili Jiang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Youling Hao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhuan Song
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhiyuan Sun
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, P. R. China
| |
Collapse
|
5
|
Yeoman MS, Fidalgo S, Marcelli G, Patel BA. Amperometry approach curve profiling to understand the regulatory mechanisms governing the concentration of intestinal extracellular serotonin. Sci Rep 2024; 14:10479. [PMID: 38714793 PMCID: PMC11076564 DOI: 10.1038/s41598-024-61296-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 05/03/2024] [Indexed: 05/10/2024] Open
Abstract
Enterochromaffin (EC) cells located within the intestinal mucosal epithelium release serotonin (5-HT) to regulate motility tones, barrier function and the immune system. Electroanalytical methodologies have been able to monitor steady state basal extracellular 5-HT levels but are unable to provide insight into how these levels are influenced by key regulatory processes such as release and uptake. We established a new measurement approach, amperometry approach curve profiling, which monitors the extracellular 5-HT level at different electrode-tissue (E-T) distances. Analysis of the current profile can provide information on contributions of regulatory components on the observed extracellular 5-HT level. Measurements were conducted from ex vivo murine ileum and colon using a boron-doped diamond (BDD) microelectrode. Amperometry approach curve profiling coupled with classical pharmacology demonstrated that extracellular 5-HT levels were significantly lower in the colon when compared to the ileum. This difference was due to a greater degree of activity of the 5-HT transporter (SERT) and a reduced amount of 5-HT released from colonic EC cells. The presence of an inhibitory 5-HT4 autoreceptor was observed in the colon, where a 40% increase in extracellular 5-HT was the half maximal inhibitory concentration for activation of the autoreceptor. This novel electroanalytical approach allows estimates of release and re-uptake and their contribution to 5-HT extracellular concentration from intestinal tissue be obtained from a single series of measurements.
Collapse
Affiliation(s)
- Mark S Yeoman
- School of Applied Sciences, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
- Centre for Lifelong Health, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
| | - Sara Fidalgo
- School of Applied Sciences, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
- Centre for Lifelong Health, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK
| | - Gianluca Marcelli
- School of Engineering, University of Kent, Jennison Building, Canterbury, CT2 7NZ, UK
| | - Bhavik Anil Patel
- School of Applied Sciences, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK.
- Centre for Lifelong Health, University of Brighton, Huxley Building, Brighton, BN2 4GJ, UK.
| |
Collapse
|
6
|
Vanslette AM, Toft PB, Lund ML, Moritz T, Arora T. Serotonin receptor 4 agonism prevents high fat diet induced reduction in GLP-1 in mice. Eur J Pharmacol 2023; 960:176181. [PMID: 37926275 DOI: 10.1016/j.ejphar.2023.176181] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/02/2023] [Accepted: 11/02/2023] [Indexed: 11/07/2023]
Abstract
Hormone-producing enteroendocrine cells (EECs) are present throughout the gastrointestinal tract and respond to various nutrient and gut microbiota produced metabolites stimuli. Two important EEC subtypes, Glucagon like peptide-1 (GLP-1) producing L-cells and serotonin (5-HT) producing enterochromaffin (EC) cells interact via paracrine signaling and exhibit bidirectional regulation of expression and secretion of produced hormones. Accordingly, in vitro studies suggest potential to modulate 5-HT secretion by GLP-1 receptor agonism, and L-cell differentiation via serotonin receptor 4 agonism. However, the importance of this cellular signaling on host metabolism is poorly understood. In this study, we found that two weeks of high fat diet (HFD) feeding reduced RNA expression of gut hormones, including proglucagon (Gcg) gene encoding GLP-1 and Tryptophan hydroxylase1 (Tph1) gene encoding rate limiting enzyme in 5-HT synthesis, specifically in the colon and reduced plasma GLP-1 levels. Levels of propionate and butyrate were also reduced following HFD. However, supplementation of sodium propionate did not improve HFD induced reduction in GLP-1. In contrast, chemical induction of serotonin receptor 4 promoted GLP-1 levels, colonic Gcg RNA expression accompanied by improvement in glucose tolerance in HFD-fed mouse. Thus, this study suggests a novel mechanism to improve glucose tolerance via serotonin receptor 4 stimulation in the HFD induced obese mouse model.
Collapse
Affiliation(s)
- Amanda Marie Vanslette
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Pernille Baumann Toft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Mari Lilith Lund
- Human Health Research, Scientific Affairs, Chr. Hansen A/S, Bøge Alle 10-12, 2970, Hørsholm, Denmark
| | - Thomas Moritz
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
7
|
Di Ciaula A, Bonfrate L, Khalil M, Garruti G, Portincasa P. Contribution of the microbiome for better phenotyping of people living with obesity. Rev Endocr Metab Disord 2023; 24:839-870. [PMID: 37119391 PMCID: PMC10148591 DOI: 10.1007/s11154-023-09798-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 05/01/2023]
Abstract
Obesity has reached epidemic proportion worldwide and in all ages. Available evidence points to a multifactorial pathogenesis involving gene predisposition and environmental factors. Gut microbiota plays a critical role as a major interface between external factors, i.e., diet, lifestyle, toxic chemicals, and internal mechanisms regulating energy and metabolic homeostasis, fat production and storage. A shift in microbiota composition is linked with overweight and obesity, with pathogenic mechanisms involving bacterial products and metabolites (mainly endocannabinoid-related mediators, short-chain fatty acids, bile acids, catabolites of tryptophan, lipopolysaccharides) and subsequent alterations in gut barrier, altered metabolic homeostasis, insulin resistance and chronic, low-grade inflammation. Although animal studies point to the links between an "obesogenic" microbiota and the development of different obesity phenotypes, the translational value of these results in humans is still limited by the heterogeneity among studies, the high variation of gut microbiota over time and the lack of robust longitudinal studies adequately considering inter-individual confounders. Nevertheless, available evidence underscores the existence of several genera predisposing to obesity or, conversely, to lean and metabolically health phenotype (e.g., Akkermansia muciniphila, species from genera Faecalibacterium, Alistipes, Roseburia). Further longitudinal studies using metagenomics, transcriptomics, proteomics, and metabolomics with exact characterization of confounders are needed in this field. Results must confirm that distinct genera and specific microbial-derived metabolites represent effective and precision interventions against overweight and obesity in the long-term.
Collapse
Affiliation(s)
- Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Gabriella Garruti
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), University of Bari Medical School, 70124 Bari, Italy
| |
Collapse
|
8
|
Pelantová H, Tomášová P, Šedivá B, Neprašová B, Mráziková L, Kuneš J, Železná B, Maletínská L, Kuzma M. Metabolomic Study of Aging in fa/ fa Rats: Multiplatform Urine and Serum Analysis. Metabolites 2023; 13:metabo13040552. [PMID: 37110210 PMCID: PMC10142631 DOI: 10.3390/metabo13040552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/27/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Zucker fatty (fa/fa) rats represent a well-established and widely used model of genetic obesity. Because previous metabolomic studies have only been published for young fa/fa rats up to 20 weeks of age, which can be considered early maturity in male fa/fa rats, the aim of our work was to extend the metabolomic characterization to significantly older animals. Therefore, the urinary profiles of obese fa/fa rats and their lean controls were monitored using untargeted NMR metabolomics between 12 and 40 weeks of age. At the end of the experiment, the rats were also characterized by NMR and LC-MS serum analysis, which was supplemented by a targeted LC-MS analysis of serum bile acids and neurotransmitters. The urine analysis showed that most of the characteristic differences detected in young obese fa/fa rats persisted throughout the experiment, primarily through a decrease in microbial co-metabolite levels, the upregulation of the citrate cycle, and changes in nicotinamide metabolism compared with the age-related controls. The serum of 40-week-old obese rats showed a reduction in several bile acid conjugates and an increase in serotonin. Our study demonstrated that the fa/fa model of genetic obesity is stable up to 40 weeks of age and is therefore suitable for long-term experiments.
Collapse
Affiliation(s)
- Helena Pelantová
- Institute of Microbiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Petra Tomášová
- Institute of Microbiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic
- First Faculty of Medicine, Charles University and General University Hospital in Prague, 128 08 Prague, Czech Republic
| | - Blanka Šedivá
- Faculty of Applied Sciences, University of West Bohemia, 306 14 Pilsen, Czech Republic
| | - Barbora Neprašová
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 160 00 Prague, Czech Republic
| | - Lucia Mráziková
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 160 00 Prague, Czech Republic
| | - Jaroslav Kuneš
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 160 00 Prague, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| | - Blanka Železná
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 160 00 Prague, Czech Republic
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, 160 00 Prague, Czech Republic
| | - Marek Kuzma
- Institute of Microbiology, Czech Academy of Sciences, 142 20 Prague, Czech Republic
| |
Collapse
|
9
|
Body weight changes and bipolar disorder: a molecular pathway analysis. Pharmacogenet Genomics 2022; 32:308-320. [DOI: 10.1097/fpc.0000000000000484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Kwon YH, Khan WI. Peripheral Serotonin: Cultivating Companionship with Gut Microbiota in Intestinal Homeostasis. Am J Physiol Cell Physiol 2022; 323:C550-C555. [PMID: 35759441 DOI: 10.1152/ajpcell.00433.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Serotonin, also known as 5-hydroxytryptamine (5-HT), is an evolutionarily ancient and phylogenetically conserved monoamine that regulates multifaceted physiological functions in mammals. 5-HT was, at one time, most extensively studied as a neurotransmitter within the central nervous system but is now known to regulate non-neuronal functions including immune responses in an autocrine-paracrine-endocrine manner. Compelling evidence from intervention studies using germ-free mice or antibiotic-associated microbiota perturbation suggests that novel interactions between 5-HT and the gut microbiota are essential in maintaining intestinal homeostasis. Importantly, recent studies reveal that bidirectional host-microbial interactions mediated by the host serotonergic system can promote distinct changes within the gut microbiota. These changes may potentially lead to a state known as 'dysbiosis' which has been strongly associated with various gut pathologies including inflammatory bowel disease (IBD). In this review, we update the current understanding of host-microbiota interaction by focusing on the impact of peripheral 5-HT signaling within this dynamic. We also briefly highlight key environmental risk factors for IBD, such as Western diet, and draw attention to the interaction of synthetic food colorants with 5-HT signaling that may facilitate future research.
Collapse
Affiliation(s)
- Yun Han Kwon
- Department of Pathology and Molecular Medicine, McMaster University; Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Waliul I Khan
- Department of Pathology and Molecular Medicine, McMaster University; Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Laboratory Medicine, Hamilton Health Sciences, Hamilton, Ontario, Canada
| |
Collapse
|
11
|
Krivošíková K, Krivošíková Z, Wsolová L, Seeman T, Podracká Ľ. Hypertension in obese children is associated with vitamin D deficiency and serotonin dysregulation. BMC Pediatr 2022; 22:289. [PMID: 35581625 PMCID: PMC9112480 DOI: 10.1186/s12887-022-03337-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 05/04/2022] [Indexed: 11/20/2022] Open
Abstract
Background Obesity and hypertension represent serious health issues affecting the pediatric population with increasing prevalence. Hypovitaminosis D has been suggested to be associated with arterial hypertension. Serotonin by modulating nitric oxide synthase affect blood pressure regulation. The biological mechanism by which vitamin D specifically regulates serotonin synthesis was recently described. The aim of this paper is to determine the associations between vitamin D, serotonin, and blood pressure in obese children. Methods One hundred and seventy-one children were enrolled in the prospective cross-sectional study. Two groups of children divided according to body mass index status to obese (BMI ≥95th percentile; n = 120) and non-obese (n = 51) were set. All children underwent office and ambulatory blood pressure monitoring and biochemical analysis of vitamin D and serotonin. Data on fasting glucose, insulin, HOMA, uric acid, and complete lipid profile were obtained in obese children. Results Hypertension was found only in the group of obese children. Compared to the control group, obese children had lower vitamin D and serotonin, especially in winter. The vitamin D seasonality and BMI-SDS were shown as the most significant predictors of systolic blood pressure changes, while diastolic blood pressure was predicted mostly by insulin and serotonin. The presence of hypertension and high-normal blood pressure in obese children was most significantly affected by vitamin D deficiency and increased BMI-SDS. Conclusions Dysregulation of vitamin D and serotonin can pose a risk of the onset and development of hypertension in obese children; therefore, their optimization together with reducing body weight may improve the long-term cardiovascular health of these children. Supplementary Information The online version contains supplementary material available at 10.1186/s12887-022-03337-8.
