351
|
Larraufie P, Martin-Gallausiaux C, Lapaque N, Dore J, Gribble FM, Reimann F, Blottiere HM. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci Rep 2018; 8:74. [PMID: 29311617 PMCID: PMC5758799 DOI: 10.1038/s41598-017-18259-0] [Citation(s) in RCA: 243] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022] Open
Abstract
Peptide-YY (PYY) and Glucagon-Like Peptide-1 (GLP-1) play important roles in the regulation of food intake and insulin secretion, and are of translational interest in the field of obesity and diabetes. PYY production is highest in enteroendocrine cells located in the distal intestine, mirroring the sites where high concentrations of short chain fatty acids (SCFAs) are produced by gut microbiota. We show here that propionate and butyrate strongly increased expression of PYY but not GCG in human cell line and intestinal primary culture models. The effect was predominantly attributable to the histone deacetylase inhibitory activity of SCFA and minor, but significant contributions of FFA2 (GPR43). Consistent with the SCFA-dependent elevation of PYY gene expression, we also observed increased basal and stimulated PYY hormone secretion. Interestingly, the transcriptional stimulation of PYY was specific to human-derived cell models and not reproduced in murine primary cultures. This is likely due to substantial differences in PYY gene structure between mouse and human. In summary, this study revealed a strong regulation of PYY production by SCFA that was evident in humans but not mice, and suggests that high fibre diets elevate plasma concentrations of the anorexigenic hormone PYY, both by targeting gene expression and hormone secretion.
Collapse
Affiliation(s)
- P Larraufie
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, WT-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
| | - C Martin-Gallausiaux
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,Sorbonne Universités, UPMC Univ Paris 06, IFD, 4 place Jussieu, 75252, Paris, cedex 05, France
| | - N Lapaque
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - J Dore
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,US 1367 MetaGenoPolis, INRA, Université Paris-Saclay, 78350, Jouy en Josas, France
| | - F M Gribble
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, WT-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - F Reimann
- University of Cambridge, Metabolic Research Laboratories and MRC Metabolic Diseases Unit, WT-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - H M Blottiere
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.,US 1367 MetaGenoPolis, INRA, Université Paris-Saclay, 78350, Jouy en Josas, France
| |
Collapse
|
352
|
Zapata RC, Singh A, Chelikani PK. Peptide YY mediates the satiety effects of diets enriched with whey protein fractions in male rats. FASEB J 2018; 32:850-861. [PMID: 29042449 DOI: 10.1096/fj.201700519rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dairy proteins-whey protein, in particular-are satiating and often recommended for weight control; however, little is known about the mechanisms by which whey protein and its components promote satiety and weight loss. We used diet-induced obese rats to determine whether the hypophagic effects of diets that are enriched with whey and its fractions, lactalbumin and lactoferrin, are mediated by the gut hormone, peptide YY (PYY). We demonstrate that high protein diets that contain whey, lactalbumin, and lactoferrin decreased food intake and body weight with a concurrent increase in PYY mRNA abundance in the colon and/or plasma PYY concentrations. Of importance, blockade of PYY neuropeptide Y receptor subtype 2 (Y2) receptors with a peripherally restricted antagonist attenuated the hypophagic effects of diets that are enriched with whey protein fractions. Diets that are enriched with whey fractions were less preferred; however, in a modified conditioned taste preference test, PYY Y2 receptor blockade induced hyperphagia of a lactoferrin diet, but caused a reduction in preference for Y2 antagonist-paired flavor, which suggested that PYY signaling is important for lactoferrin-induced satiety, but not essential for preference for lactoferrin-enriched diets. Taken together, these data provide evidence that the satiety of diets that are enriched with whey protein components is mediated, in part, via enhanced PYY secretion and action in obese male rats.-Zapata, R. C., Singh, A., Chelikani, P. K. Peptide YY mediates the satiety effects of diets enriched with whey protein fractions in male rats.