Collapse
Affiliation(s)
- Katarína Krivošíková
- Department of Pediatrics, National Institute of Children's Diseases and Faculty of Medicine, Comenius University, Limbová 1, Bratislava, 831 01, Slovak Republic.
| | - Zora Krivošíková
- Department of Clinical and Experimental Pharmacotherapy, Faculty of Medicine, Slovak Medical University, Bratislava, Slovak Republic
| | - Ladislava Wsolová
- Department of Biophysics, Informatics and Biostatistics, Faculty of Public Health, Slovak Medical University, Bratislava, Slovak Republic
| | - Tomáš Seeman
- Department of Pediatrics, 2nd Medical Faculty, Charles University Prague, Prague, Czech Republic.,Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Ľudmila Podracká
- Department of Pediatrics, National Institute of Children's Diseases and Faculty of Medicine, Comenius University, Limbová 1, Bratislava, 831 01, Slovak Republic
| |
Collapse
|
12
|
Hatton-Jones KM, du Toit EF, Cox AJ. Effect of chronic restraint stress and western-diet feeding on colonic regulatory gene expression in mice. Neurogastroenterol Motil 2022; 34:e14300. [PMID: 34825433 DOI: 10.1111/nmo.14300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/07/2021] [Accepted: 11/10/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Diet-induced obesity (DIO) and psychological stress are significant independent regulators of gastrointestinal physiology; however, our understanding of how these two disorders influence the host-microbe interface is still poorly characterized. The aim of this study was to assess the combined influences of diet-induced obesity and psychological stress on microbiome composition and colonic gene expression. METHODS C57BL/6J mice (n = 48) were subject to a combination of 22 weeks of Western diet (WD) feeding and a chronic restraint stressor (CRS) for the last 4 weeks of feeding. At the end of the combined intervention, microbiome composition was determined from cecal contents, and colonic tissue gene expression was assessed by multiplex analysis using NanoString nCounter System and real-time qPCR. RESULTS WD feeding induced a DIO phenotype with increased body weight, worsened metabolic markers, and alterations to microbiome composition. CRS reduced body weight in both dietary groups while having differential effects on glucose metabolism. CRS improved the Firmicutes/Bacteroidetes ratio in WD-fed animals while expanding the Proteobacteria phyla. Significantly lower expression of colonic Tlr4 (p = 0.008), Ocln (p = 0.004), and Cldn3 (p = 0.004) were noted in WD-fed animals compared to controls with no synergistic effects observed when combined with CRS. No changes to colonic expression of downstream inflammatory mediators were observed. Interestingly, higher levels of expression of Cldn2 (p = 0.04) and bile acid receptor Nr1h4 (p = 0.02) were seen in mice exposed to CRS. CONCLUSION Differential but not synergistic effects of WD and CRS were noted at the host-microbe interface suggesting multifactorial responses that require further investigation.
Collapse
Affiliation(s)
- Kyle M Hatton-Jones
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Eugene F du Toit
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Amanda J Cox
- School of Medical Science, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
13
|
Tao E, Zhu Z, Hu C, Long G, Chen B, Guo R, Fang M, Jiang M. Potential Roles of Enterochromaffin Cells in Early Life Stress-Induced Irritable Bowel Syndrome. Front Cell Neurosci 2022; 16:837166. [PMID: 35370559 PMCID: PMC8964523 DOI: 10.3389/fncel.2022.837166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/09/2022] [Indexed: 12/04/2022] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most common functional gastrointestinal disorders, also known as disorders of the gut–brain interaction; however, the pathophysiology of IBS remains unclear. Early life stress (ELS) is one of the most common risk factors for IBS development. However, the molecular mechanisms by which ELS induces IBS remain unclear. Enterochromaffin cells (ECs), as a prime source of peripheral serotonin (5-HT), play a pivotal role in intestinal motility, secretion, proinflammatory and anti-inflammatory effects, and visceral sensation. ECs can sense various stimuli and microbiota metabolites such as short-chain fatty acids (SCFAs) and secondary bile acids. ECs can sense the luminal environment and transmit signals to the brain via exogenous vagal and spinal nerve afferents. Increasing evidence suggests that an ECs-5-HT signaling imbalance plays a crucial role in the pathogenesis of ELS-induced IBS. A recent study using a maternal separation (MS) animal model mimicking ELS showed that MS induced expansion of intestinal stem cells and their differentiation toward secretory lineages, including ECs, leading to ECs hyperplasia, increased 5-HT production, and visceral hyperalgesia. This suggests that ELS-induced IBS may be associated with increased ECs-5-HT signaling. Furthermore, ECs are closely related to corticotropin-releasing hormone, mast cells, neuron growth factor, bile acids, and SCFAs, all of which contribute to the pathogenesis of IBS. Collectively, ECs may play a role in the pathogenesis of ELS-induced IBS. Therefore, this review summarizes the physiological function of ECs and focuses on their potential role in the pathogenesis of IBS based on clinical and pre-clinical evidence.
Collapse
Affiliation(s)
- Enfu Tao
- Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
- Wenling Maternal and Child Health Care Hospital, Wenling, China
| | - Zhenya Zhu
- Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Chenmin Hu
- Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Gao Long
- Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Bo Chen
- Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Rui Guo
- Endoscopy Center and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
| | - Marong Fang
- Institute of Neuroscience and Gastrointestinal Laboratory, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mizu Jiang
- Department of Gastroenterology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, China
- *Correspondence: Mizu Jiang,
| |
Collapse
|
14
|
Stępniowska A, Tutaj K, Juśkiewicz J, Ognik K. Effect of a high-fat diet and chromium on hormones level and Cr retention in rats. J Endocrinol Invest 2022; 45:527-535. [PMID: 34550535 PMCID: PMC8850218 DOI: 10.1007/s40618-021-01677-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 09/12/2021] [Indexed: 12/17/2022]
Abstract
AIMS The aim of the study was to determine how the administration of a high-fat diet supplemented with various forms of chromium to rats affects accumulation of this element in the tissues and levels of leptin, ghrelin, insulin, glucagon, serotonin, noradrenaline and histamine, as well as selected mineral elements. METHODS The experiment was conducted on 56 male Wistar rats, which were divided into 8 experimental groups. The rats received standard diet or high fat diet (HFD) with addition of 0.3 mg/kg body weight of chromium(III) picolinate (Cr-Pic), chromium(III)-methioninate (Cr-Met), or chromium nanoparticles (Cr-NP). RESULTS Chromium in organic forms was found to be better retained in the body of rats than Cr in nanoparticles form. However, Cr-Pic was the only form that increased the insulin level, which indicates its beneficial effect on carbohydrate metabolism. In blood plasma of rats fed a high-fat diet noted an increased level of serotonin and a reduced level of noradrenaline. The addition of Cr to the diet, irrespective of its form, also increased the serotonin level, which should be considered a beneficial effect. Rats fed a high-fat diet had an unfavourable reduction in the plasma concentrations of Ca, P, Mg and Zn. The reduction of P in the plasma induced by supplementation with Cr in the form of Cr-Pic or Cr-NP may exacerbate the adverse effect of a high-fat diet on the level of this element. CONCLUSION A high-fat diet was shown to negatively affect the level of hormones regulating carbohydrate metabolism (increasing leptin levels and decreasing levels of ghrelin and insulin).
Collapse
Affiliation(s)
- A Stępniowska
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland.
| | - K Tutaj
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| | - J Juśkiewicz
- Division of Food Science, Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| | - K Ognik
- Department of Biochemistry and Toxicology, Faculty of Animal Sciences and Bioeconomy, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| |
Collapse
|
15
|
France M, Galligan JJ, Swain GM. In vitro electrochemical measurement of serotonin release in the human jejunum mucosa using a diamond microelectrode. Analyst 2022; 147:2523-2532. [PMID: 35543208 PMCID: PMC9599047 DOI: 10.1039/d2an00487a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report herein on the use of a boron-doped diamond microelectrode (DME) to record oxidation currents in vitro associated with the release of serotonin from enterochromaffin cells in the epithelium of the human intestinal mucosa.
Collapse
Affiliation(s)
- Marion France
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
| | - James J. Galligan
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| | - Greg M. Swain
- Department of Chemistry, Michigan State University, East Lansing, MI 48824, USA
- Neuroscience Program, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
16
|
Altered intestinal epithelial nutrient transport: an underappreciated factor in obesity modulated by diet and microbiota. Biochem J 2021; 478:975-995. [PMID: 33661278 DOI: 10.1042/bcj20200902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/31/2022]
Abstract
Dietary nutrients absorbed in the proximal small intestine and assimilated in different tissues have a profound effect on overall energy homeostasis, determined by a balance between body's energy intake and expenditure. In obesity, altered intestinal absorption and consequently tissue assimilation of nutrients may disturb the energy balance leading to metabolic abnormalities at the cellular level. The absorption of nutrients such as sugars, amino acids and fatty acids released from food digestion require high-capacity transporter proteins expressed in the intestinal epithelial absorptive cells. Furthermore, nutrient sensing by specific transporters/receptors expressed in the epithelial enteroendocrine cells triggers release of gut hormones involved in regulating energy homeostasis via their effects on appetite and food intake. Therefore, the intestinal epithelial cells play a pivotal role in the pathophysiology of obesity and associated complications. Over the past decade, gut microbiota has emerged as a key factor contributing to obesity via its effects on digestion and absorption of nutrients in the small intestine, and energy harvest from dietary fiber, undigested component of food, in the large intestine. Various mechanisms of microbiota effects on obesity have been implicated. However, the impact of obesity-associated microbiota on the intestinal nutrient transporters needs extensive investigation. This review marshals the limited studies addressing the altered structure and function of the gut epithelium in obesity with special emphasis on nutrient transporters and role of diet and microbiota. The review also discusses the thoughts and controversies and research gaps in this field.
Collapse
|
17
|
Deregulated Serotonin Pathway in Women with Morbid Obesity and NAFLD. Life (Basel) 2020; 10:life10100245. [PMID: 33081272 PMCID: PMC7603041 DOI: 10.3390/life10100245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) extends from simple steatosis (SS) to non-alcoholic steatohepatitis (NASH). Peripheral serotonin (5-HT) has become as an important regulator of different metabolic pathways. 5-HT has been related to obesity and lipid accumulation in the liver. The objective of this study was to assess the relationship between the 5-HT signaling pathway and the degree of NAFLD, as well as to investigate whether peripheral 5-HT levels are related to the hepatic and jejunal mRNA abundance of serotonin receptors (HTR) in a cohort of women with morbid obesity (MO) and NAFLD. ELISA was used to quantify the serum 5-HT from normal-weight subjects (n = 26) and patients with MO (n = 58). We used RTq-PCR analysis to evaluate the relative expression of HTR in women with MO with normal liver (n = 22), SS (n = 21), and NASH (n = 15). The 5-HT was diminished in women with MO under a hypocaloric diet, regardless of the presence of NAFLD. Additionally, we report a negative correlation of 5-HT levels with metabolic syndrome criteria, suggesting that serotonin may have a protective role in obesity. Additionally, the hepatic expression of HTR2A and HTR2B were decreased in women with MO and NAFLD, but no significant differences in the HTR jejunal expression according to the presence of NAFLD were found.