Collapse
Affiliation(s)
- Rizaldy C Zapata
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Arashdeep Singh
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Prasanth K Chelikani
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada.,Gastrointestinal Research Group, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
353
|
Abstract
The prevalence of obesity and overweight has plateaued in developed countries, although at high levels, but in most parts of the world, it continues to increase. Current recommendations for preventing and treating obesity are based mainly on the notion that overeating results from hedonic eating as a result of unlimited access to palatable foods, particularly those high in sugar and fat, and that hedonic centers are able to "override" the body's homeostatic mechanisms. This article proposes that the homeostatic mechanisms affecting appetite and satiety are more important in chronic overeating, and that sufficient evidence exists for adopting a new paradigm for controlling individual and global obesity based on controlling energy homeostasis via the enteroendocrine and gut microbiota systems. Many obese children and adolescents have chronic hunger, supporting the notion that they have a homeostatic rather than hedonic abnormality. The effectiveness of weight loss drugs and bariatric surgery suggests that the brain centers controlling energy homeostasis are able to override centers controlling hedonic drives. Energy homeostasis can also be influenced by nutrition, in particular, by avoiding sweetened drinks and consuming whole grains, vegetables, fruits and other foods that are high in dietary fiber, and thereby influence appetite and satiety. New recommendations are outlined for preventing and treating individual and global obesity based on a paradigm that targets appetite and satiety.
Collapse
|
354
|
Liu H, Wang J, He T, Becker S, Zhang G, Li D, Ma X. Butyrate: A Double-Edged Sword for Health? Adv Nutr 2018; 9:21-29. [PMID: 29438462 PMCID: PMC6333934 DOI: 10.1093/advances/nmx009] [Citation(s) in RCA: 605] [Impact Index Per Article: 100.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 11/11/2017] [Indexed: 02/06/2023] Open
Abstract
Butyrate, a four-carbon short-chain fatty acid, is produced through microbial fermentation of dietary fibers in the lower intestinal tract. Endogenous butyrate production, delivery, and absorption by colonocytes have been well documented. Butyrate exerts its functions by acting as a histone deacetylase (HDAC) inhibitor or signaling through several G protein-coupled receptors (GPCRs). Recently, butyrate has received particular attention for its beneficial effects on intestinal homeostasis and energy metabolism. With anti-inflammatory properties, butyrate enhances intestinal barrier function and mucosal immunity. However, the role of butyrate in obesity remains controversial. Growing evidence has highlighted the impact of butyrate on the gut-brain axis. In this review, we summarize the present knowledge on the properties of butyrate, especially its potential effects and mechanisms involved in intestinal health and obesity.
Collapse
Affiliation(s)
- Hu Liu
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ji Wang
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Ting He
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Sage Becker
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Guolong Zhang
- Department of Animal Science, Oklahoma State University, Stillwater, OK; Departments of Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Defa Li
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, Beijing, China,Internal Medicine and Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX,Address correspondence to XM (e-mail: )
| |
Collapse
|
355
|
Keel PK, Eckel LA, Hildebrandt BA, Haedt-Matt AA, Appelbaum J, Jimerson DC. Disturbance of gut satiety peptide in purging disorder. Int J Eat Disord 2018; 51:53-61. [PMID: 29219202 DOI: 10.1002/eat.22806] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/06/2017] [Accepted: 11/12/2017] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Little is known about biological factors that contribute to purging after normal amounts of food-the central feature of purging disorder (PD). This study comes from a series of nested studies examining ingestive behaviors in bulimic syndromes and specifically evaluated the satiety peptide YY (PYY) and the hunger peptide ghrelin in women with PD (n = 25), bulimia nervosa-purging (BNp) (n = 26), and controls (n = 26). Based on distinct subjective responses to a fixed meal in PD (Keel, Wolfe, Liddle, DeYoung, & Jimerson, ), we tested whether postprandial PYY response was significantly greater and ghrelin levels significantly lower in women with PD compared to controls and women with BNp. METHOD Participants completed structured clinical interviews, self-report questionnaires, and laboratory assessments of gut peptide and subjective responses to a fixed meal. RESULTS Women with PD demonstrated a significantly greater postprandial PYY response compared to women with BNp and controls, who did not differ significantly. PD women also endorsed significantly greater gastrointestinal distress, and PYY predicted gastrointestinal intestinal distress. Ghrelin levels were significantly greater in PD and BNp compared to controls, but did not differ significantly between eating disorders. Women with BNp endorsed significantly greater postprandial hunger, and ghrelin predicted hunger. DISCUSSION PD is associated with a unique disturbance in PYY response. Findings contribute to growing evidence of physiological distinctions between PD and BNp. Future research should examine whether these distinctions account for differences in clinical presentation as this could inform the development of specific interventions for patients with PD.
Collapse
Affiliation(s)
- Pamela K Keel
- Department of Psychology, Florida State University, Tallahassee, FL
| | - Lisa A Eckel
- Department of Psychology, Florida State University, Tallahassee, FL
| | | | | | | | - David C Jimerson
- Department of Psychiatry, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
356
|
Qiu NC, Cen XX, Liu ME, Liu Q, Zha SL, Shan CX, Zhang W, Wang LD, Wang Y, Qiu M. Greater Curvature Plication with Duodenal–Jejunal Bypass: a Novel Metabolic Surgery for Type 2 Diabetes Mellitus. Obes Surg 2017; 28:1595-1601. [DOI: 10.1007/s11695-017-3057-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
357
|
Luo QQ, Zhou G, Huang SN, Mu MD, Chen YJ, Qian ZM. Ghrelin is Negatively Correlated with Iron in the Serum in Human and Mice. ANNALS OF NUTRITION AND METABOLISM 2017; 72:37-42. [PMID: 29241202 DOI: 10.1159/000484698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/26/2017] [Indexed: 01/15/2023]
Abstract
BACKGROUND/AIMS The studies in the patients with iron deficiency anemia (IDA) implied the existence of the association of ghrelin with iron or hepcidin levels in the plasma under the pathophysiological conditions. We hypothesized that fasting may be able to affect iron metabolism via ghrelin under the physiological conditions. METHODS We investigated the effects of fasting on serum ghrelin and iron contents in healthy volunteers (23-31 years) and C57BL/6 male mice (8-week-olds) under the physiological conditions. RESULTS Fasting induced a significant elevation in both total ghrelin and acylated ghrelin and a reduction in iron levels in the serum of both human and mice. Correlation analysis demonstrated that total ghrelin or acylated ghrelin is negatively correlated with iron in the serum in human and mice. CONCLUSION Ghrelin has a role to reduce serum iron under the conditions of fasting.
Collapse
Affiliation(s)
- Qian-Qian Luo
- Department of Pharmacology and Laboratory of Prevention and Treatment of Neurodegenerative Diseases, Department of Biochemistry, Nantong University Medical School, Nantong, China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Su-Na Huang
- Department of Pharmacology and Laboratory of Prevention and Treatment of Neurodegenerative Diseases, Department of Biochemistry, Nantong University Medical School, Nantong, China
| | - Ming-Dao Mu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yun-Jin Chen
- Department of Pharmacology and Laboratory of Prevention and Treatment of Neurodegenerative Diseases, Department of Biochemistry, Nantong University Medical School, Nantong, China
| | - Zhong-Ming Qian
- Department of Pharmacology and Laboratory of Prevention and Treatment of Neurodegenerative Diseases, Department of Biochemistry, Nantong University Medical School, Nantong, China
| |
Collapse
|
358
|
Alkhatib A, Tsang C, Tiss A, Bahorun T, Arefanian H, Barake R, Khadir A, Tuomilehto J. Functional Foods and Lifestyle Approaches for Diabetes Prevention and Management. Nutrients 2017; 9:E1310. [PMID: 29194424 PMCID: PMC5748760 DOI: 10.3390/nu9121310] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/26/2017] [Accepted: 11/27/2017] [Indexed: 02/07/2023] Open
Abstract
Functional foods contain biologically active ingredients associated with physiological health benefits for preventing and managing chronic diseases, such as type 2 diabetes mellitus (T2DM). A regular consumption of functional foods may be associated with enhanced anti-oxidant, anti-inflammatory, insulin sensitivity, and anti-cholesterol functions, which are considered integral to prevent and manage T2DM. Components of the Mediterranean diet (MD)-such as fruits, vegetables, oily fish, olive oil, and tree nuts-serve as a model for functional foods based on their natural contents of nutraceuticals, including polyphenols, terpenoids, flavonoids, alkaloids, sterols, pigments, and unsaturated fatty acids. Polyphenols within MD and polyphenol-rich herbs-such as coffee, green tea, black tea, and yerba maté-have shown clinically-meaningful benefits on metabolic and microvascular activities, cholesterol and fasting glucose lowering, and anti-inflammation and anti-oxidation in high-risk and T2DM patients. However, combining exercise with functional food consumption can trigger and augment several metabolic and cardiovascular protective benefits, but it is under-investigated in people with T2DM and bariatric surgery patients. Detecting functional food benefits can now rely on an "omics" biological profiling of individuals' molecular, genetics, transcriptomics, proteomics, and metabolomics, but is under-investigated in multi-component interventions. A personalized approach for preventing and managing T2DM should consider biological and behavioral models, and embed nutrition education as part of lifestyle diabetes prevention studies. Functional foods may provide additional benefits in such an approach.