Collapse
|
18
|
Interplay between Peripheral and Central Inflammation in Obesity-Promoted Disorders: The Impact on Synaptic Mitochondrial Functions. Int J Mol Sci 2020; 21:ijms21175964. [PMID: 32825115 PMCID: PMC7504224 DOI: 10.3390/ijms21175964] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/12/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022] Open
Abstract
The metabolic dysfunctions induced by high fat diet (HFD) consumption are not limited to organs involved in energy metabolism but cause also a chronic low-grade systemic inflammation that affects the whole body including the central nervous system. The brain has been considered for a long time to be protected from systemic inflammation by the blood–brain barrier, but more recent data indicated an association between obesity and neurodegeneration. Moreover, obesity-related consequences, such as insulin and leptin resistance, mitochondrial dysfunction and reactive oxygen species (ROS) production, may anticipate and accelerate the physiological aging processes characterized by systemic inflammation and higher susceptibility to neurological disorders. Here, we discussed the link between obesity-related metabolic dysfunctions and neuroinflammation, with particular attention to molecules regulating the interplay between energetic impairment and altered synaptic plasticity, for instance AMP-activated protein kinase (AMPK) and Brain-derived neurotrophic factor (BDNF). The effects of HFD-induced neuroinflammation on neuronal plasticity may be mediated by altered brain mitochondrial functions. Since mitochondria play a key role in synaptic areas, providing energy to support synaptic plasticity and controlling ROS production, the negative effects of HFD may be more pronounced in synapses. In conclusion, it will be emphasized how HFD-induced metabolic alterations, systemic inflammation, oxidative stress, neuroinflammation and impaired brain plasticity are tightly interconnected processes, implicated in the pathogenesis of neurological diseases.
Collapse
|
19
|
Martin AM, Jones LA, Jessup CF, Sun EW, Keating DJ. Diet differentially regulates enterochromaffin cell serotonin content, density and nutrient sensitivity in the mouse small and large intestine. Neurogastroenterol Motil 2020; 32:e13869. [PMID: 32378785 DOI: 10.1111/nmo.13869] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 04/03/2020] [Accepted: 04/09/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Enterochromaffin (EC) cells are specialized enteroendocrine cells lining the gastrointestinal (GI) tract and the source of almost all serotonin (5-hydroxytryptamine; 5-HT) in the body. Gut-derived 5-HT has a plethora of physiological roles, including regulation of gastrointestinal motility, and has been implicated as a driver of obesity and metabolic disease. This is due to 5-HT influencing key metabolic processes, such as hepatic gluconeogenesis, adipose tissue lipolysis and hindering thermogenic capacity. Increased circulating 5-HT occurs in humans with obesity and type 2 diabetes. However, despite the known metabolic roles of gut-derived 5-HT, the mechanisms underlying the cellular-level change in EC cells under obesogenic conditions remains unknown. METHODS We use a mouse model of diet-induced obesity (DIO) to identify the regional changes that occur in primary EC cells from the duodenum and colon. Transcriptional changes in the nutrient sensing profile of primary EC cells were assessed, and responses to nutrient stimuli in culture were determined by 5-HT ELISA. KEY RESULTS We find that obesogenic conditions affect EC cells in a region-dependent manner. Duodenal EC cells from DIO mice have impaired sugar sensing even in the presence of increased 5-HT content per cell, while colonic EC cell numbers are significantly increased, but have unaltered nutrient sensing capacity. CONCLUSIONS & INFERENCES Our findings from this study add novel insights into the mechanisms by which functional changes to EC cells occur at a cellular level, which may contribute to the altered circulating 5-HT seen with obesity and metabolic disease, and associated gastrointestinal disorders.
Collapse
Affiliation(s)
- Alyce M Martin
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Lauren A Jones
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Claire F Jessup
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Emily W Sun
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Damien J Keating
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, Australia
| |
Collapse
|
20
|
Amino Acid Metabolites and Slow Weight Loss in the Early Postoperative Period after Sleeve Gastrectomy. J Clin Med 2020; 9:jcm9082348. [PMID: 32717870 PMCID: PMC7463855 DOI: 10.3390/jcm9082348] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/22/2020] [Indexed: 01/16/2023] Open
Abstract
Background: Profiles of amino acid metabolites (AAMs) have been linked to obesity and energy homeostasis. We investigated whether baseline obesity-related AAMs were associated with weight status in the early postoperative period after sleeve gastrectomy. Methods: In this prospective, single-arm, longitudinal study, 27 bariatric patients underwent sleeve gastrectomy. Twenty obesity-related AAMs were comprehensively quantified prior to surgery, and slow weight loss was defined as the lowest 40% of the percentage excess weight loss (%EWL) at three and six months postoperatively. Linear regression models were used to assess the association between baseline obesity-related AAMs and %EWL, and receiver operating characteristic curves were assessed. Results: Isoleucine and metabolites from the serotonin pathway were significantly associated with the %EWL at three and six months after sleeve gastrectomy. Among the metabolites identified to be significant in the regression analyses, serotonin (area under receiver operating characteristic curves (AUROC): 0.79, 95% confidence interval (CI): 0.59–0.97) and serotonin/5-hydroxytryptophan ratio (AUROC: 0.80, 95% CI: 0.58–1.00) showed superior performance in predicting slow weight loss six months after sleeve gastrectomy. Conclusions: Our findings underscore the importance of baseline AAM profiles, especially serotonin and serotonin/5-hydroxytryptophan ratio, in predicting slow weight loss in the early postoperative period after sleeve gastrectomy.
Collapse
|
21
|
Xie Y, Wang C, Zhao D, Wang C, Li C. Dietary Proteins Regulate Serotonin Biosynthesis and Catabolism by Specific Gut Microbes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:5880-5890. [PMID: 32363863 DOI: 10.1021/acs.jafc.0c00832] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
More than 90% of serotonin is produced in the intestine. Previous studies have shown that different protein diets significantly affect serum serotonin levels. Here, the colonic microbiota and intestinal serotonin were measured to elaborate how protein diets affect serotonin production in a mouse model. The emulsion-type sausage protein and cooked pork protein diets increased the mRNA levels of tryptophan hydroxylase 1 (Tph1) and monoamine oxidase A (Maoa) and serotonin level as well but reduced the number of enterochromaffin cells. However, the soy protein diet increased the number of enterochromaffin cells and Tph1 mRNA level but decreased the Maoa mRNA level and the serotonin content. Specific gut microbes that responded to dietary changes and affected the content of short-chain fatty acids were significantly related to serotonin-associated biomarkers. These results suggest that dietary proteins may regulate serotonin biosynthesis and catabolism by altering specific gut microbes.
Collapse
Affiliation(s)
- Yunting Xie
- Key Laboratory of Meat Processing and Quality Control, MOE; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; Key Laboratory of Meat Products Processing, MOA, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Chong Wang
- Key Laboratory of Meat Processing and Quality Control, MOE; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; Key Laboratory of Meat Products Processing, MOA, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Di Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; Key Laboratory of Meat Products Processing, MOA, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Chao Wang
- Key Laboratory of Meat Processing and Quality Control, MOE; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; Key Laboratory of Meat Products Processing, MOA, Nanjing Agricultural University, Nanjing 210095, P. R. China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE; Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control; Key Laboratory of Meat Products Processing, MOA, Nanjing Agricultural University, Nanjing 210095, P. R. China
- Joint International Research Laboratory of Animal Health and Food Safety, MOE, Nanjing Agricultural University, Nanjing 210095, P. R. China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, P. R. China
| |
Collapse
|
22
|
Wang L, Fan X, Han J, Cai M, Wang X, Wang Y, Shang J. Gut-Derived Serotonin Contributes to the Progression of Non-Alcoholic Steatohepatitis via the Liver HTR2A/PPARγ2 Pathway. Front Pharmacol 2020; 11:553. [PMID: 32477107 PMCID: PMC7240039 DOI: 10.3389/fphar.2020.00553] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 04/14/2020] [Indexed: 12/17/2022] Open
Abstract
The precipitous increase in occurrence of non-alcoholic steatohepatitis (NASH) is a serious threat to public health worldwide. The pathogenesis of NASH has not yet been thoroughly studied. We aimed to elucidate the interplay between serotonin (5-hydroxytryptamine, 5-HT) and NASH. The serum 5-HT levels in patients with non-alcoholic fatty liver disease (NAFLD) and a rat fed with high fat-sucrose diet (HFSD) were evaluated using liquid chromatography-hybrid quadrupole time-of-flight mass spectrometry (LC-QTOF MS)/MS. The peripheral Tph1 inhibitor, LP533401, and a tryptophan (TRP)-free diet were administered to rats with NASH, induced by HFSD. BRL-3A cells were treated with 1 mM free fatty acids (FFAs) and/or 50 μM 5-HT, and then small interfering RNA (siRNA) targeting the 5-HT2A receptor (HTR2A) and the PPARγ pharmaceutical agonist, pioglitazone, were applied. We found a marked correlation between 5-HT and NASH. The absence of 5-HT, through the pharmaceutical blockade of Tph1 (LP533401) and dietary control (TRP-free diet), suppressed hepatic lipid load and the expression of inflammatory factors (Tnfα, Il6, and Mcp-1). In BRL-3A cells, 50 μM 5-HT induced lipid accumulation and upregulated the expression of lipogenesis-ralated genes (Fas, Cd36, and Plin2) and the inflammatory response. Specifically, HTR2A knockdown and evaluation of PPARγ agonist activity revealed that HTR2A promoted hepatic steatosis and inflammation by activating PPARγ2. These results suggested that duodenal 5-HT was a risk factor in the pathological progression of NASH. Correspondingly, it may represent an attractive therapeutic target for preventing the development of NASH via the regulation of the HTR2A/PPARγ2 signaling pathway.
Collapse
Affiliation(s)
- Lulu Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Division of Clinical Pharmacy, Department of Pharmacy, Drum Tower Hospital Affiliated to Medical School of Nanjing University, Nanjing, China
| | - Xiangcheng Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, China
| | - Jichun Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, China
| | - Minxuan Cai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, China
| | - Xiaozhong Wang
- Department of Hepatology, Traditional Chinese Medicine Hospital, Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yan Wang
- Department of Traditional Chinese Medicine, Chengdu Fifth People's Hospital, Chengdu, China
| | - Jing Shang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
23
|
Sefidgari-Abrasi S, Roshangar L, Karimi P, Morshedi M, Rahimiyan-Heravan M, Saghafi-Asl M. From the gut to the heart: L. plantarum and inulin administration as a novel approach to control cardiac apoptosis via 5-HT2B and TrkB receptors in diabetes. Clin Nutr 2020; 40:190-201. [PMID: 32446786 DOI: 10.1016/j.clnu.2020.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 04/23/2020] [Accepted: 05/05/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS Type 2 diabetes mellitus, as a metabolic disorder, can lead to diabetic cardiomyopathy, identified by cardiomyocyte apoptosis and myocardial fibrosis. Brain-derived neurotrophic factor (BDNF) and serotonin are two neurotransmitters that can control cardiomyocyte apoptosis and myocardial fibrosis through their cardiac receptors. In the present study, we investigated the impacts of L. plantarum and inulin supplementation on the inhibition of cardiac apoptosis and fibrosis by modulating intestinal, serum, and cardiac levels of serotonin and BDNF as well as their cardiac receptors. METHODS Diabetes was induced by a high-fat diet and streptozotocin in male Wistar rats. Rats were divided into six groups and were supplemented with L. plantarum, inulin or their combination for 8 weeks. Finally, the rats were killed and levels of intestinal, serum, and cardiac parameters were evaluated. RESULTS Concurrent administration of L. plantarum and inulin caused a significant rise in the expression of cardiac serotonin and BDNF receptors (P < 0.001) as well as a significant fall in cardiac interstitial and perivascular fibrosis (P < 0.001, both) and apoptosis (P = 0.01). Moreover, there was a strong correlation of cardiac 5-Hydroxytryptamine 2B (5-HT2B) and tropomyosin receptor kinase B (TrkB) receptors with interstitial/perivascular fibrosis and apoptosis (P < 0.001, both). CONCLUSIONS/INTERPRETATION Results revealed beneficial effects of L. plantarum, inulin or their combination on intestinal, serum, and cardiac serotonin and BDNF accompanied by higher expression of their cardiac receptors and lower levels of cardiac apoptotic and fibrotic markers. It seems that L. plantarum and inulin supplementation could be considered as a novel adjunct therapy to reduce cardiac complications of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Safa Sefidgari-Abrasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pouran Karimi
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Morshedi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Rahimiyan-Heravan
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Saghafi-Asl
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Nutrition Research Center, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
24
|
Veniaminova E, Oplatchikova M, Bettendorff L, Kotenkova E, Lysko A, Vasilevskaya E, Kalueff AV, Fedulova L, Umriukhin A, Lesch KP, Anthony DC, Strekalova T. Prefrontal cortex inflammation and liver pathologies accompany cognitive and motor deficits following Western diet consumption in non-obese female mice. Life Sci 2019; 241:117163. [PMID: 31837337 DOI: 10.1016/j.lfs.2019.117163] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022]
Abstract
AIMS The high sugar and lipid content of the Western diet (WD) is associated with metabolic dysfunction, non-alcoholic steatohepatitis, and it is an established risk factor for neuropsychiatric disorders. Our previous studies reported negative effects of the WD on rodent emotionality, impulsivity, and sociability in adulthood. Here, we investigated the effect of the WD on motor coordination, novelty recognition, and affective behavior in mice as well as molecular and cellular endpoints in brain and peripheral tissues. MAIN METHODS Female C57BL/6 J mice were fed the WD for three weeks and were investigated for glucose tolerance, insulin resistance, liver steatosis, and changes in motor coordination, object recognition, and despair behavior in the swim test. Lipids and liver injury markers, including aspartate-transaminase, alanine-transaminase and urea were measured in blood. Serotonin transporter (SERT) expression, the density of Iba1-positive cells and concentration of malondialdehyde were measured in brain. KEY FINDINGS WD-fed mice exhibited impaired glucose tolerance and insulin resistance, a loss of motor coordination, deficits in novel object exploration and recognition, increased helplessness, dyslipidemia, as well as signs of a non-alcoholic steatohepatitis (NASH)-like syndrome: liver steatosis and increased liver injury markers. Importantly, these changes were accompanied by decreased SERT expression, elevated numbers of microglia cells and malondialdehyde levels in, and restricted to, the prefrontal cortex. SIGNIFICANCE The WD induces a spectrum of behaviors that are more reminiscent of ADHD and ASD than previously recognized and suggests that, in addition to the impairment of impulsivity and sociability, the consumption of a WD might be expected to exacerbate motor dysfunction that is also known to be associated with adult ADHD and ASD.
Collapse
Affiliation(s)
- Ekaterina Veniaminova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia
| | - Margarita Oplatchikova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia
| | - Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, 4000 Liège, Belgium
| | - Elena Kotenkova
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Tallalikhina Str. 26, 109316 Moscow, Russia
| | - Alexander Lysko
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia
| | - Ekaterina Vasilevskaya
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Tallalikhina Str. 26, 109316 Moscow, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, 400715 Chongqing, China; Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya Nab. 7-9, 199034 St. Petersburg, Russia; Ural Federal University, Mira Str. 19, 620002 Ekaterinburg, Russia
| | - Liliya Fedulova
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Tallalikhina Str. 26, 109316 Moscow, Russia
| | - Aleksei Umriukhin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia; Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080 Würzburg, Germany
| | - Daniel C Anthony
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia; Department of Pharmacology, Oxford University, Mansfield Road, OX1 3QT Oxford, UK
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia; Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080 Würzburg, Germany.
| |
Collapse
|
25
|
Miron I, Dumitrascu D. GASTROINTESTINAL MOTILITY DISORDERS IN OBESITY. ACTA ENDOCRINOLOGICA (BUCHAREST, ROMANIA : 2005) 2019; 15:497-504. [PMID: 32377248 PMCID: PMC7200119 DOI: 10.4183/aeb.2019.497] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal (GI) motility, which is important for the digestion and absorption, may be altered in obesity. The aim of this review is to present the GI motility changes occurring in obesity, as well as their underlying mechanisms. We have conducted a systematic review of the published literature concerning GI motility and obesity and have described recent published data on the changes throughout the entire GI tract. Most recent discoveries include evidence supporting the increase of gastroesophageal reflux disease in obesity and inhibition of gastric motility. Intestinal transit of the distal small bowel generally slows down, ensuring enough time for digestion and absorption. Constipation is more frequent in obese patients than in those with a normal weight. The gut-brain axis plays an important role in the pathophysiology of GI motility disorders in obesity. This bidirectional communication is achieved by way of neurons, hormones, metabolites derived from intestinal microbiota and cytokines. The molecular mechanisms of GI motility changes in obesity are complex. Current data offer a starting point for further research needed to clarify the association of obesity with GI motility disorders.
Collapse
Affiliation(s)
- I. Miron
- “Iuliu Hatieganu” University of Medicine and Pharmacy, 3 Medical Clinic, Cluj-Napoca, Romania
| | - D.L. Dumitrascu
- “Iuliu Hatieganu” Dept of Internal Medicine, Cluj-Napoca, Romania
| |
Collapse
|
26
|
Yabut JM, Crane JD, Green AE, Keating DJ, Khan WI, Steinberg GR. Emerging Roles for Serotonin in Regulating Metabolism: New Implications for an Ancient Molecule. Endocr Rev 2019; 40:1092-1107. [PMID: 30901029 PMCID: PMC6624793 DOI: 10.1210/er.2018-00283] [Citation(s) in RCA: 240] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
Serotonin is a phylogenetically ancient biogenic amine that has played an integral role in maintaining energy homeostasis for billions of years. In mammals, serotonin produced within the central nervous system regulates behavior, suppresses appetite, and promotes energy expenditure by increasing sympathetic drive to brown adipose tissue. In addition to these central circuits, emerging evidence also suggests an important role for peripheral serotonin as a factor that enhances nutrient absorption and storage. Specifically, glucose and fatty acids stimulate the release of serotonin from the duodenum, promoting gut peristalsis and nutrient absorption. Serotonin also enters the bloodstream and interacts with multiple organs, priming the body for energy storage by promoting insulin secretion and de novo lipogenesis in the liver and white adipose tissue, while reducing lipolysis and the metabolic activity of brown and beige adipose tissue. Collectively, peripheral serotonin acts as an endocrine factor to promote the efficient storage of energy by upregulating lipid anabolism. Pharmacological inhibition of serotonin synthesis or signaling in key metabolic tissues are potential drug targets for obesity, type 2 diabetes, and nonalcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Julian M Yabut
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Justin D Crane
- Department of Biology, Northeastern University, Boston, Massachusetts
| | - Alexander E Green
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Damien J Keating
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Waliul I Khan
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
27
|
Mao Z, Lin H, Su W, Li J, Zhou M, Li Z, Zhou B, Yang Q, Zhou M, Pan K, He J, Zhang W. Deficiency of ZnT8 Promotes Adiposity and Metabolic Dysfunction by Increasing Peripheral Serotonin Production. Diabetes 2019; 68:1197-1209. [PMID: 30936149 DOI: 10.2337/db18-1321] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/04/2019] [Indexed: 02/05/2023]
Abstract
ZnT8 is a zinc transporter enriched in pancreatic β-cells, and its polymorphism is associated with increased susceptibility to type 2 diabetes. However, the exact role of ZnT8 in systemic energy metabolism remains elusive. In this study, we found that ZnT8 knockout mice displayed increased adiposity without obvious weight gain. We also observed that the intestinal tract morphology, motility, and gut microbiota were changed in ZnT8 knockout mice. Further study demonstrated that ZnT8 was expressed in enteroendocrine cells, especially in 5-hydroxytryptamine (5-HT)-positive enterochromaffin cells. Lack of ZnT8 resulted in an elevated circulating 5-HT level owing to enhanced expression of tryptophan hydroxylase 1. Blocking 5-HT synthesis in ZnT8-deficient mice restored adiposity, high-fat diet-induced obesity, and glucose intolerance. Moreover, overexpression of human ZnT8 diabetes high-risk allele R325W increased 5-HT levels relative to the low-risk allele in RIN14B cells. Our study revealed an unexpected role of ZnT8 in regulating peripheral 5-HT biogenesis and intestinal microenvironment, which might contribute to the increased risk of obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Zhuo Mao
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Hui Lin
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Wen Su
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Jinghui Li
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Minsi Zhou
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Zhuoran Li
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Beibei Zhou
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Qing Yang
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Mingyan Zhou
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Ke Pan
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Jinhan He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Weizhen Zhang
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
- Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, China
| |
Collapse
|
28
|
Ishida Y, Sugiura Y, Magome T, Kamakura T, Takimoto Y, Hanada Y, Kitayama K, Nakamura Y, Shimada S, Ohta N, Naono-Nakayama R, Kamijo K. Expression Analysis of Serotonin Receptors, Serotonin Transporter and l-Amino Acid Decarboxylase in the Mouse Sphenopalatine Ganglion by RT-PCR, Northern Blot Analysis and In Situ Hybridization. Neuroscience 2019; 411:23-36. [PMID: 31128160 DOI: 10.1016/j.neuroscience.2019.05.028] [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: 01/19/2019] [Revised: 05/03/2019] [Accepted: 05/14/2019] [Indexed: 11/26/2022]
Abstract
The sphenopalatine ganglion (SPG) is a gathering of the cell bodies of parasympathetic fibers that dominate the nasal gland, lacrimal gland and cerebral blood vessels. The SPG controls nasal secretions, tears, and the dilation of cerebral blood vessels. However, it is unclear how serotonin regulates SPG functions. In this study, we investigated the expression of genes involved in the serotonergic system in the mouse SPG. We examined the mRNA expression levels of 5-HT1A, 5-HT1B, 5-HT1D, 5-HT1F, 5-HT2A, 5-HT2B, 5-HT2C, 5-HT3A, 5-HT3B, 5-HT4, 5-HT5A, 5-HT5B, 5-HT6 and 5-HT7 receptors, as well as serotonin transporter, tryptophan hydroxylases 1 and 2, and L-amino acid decarboxylase (AADC) by RT-PCR. It revealed that the 5-HT3A and 5-HT3B ionotropic receptors and AADC were likely to be highly expressed in the SPG, as measured by RT-PCR. We next performed in situ hybridization on the SPG to examine the expression of these three genes at the cellular level after validating the specificity of each cRNA probe by northern blotting. The 5-HT3A receptor, 5-HT3B receptor, and AADC were expressed in 96.5% ± 1.0%, 29.7% ± 10.7%, and 57.4% ± 2.9% of neuronal cell bodies in the SPG, respectively, indicating that the 5-HT3A receptor was virtually expressed in all SPG neurons. Our results on the expression of these critical serotonin system genes in the parasympathetic SPG provide insight into the pathogenetics of rhinitis, conjunctivitis and headache. Furthermore, our findings suggest that targeting the 5-HT3A receptor might have therapeutic potential in the treatment of these ailments.