Collapse
Affiliation(s)
- Ahmad Alkhatib
- Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait.
| | - Catherine Tsang
- Faculty of Health and Social Care, Edge Hill University, St. Helens Road, Ormskirk, Lancashire L39 4QP, UK.
| | - Ali Tiss
- Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait.
| | - Theeshan Bahorun
- ANDI Centre of Excellence for Biomedical and Biomaterials Research, University of Mauritius, MSIRI Building, Réduit 80837, Mauritius.
| | | | - Roula Barake
- Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait.
| | | | - Jaakko Tuomilehto
- Dasman Diabetes Institute, P.O. Box 1180, Dasman 15462, Kuwait.
- Diabetes Research Group, King Abdulaziz University, P.O. Box 80200, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
359
|
Bird JK, Raederstorff D, Weber P, Steinert RE. Cardiovascular and Antiobesity Effects of Resveratrol Mediated through the Gut Microbiota. Adv Nutr 2017; 8:839-849. [PMID: 29141969 PMCID: PMC5682996 DOI: 10.3945/an.117.016568] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Encouraging scientific research into the health effects of dietary bioactive resveratrol has been confounded by its rapid first-pass metabolism, which leads to low in vivo bioavailability. Preliminary studies have shown that resveratrol can modulate gut microbiota composition, undergo biotransformation to active metabolites via the intestinal microbiota, or affect gut barrier function. In rodents, resveratrol can modify the relative Bacteroidetes:Firmicutes ratio and reverse the gut microbial dysbiosis caused by a high-fat diet. By upregulating the expression of genes involved in maintaining tight junctions between intestinal cells, resveratrol contributes to gut barrier integrity. The composition of the gut microbiome and rapid metabolism of resveratrol determines the production of resveratrol metabolites, which are found at greater concentrations in humans after ingestion than their parent molecule and can have similar biological effects. Resveratrol may affect cardiovascular risk factors such as elevated blood cholesterol or trimethylamine N-oxide concentrations. Modulating the composition of the gut microbiota by resveratrol may affect central energy metabolism and modify concentrations of satiety hormones to produce antiobesity effects. Encouraging research from animal models could be tested in humans.
Collapse
Affiliation(s)
- Julia K Bird
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and
| | - Daniel Raederstorff
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and
| | - Peter Weber
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and
| | - Robert E Steinert
- Human Nutrition and Health, DSM Nutritional Products, Basel, Switzerland, and,Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
360
|
Machytka E, Bužga M, Zonca P, Lautz DB, Ryou M, Simonson DC, Thompson CC. Partial jejunal diversion using an incisionless magnetic anastomosis system: 1-year interim results in patients with obesity and diabetes. Gastrointest Endosc 2017; 86:904-912. [PMID: 28716404 DOI: 10.1016/j.gie.2017.07.009] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 07/06/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Most patients with type 2 diabetes mellitus have obesity. Studies show that bariatric surgery is superior to medical treatment for remission of type 2 diabetes mellitus. Nevertheless, very few patients undergo surgery, and a less-invasive endoscopic alternative is desirable. METHODS This was a single-arm first-in-human pilot study designed to evaluate the technical feasibility, safety, and clinical performance of the incisionless magnetic anastomosis system (IMAS) to create a partial jejunal diversion (PJD). Ten patients with obesity and type 2 diabetes mellitus, prediabetes, or no diabetes were enrolled. A PJD to the ileum was attempted in all patients under general anesthesia. The IMAS was delivered through the working channel of a colonoscope, with laparoscopic supervision. The patients were not required to participate in an intensive lifestyle/diet management program. Endoscopic visualization of the anastomosis was obtained at 2, 6, and 12 months. Patient weight, glycemic profile, and metabolic panels were acquired at 0.5, 1, 2, 3, 6, 9, and 12 months. RESULTS A PJD was created in all patients with no device-related serious adverse events. The anastomosis remained widely patent in all patients at 1 year. Average total weight loss was 14.6% (40.2% excess weight loss at 12 months). A significant reduction in glycated hemoglobin level was observed in all diabetic (1.9%) and prediabetic (1.0%) patients, while reducing or eliminating the use of diabetes medications. CONCLUSIONS Permanent anastomosis for PJD was created in all patients with the IMAS. This resulted in improvement in measures of hyperglycemia and progressive weight loss. (Clinical trial registration number: NCT02839512.).