Collapse
Affiliation(s)
- Yusuke Ishida
- Division of Anatomy and Cell Biology, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan; Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Division of Otorhinolaryngology, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan.
| | - Yusuke Sugiura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Takuya Magome
- Department of medicine for Sports and Performing Arts, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Takefumi Kamakura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yasumitsu Takimoto
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yukiko Hanada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Otorhinolaryngology-Head and Neck Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Kazuki Kitayama
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yukiko Nakamura
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Nobuo Ohta
- Division of Otorhinolaryngology, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| | - Rumi Naono-Nakayama
- Division of Anatomy and Cell Biology, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| | - Keiju Kamijo
- Division of Anatomy and Cell Biology, Tohoku Medical and Pharmaceutical University, 1-15-1 Fukumuro, Miyagino-ku, Sendai 983-8536, Japan
| |
Collapse
|
29
|
Nyavor Y, Estill R, Edwards H, Ogden H, Heideman K, Starks K, Miller C, May G, Flesch L, McMillan J, Gericke M, Forney L, Balemba O. Intestinal nerve cell injury occurs prior to insulin resistance in female mice ingesting a high-fat diet. Cell Tissue Res 2019; 376:325-340. [PMID: 30778729 DOI: 10.1007/s00441-019-03002-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 01/30/2019] [Indexed: 12/11/2022]
Abstract
Diabetic patients suffer from gastrointestinal disorders associated with dysmotility, enteric neuropathy and dysbiosis of gut microbiota; however, gender differences are not fully known. Previous studies have shown that a high-fat diet (HFD) causes type two diabetes (T2D) in male mice after 4-8 weeks but only does so in female mice after 16 weeks. This study seeks to determine whether sex influences the development of intestinal dysmotility, enteric neuropathy and dysbiosis in mice fed HFD. We fed 8-week-old C57BL6 male and female mice a standard chow diet (SCD) or a 72% kcal HFD for 8 weeks. We analyzed the associations between sex and intestinal dysmotility, neuropathy and dysbiosis using motility assays, immunohistochemistry and next-generation sequencing. HFD ingestion caused obesity, glucose intolerance and insulin resistance in male but not female mice. However, HFD ingestion slowed intestinal propulsive motility in both male and female mice. This was associated with decreased inhibitory neuromuscular transmission, loss of myenteric inhibitory motor neurons and axonal swelling and loss of cytoskeletal filaments. HFD induced dysbiosis and changed the abundance of specific bacteria, especially Allobaculum, Bifidobacterium and Lactobacillus, which correlated with dysmotility and neuropathy. Female mice had higher immunoreactivity and numbers of myenteric inhibitory motor neurons, matching larger amplitudes of inhibitory junction potentials. This study suggests that sex influences the development of HFD-induced metabolic syndrome but dysmotility, neuropathy and dysbiosis occur independent of sex and prior to T2D conditions. Gastrointestinal dysmotility, neuropathy and dysbiosis might play a crucial role in the pathophysiology of T2D in humans irrespective of sex.
Collapse
Affiliation(s)
- Yvonne Nyavor
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Rachel Estill
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Hannah Edwards
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Hailey Ogden
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Kaila Heideman
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Kiefer Starks
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Christopher Miller
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - George May
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Lance Flesch
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - John McMillan
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Martin Gericke
- Institute for Anatomy, University of Leipzig, Liebigstraße 13, 04103, Leipzig, Germany
| | - Larry Forney
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA
| | - Onesmo Balemba
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive, LSS 252, Moscow, ID, 83844, USA.
| |
Collapse
|
30
|
Clostridium ramosum regulates enterochromaffin cell development and serotonin release. Sci Rep 2019; 9:1177. [PMID: 30718836 PMCID: PMC6362283 DOI: 10.1038/s41598-018-38018-z] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/14/2018] [Indexed: 12/22/2022] Open
Abstract
Peripheral serotonin (5-hydroxytryptamine: 5-HT) synthesized in the intestine by enterochromaffin cells (ECs), plays an important role in the regulation of peristaltic of the gut, epithelial secretion and promotes the development and maintenance of the enteric neurons. Recent studies showed that the indigenous gut microbiota modulates 5-HT signalling and that ECs use sensory receptors to detect dietary and microbiota-derived signals from the lumen to subsequently transduce the information to the nervous system. We hypothesized that Clostridium ramosum by increasing gut 5-HT availability consequently contributes to high-fat diet-induced obesity. Using germ-free mice and mice monoassociated with C. ramosum, intestinal cell lines and mouse organoids, we demonstrated that bacterial cell components stimulate host 5-HT secretion and program the differentiation of colonic intestinal stem progenitors toward the secretory 5-HT-producing lineage. An elevated 5-HT level regulates the expression of major proteins involved in intestinal fatty acid absorption in vitro, suggesting that the presence of C. ramosum in the gut promotes 5-HT secretion and thereby could facilitates intestinal lipid absorption and the development of obesity.
Collapse
|
31
|
Sugar Responses of Human Enterochromaffin Cells Depend on Gut Region, Sex, and Body Mass. Nutrients 2019; 11:nu11020234. [PMID: 30678223 PMCID: PMC6412251 DOI: 10.3390/nu11020234] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 12/15/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Gut-derived serotonin (5-HT) is released from enterochromaffin (EC) cells in response to nutrient cues, and acts to slow gastric emptying and modulate gastric motility. Rodent studies also evidence a role for gut-derived 5-HT in the control of hepatic glucose production, lipolysis and thermogenesis, and in mediating diet-induced obesity. EC cell number and 5-HT content is increased in the small intestine of obese rodents and human, however, it is unknown whether EC cells respond directly to glucose in humans, and whether their capacity to release 5-HT is perturbed in obesity. We therefore investigated 5-HT release from human duodenal and colonic EC cells in response to glucose, sucrose, fructose and α-glucoside (αMG) in relation to body mass index (BMI). EC cells released 5-HT only in response to 100 and 300 mM glucose (duodenum) and 300 mM glucose (colon), independently of osmolarity. Duodenal, but not colonic, EC cells also released 5-HT in response to sucrose and αMG, but did not respond to fructose. 5-HT content was similar in all EC cells in males, and colonic EC cells in females, but 3 to 4-fold higher in duodenal EC cells from overweight females (p < 0.05 compared to lean, obese). Glucose-evoked 5-HT release was 3-fold higher in the duodenum of overweight females (p < 0.05, compared to obese), but absent here in overweight males. Our data demonstrate that primary human EC cells respond directly to dietary glucose cues, with regional differences in selectivity for other sugars. Augmented glucose-evoked 5-HT release from duodenal EC is a feature of overweight females, and may be an early determinant of obesity.
Collapse
|
32
|
Hajjarzadeh S, Mahdavi R, Shalilahmadi D, Nikniaz Z. The association of dietary patterns with migraine attack frequency in migrainous women. Nutr Neurosci 2018; 23:724-730. [DOI: 10.1080/1028415x.2018.1550890] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Samaneh Hajjarzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Mahdavi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Zeinab Nikniaz
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Duan Y, Zeng L, Zheng C, Song B, Li F, Kong X, Xu K. Inflammatory Links Between High Fat Diets and Diseases. Front Immunol 2018; 9:2649. [PMID: 30483273 PMCID: PMC6243058 DOI: 10.3389/fimmu.2018.02649] [Citation(s) in RCA: 302] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, chronic overnutrition, such as consumption of a high-fat diet (HFD), has been increasingly viewed as a significant modifiable risk factor for diseases such as diabetes and certain types of cancer. However, the mechanisms by which HFDs exert adverse effects on human health remains poorly understood. Here, this paper will review the recent scientific literature about HFD-induced inflammation and subsequent development of diseases and cancer, with an emphasis on mechanisms involved. Given the expanding global epidemic of excessive HFD intake, understanding the impacts of a HFD on these medical conditions, gaining great insights into possible underlying mechanisms, and developing effective therapeutic strategies are of great importance.
Collapse
Affiliation(s)
- Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Liming Zeng
- Science College of Jiangxi Agricultural University, Nanchang, China
| | - Changbing Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| | - Bo Song
- Guangdong Provincial Key Laboratory of Animal Nutrition Regulation, South China Agricultural University, Guangzhou, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Xiangfeng Kong
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| | - Kang Xu
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, China
| |
Collapse
|
34
|
Young RL, Lumsden AL, Martin AM, Schober G, Pezos N, Thazhath SS, Isaacs NJ, Cvijanovic N, Sun EWL, Wu T, Rayner CK, Nguyen NQ, Fontgalland DD, Rabbitt P, Hollington P, Sposato L, Due SL, Wattchow DA, Liou AP, Jackson VM, Keating DJ. Augmented capacity for peripheral serotonin release in human obesity. Int J Obes (Lond) 2018; 42:1880-1889. [PMID: 29568107 DOI: 10.1038/s41366-018-0047-8] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/08/2018] [Accepted: 01/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND/OBJECTIVES Evidence from animal studies highlights an important role for serotonin (5-HT), derived from gut enterochromaffin (EC) cells, in regulating hepatic glucose production, lipolysis and thermogenesis, and promoting obesity and dysglycemia. Evidence in humans is limited, although elevated plasma 5-HT concentrations are linked to obesity. SUBJECTS/METHODS We assessed (i) plasma 5-HT concentrations before and during intraduodenal glucose infusion (4 kcal/min for 30 min) in non-diabetic obese (BMI 44 ± 4 kg/m2, N = 14) and control (BMI 24 ± 1 kg/m2, N = 10) subjects, (ii) functional activation of duodenal EC cells (immunodetection of phospho-extracellular related-kinase, pERK) in response to glucose, and in separate subjects, (iii) expression of tryptophan hydroxylase-1 (TPH1) in duodenum and colon (N = 39), and (iv) 5-HT content in primary EC cells from these regions (N = 85). RESULTS Plasma 5-HT was twofold higher in obese than control responders prior to (P = 0.025), and during (iAUC, P = 0.009), intraduodenal glucose infusion, and related positively to BMI (R2 = 0.334, P = 0.003) and HbA1c (R2 = 0.508, P = 0.009). The density of EC cells in the duodenum was twofold higher at baseline in obese subjects than controls (P = 0.023), with twofold more EC cells activated by glucose infusion in the obese (EC cells co-expressing 5-HT and pERK, P = 0.001), while the 5-HT content of EC cells in duodenum and colon was similar; TPH1 expression was 1.4-fold higher in the duodenum of obese subjects (P = 0.044), and related positively to BMI (R2 = 0.310, P = 0.031). CONCLUSIONS Human obesity is characterized by an increased capacity to produce and release 5-HT from the proximal small intestine, which is strongly linked to higher body mass, and glycemic control. Gut-derived 5-HT is likely to be an important driver of pathogenesis in human obesity and dysglycemia.
Collapse
Affiliation(s)
- Richard L Young
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Amanda L Lumsden
- Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia
| | - Alyce M Martin
- Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia
| | - Gudrun Schober
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Nektaria Pezos
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Sony S Thazhath
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Nicole J Isaacs
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Nada Cvijanovic
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| | - Emily W L Sun
- Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia
| | - Tongzhi Wu
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Christopher K Rayner
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Nam Q Nguyen
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Dayan de Fontgalland
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Philippa Rabbitt
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Paul Hollington
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Luigi Sposato
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Steven L Due
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - David A Wattchow
- Department of Surgery, Flinders Medical Centre, Bedford Park, SA, 5042, Australia
| | - Alice P Liou
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, 02139, USA
| | - V Margaret Jackson
- Cardiovascular, Metabolic, and Endocrine Diseases Research Unit, Pfizer Worldwide Research and Development, Cambridge, MA, 02139, USA
| | - Damien J Keating
- Nutrition & Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia.