Collapse
Affiliation(s)
- Evžen Machytka
- Department of Surgical Studies, Faculty of Medicine, University Hospital Ostrava, Ostrava, Czech Republic
| | - Marek Bužga
- Department of Physiology and Pathophysiology, University of Ostrava Faculty of Medicine, Ostrava, Czech Republic
| | - Pavel Zonca
- Department of Surgical Studies, Faculty of Medicine, University Hospital Ostrava, Ostrava, Czech Republic
| | - David B Lautz
- Emerson Hospital, Concord, Massachusetts, USA; Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
361
|
Steinert RE, Ullrich SS, Geary N, Asarian L, Bueter M, Horowitz M, Feinle-Bisset C. Comparative effects of intraduodenal amino acid infusions on food intake and gut hormone release in healthy males. Physiol Rep 2017; 5:e13492. [PMID: 29138359 PMCID: PMC5688783 DOI: 10.14814/phy2.13492] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/12/2017] [Indexed: 02/07/2023] Open
Abstract
In contrast to the many studies of the effects of individual amino acids (AAs) on eating, no studies have compared the effects of different AAs on eating and underlying preabsorptive gastrointestinal mechanisms. To compare the effects of intraduodenal infusions of l-tryptophan (TRP), l-leucine (LEU), l-phenylalanine (PHE) and l-glutamine (GLN) on appetite, gastrointestinal hormone responses (including ghrelin, cholecystokinin (CCK), peptide YY (PYY) and glucagon-like peptide-1 [GLP-1]), glycemia (glucagon, insulin and glucose) and test meal size in healthy males, we retrospectively analyzed data from four published independent, randomized, double-blind, placebo-controlled studies of 90-min intraduodenal infusions of the individual AAs. The designs of the studies were identical, except the dose of TRP (0.15 kcal/min) was lower than that of the other AAs (0.45 kcal/min) because higher doses of this AA were not well tolerated. TRP and LEU decreased intake more than PHE (reductions relative to control, ~219 ± 68, ~170 ± 48 and ~12 ± 57 kcal, respectively), and TRP decreased intake more than GLN (~31 ± 82 kcal). These effects of TRP and LEU versus GLN, but not versus PHE, were paralleled by greater decreases in plasma ghrelin, and increases in CCK, concentrations. TRP increased PYY more than GLN or LEU, but not PHE. LEU increased PYY less than PHE. No significant differences were detected for GLP-1. PHE increased glucagon more than TRP or LEU, and increased insulin more than TRP. Under our experimental conditions, intraduodenal TRP and LEU were more satiating than PHE and GLN. The greater satiating efficacy of LEU versus PHE was significantly dissociated from the effects of these AAs on PYY, while the greater satiating potency of TRP versus PHE was significantly dissociated from the effects of these AAs on insulin and glucagon. In contrast, ghrelin and CCK, and potentially other mechanisms, including central sensing of individual AAs, appear to be stronger candidate mechanisms for the relative satiating effects obtained.
Collapse
Affiliation(s)
- Robert E Steinert
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, South Australia, Australia
- Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Sina S Ullrich
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, South Australia, Australia
| | - Nori Geary
- Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- Department of Medicine-Immunobiology, Robert Larner College of Medicine University of Vermont, Burlington, Vermont
| | - Marco Bueter
- Department of Surgery, Division of Visceral and Transplantation Surgery, University Hospital Zürich, Zürich, Switzerland
| | - Michael Horowitz
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, South Australia, Australia
| | - Christine Feinle-Bisset
- Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
- NHMRC Centre of Research Excellence in Translating Nutritional Science to Good Health University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
362
|
Sasaki T. Neural and Molecular Mechanisms Involved in Controlling the Quality of Feeding Behavior: Diet Selection and Feeding Patterns. Nutrients 2017; 9:nu9101151. [PMID: 29053636 PMCID: PMC5691767 DOI: 10.3390/nu9101151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 12/20/2022] Open
Abstract
We are what we eat. There are three aspects of feeding: what, when, and how much. These aspects represent the quantity (how much) and quality (what and when) of feeding. The quantitative aspect of feeding has been studied extensively, because weight is primarily determined by the balance between caloric intake and expenditure. In contrast, less is known about the mechanisms that regulate the qualitative aspects of feeding, although they also significantly impact the control of weight and health. However, two aspects of feeding quality relevant to weight loss and weight regain are discussed in this review: macronutrient-based diet selection (what) and feeding pattern (when). This review covers the importance of these two factors in controlling weight and health, and the central mechanisms that regulate them. The relatively limited and fragmented knowledge on these topics indicates that we lack an integrated understanding of the qualitative aspects of feeding behavior. To promote better understanding of weight control, research efforts must focus more on the mechanisms that control the quality and quantity of feeding behavior. This understanding will contribute to improving dietary interventions for achieving weight control and for preventing weight regain following weight loss.
Collapse
Affiliation(s)
- Tsutomu Sasaki
- Laboratory for Metabolic Signaling, Institute for Molecular and Cellular Regulation, Gunma University, 3-39-15 Showa-machi, Maebashi, Gunma 371-8512, Japan.
| |
Collapse
|
363
|
Petersen KU. Pepsin and Its Importance for Functional Dyspepsia: Relic, Regulator or Remedy? Dig Dis 2017; 36:98-105. [PMID: 28982106 DOI: 10.1159/000481399] [Citation(s) in RCA: 5] [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: 05/09/2017] [Accepted: 09/06/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND Functional dyspepsia is a heterogeneous disorder lacking an established therapeutic strategy. Historical treatment attempts with pepsin products were shrugged off, as a simple calculation shows that quantitative substitution is pointless. However, such attempts might have been right for the wrong reason. SUMMARY Today, the role of pepsins is primarily seen in the provision of signalling amino acids (especially phenylalanine and tryptophan) and peptides, which initiate processes promoting digestion. Proteolysis benefits from pepsin variants showing, contrary to common belief, activities of up to a pH value of 5.0. Non-clinical and clinical studies support the view that liberated amino acids produce a variety of direct and indirect effects. Signal chains stimulated by (mostly aromatic) amino acids lead to secretion of gastrin and cholecystokinin (CCK), mediated, respectively, by CCK2 (gastrin) and Ca2+-sensing receptors in the parietal cell, and Ca2+-sensing receptors in the antral and duodenal mucosa. Thus, CCK effects such as secretion of pancreatic enzymes and promotion of gastric accommodation are (also) consequential to peptic liberation of amino acids. Key Message: As functional dyspepsia represents a heterogeneous disorder, it may be intriguing to view pepsin as a potential (although still to be proven) treatment modality, distinguished by a diversity of pro-digestive effects.