- Centre for Neuroscience & Department of Human Physiology, Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
35
|
Kim JW, Lee YS, Seol DJ, Cho IJ, Ku SK, Choi JS, Lee HJ. Anti-obesity and fatty liver-preventing activities of Lonicera caerulea in high-fat diet-fed mice. Int J Mol Med 2018; 42:3047-3064. [PMID: 30221679 PMCID: PMC6202101 DOI: 10.3892/ijmm.2018.3879] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 08/27/2018] [Indexed: 12/26/2022] Open
Abstract
Blue honeysuckle (BH, Lonicera caerulea) is used as a traditional medicine in Russia, Japan and China, but is not commonly considered as an edible berry in Europe, USA or Korea. BH has been revealed to decrease serum cholesterol and triacylglycerol (triglyceride or TG) levels through the activation of AMP-activated protein kinase (AMPK), thus it is expected to be a health functional food and pharmaceutical agent for the prevention of non-alcoholic liver damage, in addition to effects as a suppressor of hyperlipidemia and as an anti-obesity agent. In the present study, the pharmacological activity of BH extract (BHe) was observed in high-fat diet (HFD)-fed mice. Significant increases in fat pad weight, body weight, fat accumulation (body and abdominal fat density, and thickness of the periovarian and abdominal wall) and serum biochemical levels (aspartate transaminase, alanine amino-transferase, alkaline phosphatase, lactate dehydrogenase, γ-glutamyltransferase, total cholesterol, low-density lipoprotein and TG, with the exception of high-density lipoprotein) were observed in HFD-fed mice. In addition, increases in adipocyte hypertrophy, the area of steatohepatitis and hepatocyte hypertrophy were observed, whereas decreased zymogen content was identified upon histopathological observation. Increased deterioration of the endogenous antioxidant defense system (liver catalase, glutathione and superoxide dismutase) and hepatic lipid peroxidation was observed. In addition, there were decreases in hepatic glucokinase activity, AMPKα1 and AMPKα2 mRNA expression, adipose tissue uncoupling protein 2 expression, and adiponectin mRNA expression, increases in phosphoenolpyruvate carboxykinase and glucose-6-phosphatase activity, hepatic acetyl-CoA carboxylase 1 mRNA expression, and the expression of leptin, CCAAT/enhancer-binding protein (C/EBP) α, C/EBPβ and sterol-regulatory-element-binding protein 1c mRNA in the periovarian tissue. Furthermore, non-alcoholic fatty liver disease (NAFLD) and obesity were significantly inhibited by the continuous administration of BHe for 84 days. These results revealed that BHe may be a promising novel drug or functional food candidate for the treatment of obesity and NAFLD.
Collapse
Affiliation(s)
- Joo Wan Kim
- Aribio Co. Ltd., Seongnam, Gyeonggi 13487, Republic of Korea
| | - You-Suk Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi 13120, Republic of Korea
| | - Du Jin Seol
- Aribio Co. Ltd., Seongnam, Gyeonggi 13487, Republic of Korea
| | - Il Je Cho
- The Medical Research Center for Globalization of Herbal Formulation and Department of Herbal Formulation, College of Oriental Medicine, Gyeongsan, Gyeongsangbuk 38610, Republic of Korea
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Korean Medicine, Daegu Haany University, Gyeongsan, Gyeongsangbuk 38610, Republic of Korea
| | - Jae-Suk Choi
- Division of Bioindustry, College of Medical and Life Sciences, Silla University, Sasang, Busan 46958, Republic of Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam, Gyeonggi 13120, Republic of Korea
| |
Collapse
|
36
|
Antonioli L, Caputi V, Fornai M, Pellegrini C, Gentile D, Giron MC, Orso G, Bernardini N, Segnani C, Ippolito C, Csóka B, Haskó G, Németh ZH, Scarpignato C, Blandizzi C, Colucci R. Interplay between colonic inflammation and tachykininergic pathways in the onset of colonic dysmotility in a mouse model of diet-induced obesity. Int J Obes (Lond) 2018; 43:331-343. [PMID: 30082748 DOI: 10.1038/s41366-018-0166-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 05/24/2018] [Accepted: 06/24/2018] [Indexed: 12/15/2022]
Abstract
BACKGROUND The murine model of high fat diet (HFD)-induced obesity is characterized by an increment of intestinal permeability, secondary to an impairment of mucosal epithelial barrier and enteric inflammation, followed by morphofunctional rearrangement of the enteric nervous system. The present study investigated the involvement of abdominal macrophages in the mechanisms underlying the development of enteric dysmotility associated with obesity. METHODS Wild type C57BL/6J mice were fed with HFD (60% kcal from fat) or normocaloric diet (NCD, 18% kcal from fat) for 8 weeks. Groups of mice fed with NCD or HFD were treated with clodronate encapsulated into liposomes to deplete abdominal macrophages. Tachykininergic contractions, elicited by electrical stimulation or exogenous substance P (SP), were recorded in vitro from longitudinal muscle colonic preparations. Substance P distribution was examined by confocal immunohistochemistry. The density of macrophages in the colonic wall was examined by immunohistochemical analysis. Malondialdehyde (MDA, colorimetric assay) and IL-1β (ELISA assay) levels were also evaluated. RESULTS MDA and IL-1β levels were increased in colonic tissues from HFD-treated animals. In colonic preparations, electrically evoked tachykininergic contractions were enhanced in HFD mice. Immunohistochemistry displayed an increase in substance P immunoreactivity in myenteric ganglia, as well as in the muscular layers of colonic cryosections from obese mice. Macrophage depletion in HFD mice was associated with a significant reduction of colonic inflammation. In addition, the decrease in macrophage density attenuated the morphofunctional alterations of tachykininergic pathways observed in obese mice. CONCLUSION Obesity elicited by HFD determines a condition of colonic inflammation, followed by a marked rearrangement of motor excitatory tachykininergic enteric nerves. Macrophage depletion counteracted the morphofunctional changes of colonic neuromuscular compartment, suggesting a critical role for these immune cells in the onset of enteric dysmotility associated with obesity.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Department of Anesthesiology, Columbia University, New York, NY, 10032, USA
| | - Valentina Caputi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.,San Camillo Hospital, Treviso, Italy.,APC Microbiome Ireland, University College Cork, T12YT20 Cork, Ireland
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Daniela Gentile
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Genny Orso
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini-, Lecco, Italy
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Cristina Segnani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Balázs Csóka
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA
| | - Zoltán H Németh
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA.,Department of Surgery, Morristown Medical Center, Morristown, NJ, 07960, USA
| | | | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
37
|
Bliss ES, Whiteside E. The Gut-Brain Axis, the Human Gut Microbiota and Their Integration in the Development of Obesity. Front Physiol 2018; 9:900. [PMID: 30050464 PMCID: PMC6052131 DOI: 10.3389/fphys.2018.00900] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022] Open
Abstract
Obesity is a global epidemic, placing socioeconomic strain on public healthcare systems, especially within the so-called Western countries, such as Australia, United States, United Kingdom, and Canada. Obesity results from an imbalance between energy intake and energy expenditure, where energy intake exceeds expenditure. Current non-invasive treatments lack efficacy in combating obesity, suggesting that obesity is a multi-faceted and more complex disease than previously thought. This has led to an increase in research exploring energy homeostasis and the discovery of a complex bidirectional communication axis referred to as the gut-brain axis. The gut-brain axis is comprised of various neurohumoral components that allow the gut and brain to communicate with each other. Communication occurs within the axis via local, paracrine and/or endocrine mechanisms involving a variety of gut-derived peptides produced from enteroendocrine cells (EECs), including glucagon-like peptide 1 (GLP1), cholecystokinin (CCK), peptide YY3-36 (PYY), pancreatic polypeptide (PP), and oxyntomodulin. Neural networks, such as the enteric nervous system (ENS) and vagus nerve also convey information within the gut-brain axis. Emerging evidence suggests the human gut microbiota, a complex ecosystem residing in the gastrointestinal tract (GIT), may influence weight-gain through several inter-dependent pathways including energy harvesting, short-chain fatty-acids (SCFA) signalling, behaviour modifications, controlling satiety and modulating inflammatory responses within the host. Hence, the gut-brain axis, the microbiota and the link between these elements and the role each plays in either promoting or regulating energy and thereby contributing to obesity will be explored in this review.
Collapse
Affiliation(s)
- Edward S. Bliss
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | | |
Collapse
|
38
|
Banskota S, Ghia JE, Khan WI. Serotonin in the gut: Blessing or a curse. Biochimie 2018; 161:56-64. [PMID: 29909048 DOI: 10.1016/j.biochi.2018.06.008] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/12/2018] [Indexed: 12/25/2022]
Abstract
Serotonin (5-hydroxytryptamine or 5-HT) once most extensively studied as a neurotransmitter of the central nervous system, is seen to be predominantly secreted in the gut. About 95% of 5-HT is estimated to be found in gut mainly within the enterochromaffin cells whereas about 5% is found in the brain. 5-HT is an important enteric signaling molecule and is well known for playing a key role in sensory-motor and secretory functions in the gut. In recent times, studies uncovering various new functions of gut-derived 5-HT indicate that many more are yet to be discovered in coming days. Recent studies revealed that 5-HT plays a pivotal role in immune cell activation and generation/perpetuation of inflammation in the gut. In addition to its various roles in the gut, there are now emerging evidences that suggest an important role of gut-derived 5-HT in other biological processes beyond the gut, such as bone remodeling and metabolic homeostasis. This review focuses to briefly summarize the accumulated and newly updated role of 5-HT in the maintenance of normal gut physiology and in the pathogenesis of inflammation in the gut. The collected information about this multifaceted signaling molecule may aid in distinguishing its good and bad effects which may lead to the development of novel strategies to overcome the unwanted effect, such as in inflammatory bowel disease.
Collapse
Affiliation(s)
- Suhrid Banskota
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Jean-Eric Ghia
- Department of Immunology and Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Waliul I Khan
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
39
|
Steinberg GR. Cellular Energy Sensing and Metabolism-Implications for Treating Diabetes: The 2017 Outstanding Scientific Achievement Award Lecture. Diabetes 2018; 67:169-179. [PMID: 29358486 DOI: 10.2337/dbi17-0039] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/08/2017] [Indexed: 11/13/2022]
Abstract
The Outstanding Scientific Achievement Award recognizes distinguished scientific achievement in the field of diabetes, taking into consideration independence of thought and originality. Gregory R. Steinberg, PhD, professor of medicine, Canada Research Chair, J. Bruce Duncan Endowed Chair in Metabolic Diseases, and codirector of the Metabolism and Childhood Obesity Research Program at McMaster University, Hamilton, Ontario, Canada, received the prestigious award at the American Diabetes Association's 77th Scientific Sessions, 9-13 June 2017, in San Diego, CA. He presented the Outstanding Scientific Achievement Award Lecture, "Cellular Energy Sensing and Metabolism-Implications for Treating Diabetes," on Monday, 12 June 2017.The survival of all cells is dependent on the constant challenge to match energetic demands with nutrient availability, a task that is mediated through a highly conserved network of metabolic fuel sensors that orchestrate both cellular and whole-organism energy balance. A mismatch between cellular energy demand and nutrient availability is a key factor contributing to the development of type 2 diabetes; thus, understanding the fundamental mechanisms by which cells sense nutrient availability and demand may lead to the development of new treatments. Glucose-lowering therapies, such as caloric restriction, exercise, and metformin, all induce an energetic challenge that results in the activation of the cellular energy sensor AMP-activated protein kinase (AMPK). Activation of AMPK in turn suppresses lipid synthesis and inflammation while increasing glucose uptake, fatty acid oxidation, and mitochondrial function. In contrast, high levels of nutrient availability suppress AMPK activity while also increasing the production of peripheral serotonin, a gut-derived endocrine factor that suppresses β-adrenergic-induced activation of brown adipose tissue. Identifying new ways to manipulate these two ancient fuel gauges by activating AMPK and inhibiting peripheral serotonin may lead to the development of new therapies for treating type 2 diabetes.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/metabolism
- Adipose Tissue, Beige/drug effects
- Adipose Tissue, Beige/metabolism
- Adipose Tissue, Beige/pathology
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Animals
- Awards and Prizes
- Caloric Restriction
- Cell Survival/drug effects
- Combined Modality Therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Diabetes Mellitus, Type 2/prevention & control
- Diabetes Mellitus, Type 2/therapy
- Endocrinology
- Energy Intake/drug effects
- Energy Metabolism/drug effects
- Enzyme Activation/drug effects
- Exercise
- Feedback, Physiological/drug effects
- Humans
- Hypoglycemic Agents/therapeutic use
- Insulin Resistance
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Models, Biological
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Serotonin/blood
- Serotonin/metabolism
Collapse
Affiliation(s)
- Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
40
|
Dong Y, Wang Z, Qin Z, Cao J, Chen Y. Role of serotonin in the intestinal mucosal epithelium barrier in weaning mice undergoing stress-induced diarrhea. J Mol Histol 2017; 49:85-97. [PMID: 29260435 DOI: 10.1007/s10735-017-9749-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022]
Abstract
Stress-induced diarrhea is a frequent and challenging threat to humans and domestic animals. Serotonin (5-HT) has been shown to be involved in the pathological process of stress-induced diarrhea. However, the role of 5-HT in stress-induced diarrhea remains unclear. A stress-induced diarrhea model was established in 21-day-old ICR weaning mice through an intragastric administration of 0.25 mL of 0.4 g/mL folium sennae and restraint of the hind legs with adhesive tape for 4 h to determine whether 5-HT regulates the mucosal barrier to cause diarrhea. Mice with decreased levels of 5-HT were pretreated with an intraperitoneal injection of 300 mg/kg p-chlorophenylalanine (PCPA), a 5-HT synthesis inhibitor. After 5 days of treatment, the stress level, body weight and intestinal mucosal morphology indexes were measured. Compared to the controls, the mice with stress-induced diarrhea displayed a stress reaction, with increased corticosterone levels, as well as increased 5-HT-positive cells. However, the mice with stress-induced diarrhea exhibited decreased body weights, villus height to crypt depth ratios (V/C), and Occludin and Claudin1 expression. The PCPA injection reversed these effects in mice with different degrees of stress-induced diarrhea. Based on these findings, inhibition of 5-HT synthesis relieved the stress response and improved the health of the intestinal tract, including both the intestinal absorption capacity, as determined by the villus height and crypt depth, and the mucosal barrier function, as determined by the tight junction proteins of epithelial cell.
Collapse
Affiliation(s)
- Yulan Dong
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China
| | - Zixu Wang
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China
| | - Zhuoming Qin
- Institute of Poultry, Shandong Academy of Agricultural Sciences, Jinan, 250100, People's Republic of China
| | - Jing Cao
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China
| | - Yaoxing Chen
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China.
| |
Collapse
|
41
|
Patel N, Fagan-Murphy A, Covill D, Patel BA. 3D Printed Molds Encompassing Carbon Composite Electrodes To Conduct Multisite Monitoring in the Entire Colon. Anal Chem 2017; 89:11690-11696. [DOI: 10.1021/acs.analchem.7b03148] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Nirav Patel
- School
of Pharmacy and Biomolecular Sciences, ‡Centre for Stress and Age-Related
Diseases, and §School of Computing, Engineering and Mathematics, University of Brighton, Brighton BN2 4AT, U.K
| | - Aidan Fagan-Murphy
- School
of Pharmacy and Biomolecular Sciences, ‡Centre for Stress and Age-Related
Diseases, and §School of Computing, Engineering and Mathematics, University of Brighton, Brighton BN2 4AT, U.K
| | - Derek Covill
- School
of Pharmacy and Biomolecular Sciences, ‡Centre for Stress and Age-Related
Diseases, and §School of Computing, Engineering and Mathematics, University of Brighton, Brighton BN2 4AT, U.K
| | - Bhavik Anil Patel
- School
of Pharmacy and Biomolecular Sciences, ‡Centre for Stress and Age-Related
Diseases, and §School of Computing, Engineering and Mathematics, University of Brighton, Brighton BN2 4AT, U.K
| |
Collapse
|
42
|
Kim MR, Kim JW, Park JB, Hong YK, Ku SK, Choi JS. Anti-obesity effects of yellow catfish protein hydrolysate on mice fed a 45% kcal high-fat diet. Int J Mol Med 2017; 40:784-800. [PMID: 28713910 PMCID: PMC5548026 DOI: 10.3892/ijmm.2017.3063] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 06/26/2017] [Indexed: 11/25/2022] Open
Abstract
Obesity contributes to the etiologies of a variety of comorbid conditions, such as type 2 diabetes, hypertension and cardiovascular disease. In the present study, the anti-obesity effects of yellow catfish protein hydrolysate (YPh) were observed in mice fed a 45% kcal high-fat diet (HFD) compared with those of mice treated with simvastatin. The HFD-fed control mice exhibited noticeable increase in body weight, and whole-body and abdominal fat densities, periovarian and abdominal wall-deposited fat pad weight, as well as in the levels of triglycerides (TG), blood total cholesterol (TC), low-density lipoprotein, alanine aminotransferase, aspartate aminotransferase, creatinine, blood urea nitrogen, and in the fecal TG and TC contents. However, they exhibited a decrease in serum high-density lipoprotein levels. In addition, an increase was detected in periovarian and dorsal abdominally deposited fat pad thickness, adipocyte hypertrophy, the number of steatohepatitis regions, hepatocyte hypertrophy and lipid droplet deposition-related renal tubular vacuolation degenerative lesions, along with increased hepatic lipid peroxidation and a deteriorated endogenous antioxidant defense system (glutathione, catalase and superoxide dismutase). However, all the above-mentioned obesity-related complications were dose-dependently and significantly inhibited after 84 days of thye consecutive oral administration of 125, 250 and 500 mg/kg YPh. In addition, YPh dose-dependently depleted the liver endogenous antioxidant defense system and inhibited hepatic lipid peroxidation. Overall, the effects of 250 mg/kg YPh on HFD-induced obesity and related complications were similar or more potent than those of 10 mg/kg simvastatin. These results indicate that YPh is a promising new potent medicinal ingredient for possible use in the treatment of obesity and related complications.
Collapse
Affiliation(s)
- Mi-Ryung Kim
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Busan 46958
| | - Joo-Wan Kim
- Aribio Inc., Byeoksan Digital Valley, Yeongdeungpo-gu, Seoul 07286
| | - Jeong Been Park
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Busan 46958
| | - Yong-Ki Hong
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan 48513
| | - Sae Kwang Ku
- Department of Anatomy and Histology, College of Oriental Medicine, Daegu Haany University, Gyeongsan-si, Gyeongsangbuk-do 38610, Republic of Korea
| | - Jae-Suk Choi
- Major in Food Biotechnology, Division of Bioindustry, College of Medical and Life Sciences, Silla University, Busan 46958
| |
Collapse
|
43
|
Antonioli L, Pellegrini C, Fornai M, Tirotta E, Gentile D, Benvenuti L, Giron MC, Caputi V, Marsilio I, Orso G, Bernardini N, Segnani C, Ippolito C, Csóka B, Németh ZH, Haskó G, Scarpignato C, Blandizzi C, Colucci R. Colonic motor dysfunctions in a mouse model of high-fat diet-induced obesity: an involvement of A 2B adenosine receptors. Purinergic Signal 2017; 13:497-510. [PMID: 28808842 DOI: 10.1007/s11302-017-9577-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/01/2017] [Indexed: 12/13/2022] Open
Abstract
Adenosine A2B receptors (A2BR) regulate several enteric functions. However, their implication in the pathophysiology of intestinal dysmotility associated with high-fat diet (HFD)-induced obesity has not been elucidated. We investigated the expression of A2BR in mouse colon and their role in the mechanisms underlying the development of enteric dysmotility associated with obesity. Wild-type C57BL/6J mice were fed with HFD (60% kcal from fat) or normocaloric diet (NCD; 18% kcal from fat) for 8 weeks. Colonic A2BR localization was examined by immunofluorescence. The role of A2BR in the control of colonic motility was examined in functional experiments on longitudinal muscle preparations (LMPs). In NCD mice, A2BR were predominantly located in myenteric neurons; in HFD animals, their expression increased throughout the neuromuscular layer. Functionally, the A2BR antagonist MRS1754 enhanced electrically induced NK1-mediated tachykininergic contractions in LMPs from HFD mice, while it was less effective in tissues from NCD mice. The A2B receptor agonist BAY 60-6583 decreased colonic tachykininergic contractions in LMPs, with higher efficacy in preparations from obese mice. Both A2BR ligands did not affect contractions elicited by exogenous substance P. Obesity is related with a condition of colonic inflammation, leading to an increase of A2BR expression. A2BR, modulating the activity of excitatory tachykininergic nerves, participate to the enteric dysmotility associated with obesity.
Collapse
Affiliation(s)
- Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.,Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy.
| | - Erika Tirotta
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Daniela Gentile
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Valentina Caputi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy.,San Camillo Hospital, Treviso, Italy
| | - Ilaria Marsilio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Genny Orso
- Scientific Institute IRCCS Eugenio Medea, Bosisio Parini-, Lecco, Italy
| | - Nunzia Bernardini
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Cristina Segnani
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Chiara Ippolito
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Balázs Csóka
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Zoltán H Németh
- Department of Surgery, Morristown Medical Center, Morristown, NJ, USA
| | - György Haskó
- Department of Surgery and Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | | | - Corrado Blandizzi
- Department of Clinical and Experimental Medicine, University of Pisa, Via Roma 55, 56126, Pisa, Italy
| | - Rocchina Colucci
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| |
Collapse
|
44
|
Das UN. Is There a Role for Bioactive Lipids in the Pathobiology of Diabetes Mellitus? Front Endocrinol (Lausanne) 2017; 8:182. [PMID: 28824543 PMCID: PMC5539435 DOI: 10.3389/fendo.2017.00182] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 07/10/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammation, decreased levels of circulating endothelial nitric oxide (eNO) and brain-derived neurotrophic factor (BDNF), altered activity of hypothalamic neurotransmitters (including serotonin and vagal tone) and gut hormones, increased concentrations of free radicals, and imbalance in the levels of bioactive lipids and their pro- and anti-inflammatory metabolites have been suggested to play a role in diabetes mellitus (DM). Type 1 diabetes mellitus (type 1 DM) is due to autoimmune destruction of pancreatic β cells because of enhanced production of IL-6 and tumor necrosis factor-α (TNF-α) and other pro-inflammatory cytokines released by immunocytes infiltrating the pancreas in response to unknown exogenous and endogenous toxin(s). On the other hand, type 2 DM is due to increased peripheral insulin resistance secondary to enhanced production of IL-6 and TNF-α in response to high-fat and/or calorie-rich diet (rich in saturated and trans fats). Type 2 DM is also associated with significant alterations in the production and action of hypothalamic neurotransmitters, eNO, BDNF, free radicals, gut hormones, and vagus nerve activity. Thus, type 1 DM is because of excess production of pro-inflammatory cytokines close to β cells, whereas type 2 DM is due to excess of pro-inflammatory cytokines in the systemic circulation. Hence, methods designed to suppress excess production of pro-inflammatory cytokines may form a new approach to prevent both type 1 and type 2 DM. Roux-en-Y gastric bypass and similar surgeries ameliorate type 2 DM, partly by restoring to normal: gut hormones, hypothalamic neurotransmitters, eNO, vagal activity, gut microbiota, bioactive lipids, BDNF production in the gut and hypothalamus, concentrations of cytokines and free radicals that results in resetting glucose-stimulated insulin production by pancreatic β cells. Our recent studies suggested that bioactive lipids, such as arachidonic acid, eicosapentaneoic acid, and docosahexaenoic acid (which are unsaturated fatty acids) and their anti-inflammatory metabolites: lipoxin A4, resolvins, protectins, and maresins, may have antidiabetic actions. These bioactive lipids have anti-inflammatory actions, enhance eNO, BDNF production, restore hypothalamic dysfunction, enhance vagal tone, modulate production and action of ghrelin, leptin and adiponectin, and influence gut microbiota that may explain their antidiabetic action. These pieces of evidence suggest that methods designed to selectively deliver bioactive lipids to pancreatic β cells, gut, liver, and muscle may prevent type 1 and type 2 DM.
Collapse
Affiliation(s)
- Undurti N. Das
- BioScience Research Centre, Department of Medicine, Gayatri Vidya Parishad Hospital, GVP College of Engineering Campus, Visakhapatnam, India
- UND Life Sciences, Battle Ground, WA, United States
| |
Collapse
|
45
|
Abstract
Serotonin was first discovered in the gut, and its conventional actions as an intercellular signalling molecule in the intrinsic and extrinsic enteric reflexes are well recognized, as are a number of serotonin signalling pharmacotherapeutic targets for treatment of nausea, diarrhoea or constipation. The latest discoveries have greatly broadened our understanding of non-conventional actions of peripheral serotonin within the gastrointestinal tract and in a number of other tissues. For example, it is now clear that bacteria within the lumen of the bowel influence serotonin synthesis and release by enterochromaffin cells. Also, serotonin can act both as a pro-inflammatory and anti-inflammatory signalling molecule in the intestinal mucosa via activation of serotonin receptors (5-HT7 or 5-HT4 receptors, respectively). For decades, serotonin receptors have been known to exist in a variety of tissues other than the gut, but studies have now provided strong evidence for physiological roles of serotonin in several important processes, including haematopoiesis, metabolic homeostasis and bone metabolism. Furthermore, evidence for serotonin synthesis in peripheral tissues outside of the gut is emerging. In this Review, we expand the discussion beyond gastrointestinal functions to highlight the roles of peripheral serotonin in colitis, haematopoiesis, energy and bone metabolism, and how serotonin is influenced by the gut microbiota.
Collapse
|
46
|
Martin AM, Young RL, Leong L, Rogers GB, Spencer NJ, Jessup CF, Keating DJ. The Diverse Metabolic Roles of Peripheral Serotonin. Endocrinology 2017; 158:1049-1063. [PMID: 28323941 DOI: 10.1210/en.2016-1839] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 02/23/2017] [Indexed: 02/07/2023]
Abstract
Serotonin (5-hydroxytryptamine or 5-HT) is a multifunctional bioamine with important signaling roles in a range of physiological pathways. Almost all of the 5-HT in our bodies is synthesized in specialized enteroendocrine cells within the gastrointestinal (GI) mucosa called enterochromaffin (EC) cells. These cells provide all of our circulating 5-HT. We have long appreciated the important contributions of 5-HT within the gut, including its role in modulating GI motility. However, evidence of the physiological and clinical significance of gut-derived 5-HT outside of the gut has recently emerged, implicating 5-HT in regulation of glucose homeostasis, lipid metabolism, bone density, and diseases associated with metabolic syndrome, such as obesity and type 2 diabetes. Although a new picture has developed in the last decade regarding the various metabolic roles of peripheral serotonin, so too has our understanding of the physiology of EC cells. Given that they are scattered throughout the lining of the GI tract within the epithelial cell layer, these cells are typically difficult to study. Advances in isolation procedures now allow the study of pure EC-cell cultures and single cells, enabling studies of EC-cell physiology to occur. EC cells are sensory cells that are capable of integrating cues from ingested nutrients, the enteric nervous system, and the gut microbiome. Thus, levels of peripheral 5-HT can be modulated by a multitude of factors, resulting in both local and systemic effects for the regulation of a raft of physiological pathways related to metabolism and obesity.
Collapse
Affiliation(s)
- Alyce M Martin
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide 5042, Australia
| | - Richard L Young
- Nutrition and Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5001, Australia
- Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - Lex Leong
- Infection and Immunity, SAHMRI, Adelaide 5001, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University of South Australia, Adelaide 5042, Australia
| | - Geraint B Rogers
- Infection and Immunity, SAHMRI, Adelaide 5001, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University of South Australia, Adelaide 5042, Australia
| | - Nick J Spencer
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide 5042, Australia
| | - Claire F Jessup
- Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
- Discipline of Anatomy and Histology, Flinders University of South Australia, Adelaide 5042, Australia
| | - Damien J Keating
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide 5042, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5001, Australia
| |
Collapse
|
47
|
Anti-climacterium effects of pomegranate concentrated solutions in ovariectomized ddY mice. Exp Ther Med 2017; 13:1249-1266. [PMID: 28413464 DOI: 10.3892/etm.2017.4109] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 10/05/2016] [Indexed: 12/14/2022] Open
Abstract
In the present study, the complex anti-climacterium potential of standardized pomegranate concentrated solution (PCS) was investigated using bilateral ovariectomy (OVX) female ddY mice. Changes in body weight and gain during experimental periods, food consumption, serum estradiol levels, total body and abdominal fat densities, abdominal fat pads, and uterus weights were observed, along with the histopathology of abdominal fat pads and uterus for anti-obesity and estrogenic effects. In addition, liver weights, serum aspartate aminotransferase (AST), alanine aminotransferase (ALT) levels, and histopathological inspections were performed to explore the hepato-protective effects. Serum total cholesterol (TC), low density lipoprotein (LDL), high density lipoprotein, and triglyceride (TG) levels were monitored for hypolipidemic effects with total body and femur mean bone mineral density (BMD), right femur wet, dry and ash weights, strength, serum osteocalcin, bone-specific alkaline phosphatase (bALP) contents, and histological and histomorphometrical analyses for anti-osteoporosis activity. As a result of OVX, notable increases in body weight and gains, food consumption, abdominal fat mass densities, weights of abdominal fat pads deposited in the abdominal cavity, and serum AST, ALT, TC, LDL, TG, and osteocalcin levels were observed, along with decreases in the uterus, liver, and femur weights, mean total body and femur BMD, femur strength, serum bALP, and estradiol levels. In addition, marked hypertrophic alterations in adipocytes located in the deposited abdominal fat pads, liver steatosis, uterine disused atrophic changes, and decreases in bone mass and structures of the femur were also observed in OVX control mice with significant increases in bone resorption markers based on histopathological and histomorphometrical analysis. However, these estrogen-deficient climacterium symptoms were significantly (P<0.05 or P<0.01) inhibited after 84 days of continuous treatment with estradiol and PCS (1, 2 and 4 ml/kg), respectively. The present results suggested that PCS was able to effectively inhibit or refine the climacterium symptoms, including obesity, hyperlipidemia, hepatic steatosis, and osteoporosis, induced by OVX in ddY mice.
Collapse
|
48
|
Insight gained from genome-wide interaction and enrichment analysis on weight gain during citalopram treatment. Neurosci Lett 2016; 637:38-43. [PMID: 27899308 DOI: 10.1016/j.neulet.2016.11.056] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 11/24/2016] [Accepted: 11/25/2016] [Indexed: 12/11/2022]
Abstract
Weight gain is a possible side effect of the pharmacological antidepressant treatments. Defining antidepressant prescriptions based on personal genetic makeups would decrease the risk of weight gain and increase the quality of the current antidepressant pharmacological treatments. 643 depressed, citalopram treated individuals with available clinical and genome-wide genetic information were investigated to identify the molecular pathways associated with weight gain. 111 individuals experienced weight gain during citalopram treatment. The axon guidance (p.adjust=0.005) and the developmental biology pathway (p.adjust=0.01) were enriched in variations associated with weight gain. The developmental biology pathway includes molecular cascades involved in the regulation of beta-cell development, and the transcriptional regulation of white adipocyte differentiation. A number of variations were harbored by genes whose products are involved in the synthesis of collagen (COL4A3, COL5A1 and ITGA1), activity of the thyroid-hormones (NCOR1 and NCOR2), energy metabolism (ADIPOQ, PPARGC1A) and myogenic differentiation (CDON). A molecular pathway analysis conducted in a sample of depressed patients identified new candidate genes whose future investigation may provide insights in the molecular events that drive weight gain during antidepressant treatment.
Collapse
|
49
|
Lowette K, Desmet AS, Farré RM, Tack J, Vanden Berghe P. Fructose consumption impairs serotonergic signaling in the murine enteric nervous system. Neurogastroenterol Motil 2016; 28:1438-42. [PMID: 27098706 DOI: 10.1111/nmo.12827] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/03/2016] [Indexed: 02/08/2023]
Abstract
The intake of free fructose has increased substantially since the development of high-fructose corn syrup. This has not only been associated with metabolic disorders but recent evidence also indicates that chronic fructose consumption can affect neuronal and cognitive function. In this study we investigated the effects of fructose consumption on serotonergic signaling and neuronal activity in the mouse submucous plexus. Male mice were put on a control or fructose (23% solution) diet for 6 weeks or were assigned to a recovery group that received normal water (2 weeks) after 4 weeks of fructose. At the end of the diet, gene expressions and enteric neuronal activity, after depolarization with high K(+) and 5-HT, were measured using Ca(2+) imaging and RT-qPCR, respectively. Even in the lack of gain weight and the absence of changes in duodenal permeability, the total number of 5-HT-responding neurons and the depolarization and 5-HT-evoked Ca(2+) amplitudes were significantly lower after fructose consumption. Expression of synaptobrevin CaV 2.1 and CaV 2.2 mRNA did not differ after fructose intake; however, CaV 2.1 mRNA levels were significantly higher in the recovery animals. SERT mRNA concentration, isolated from submucosal plexus containing mucosal epithelium, was significantly decreased after fructose consumption. Chronic fructose consumption impairs serotonergic signaling in the mouse submucous plexus, prior to weight gain and detectable intestinal permeability problems.
Collapse
Affiliation(s)
- K Lowette
- Laboratory for Enteric NeuroScience (LENS), University of Leuven, Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - A-S Desmet
- Laboratory for Enteric NeuroScience (LENS), University of Leuven, Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - R M Farré
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - J Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | - P Vanden Berghe
- Laboratory for Enteric NeuroScience (LENS), University of Leuven, Leuven, Belgium
- Translational Research Center for Gastrointestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Selection of the Optimal Herbal Compositions of Red Clover and Pomegranate According to Their Protective Effect against Climacteric Symptoms in Ovariectomized Mice. Nutrients 2016; 8:nu8080447. [PMID: 27455321 PMCID: PMC4997362 DOI: 10.3390/nu8080447] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 07/19/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023] Open
Abstract
This study aimed to ascertain the optimal range of red clover dry extracts (RC) and dried pomegranate concentrate powder (PCP) to induce anti-climacteric effects. Thus, the dose ranges showing protective effect of mixed formulae consisting of RC and PCP were examined in ovariectomized mice. At 28 days after bilateral ovariectomy (OVX), mixed herbal compositions (RC:PCP = 1:1, 1:2, 1:4, 1:8, 2:1, 4:1, and 8:1) were administered orally, at 120 mg/kg once daily for 84 days. We evaluated that RC and PCP mixture attenuate OVX-caused obesity, hyperlipidemia, hepatic steatosis, and osteoporosis. Compared to OVX-induced control mice, body weight and abdominal fat weight in OVX-induced mice were significantly decreased, concomitantly with increase of uterus weight by RC:PCP mixture. Additionally, significant increases in serum estradiol levels were observed in all RC:PCP-treated mice. RC:PCP mixture also showed protective effect against OVX-induced hyperlipidemia, hepatic steatosis. Total body and femur mean bone mineral density (BMD), osteocalcin, bALP contents were effectively increased by RC:PCP mixture. Taken together, RC:PCP mixture (2:1, 1:1, and 4:1) has remarkable protective effects against the changes induced by OVX. In particular, RC:PCP mixture (2:1) shows the strongest effect and may be considered as a potential protective agent against climacteric symptoms.
Collapse
|