Collapse
|
364
|
Leeners B, Geary N, Tobler PN, Asarian L. Ovarian hormones and obesity. Hum Reprod Update 2017; 23:300-321. [PMID: 28333235 DOI: 10.1093/humupd/dmw045] [Citation(s) in RCA: 203] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/23/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Obesity is caused by an imbalance between energy intake, i.e. eating and energy expenditure (EE). Severe obesity is more prevalent in women than men worldwide, and obesity pathophysiology and the resultant obesity-related disease risks differ in women and men. The underlying mechanisms are largely unknown. Pre-clinical and clinical research indicate that ovarian hormones may play a major role. OBJECTIVE AND RATIONALE We systematically reviewed the clinical and pre-clinical literature on the effects of ovarian hormones on the physiology of adipose tissue (AT) and the regulation of AT mass by energy intake and EE. SEARCH METHODS Articles in English indexed in PubMed through January 2016 were searched using keywords related to: (i) reproductive hormones, (ii) weight regulation and (iii) central nervous system. We sought to identify emerging research foci with clinical translational potential rather than to provide a comprehensive review. OUTCOMES We find that estrogens play a leading role in the causes and consequences of female obesity. With respect to adiposity, estrogens synergize with AT genes to increase gluteofemoral subcutaneous AT mass and decrease central AT mass in reproductive-age women, which leads to protective cardiometabolic effects. Loss of estrogens after menopause, independent of aging, increases total AT mass and decreases lean body mass, so that there is little net effect on body weight. Menopause also partially reverses women's protective AT distribution. These effects can be counteracted by estrogen treatment. With respect to eating, increasing estrogen levels progressively decrease eating during the follicular and peri-ovulatory phases of the menstrual cycle. Progestin levels are associated with eating during the luteal phase, but there does not appear to be a causal relationship. Progestins may increase binge eating and eating stimulated by negative emotional states during the luteal phase. Pre-clinical research indicates that one mechanism for the pre-ovulatory decrease in eating is a central action of estrogens to increase the satiating potency of the gastrointestinal hormone cholecystokinin. Another mechanism involves a decrease in the preference for sweet foods during the follicular phase. Genetic defects in brain α-melanocycte-stimulating hormone-melanocortin receptor (melanocortin 4 receptor, MC4R) signaling lead to a syndrome of overeating and obesity that is particularly pronounced in women and in female animals. The syndrome appears around puberty in mice with genetic deletions of MC4R, suggesting a role of ovarian hormones. Emerging functional brain-imaging data indicates that fluctuations in ovarian hormones affect eating by influencing striatal dopaminergic processing of flavor hedonics and lateral prefrontal cortex processing of cognitive inhibitory controls of eating. There is a dearth of research on the neuroendocrine control of eating after menopause. There is also comparatively little research on the effects of ovarian hormones on EE, although changes in ovarian hormone levels during the menstrual cycle do affect resting EE. WIDER IMPLICATIONS The markedly greater obesity burden in women makes understanding the diverse effects of ovarian hormones on eating, EE and body adiposity urgent research challenges. A variety of research modalities can be used to investigate these effects in women, and most of the mechanisms reviewed are accessible in animal models. Therefore, human and translational research on the roles of ovarian hormones in women's obesity and its causes should be intensified to gain further mechanistic insights that may ultimately be translated into novel anti-obesity therapies and thereby improve women's health.
Collapse
Affiliation(s)
- Brigitte Leeners
- Division of Reproductive Endocrinology, University Hospital Zurich, Frauenklinikstr. 10, CH 8091 Zurich, Switzerland.,Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland
| | - Nori Geary
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Philippe N Tobler
- Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland.,Laboratory for Social and Neural Systems Research, Department of Economics, University of Zurich, 8006 Zurich, Switzerland
| | - Lori Asarian
- Center for Integrative Human Physiology (ZIHP), University of Zurich, 8057 Zurich, Switzerland.,Institute of Veterinary Physiology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
365
|
Westfall S, Lomis N, Kahouli I, Dia SY, Singh SP, Prakash S. Microbiome, probiotics and neurodegenerative diseases: deciphering the gut brain axis. Cell Mol Life Sci 2017; 74:3769-3787. [PMID: 28643167 PMCID: PMC11107790 DOI: 10.1007/s00018-017-2550-9] [Citation(s) in RCA: 321] [Impact Index Per Article: 45.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/05/2017] [Accepted: 05/29/2017] [Indexed: 02/07/2023]
Abstract
The gut microbiota is essential to health and has recently become a target for live bacterial cell biotherapies for various chronic diseases including metabolic syndrome, diabetes, obesity and neurodegenerative disease. Probiotic biotherapies are known to create a healthy gut environment by balancing bacterial populations and promoting their favorable metabolic action. The microbiota and its respective metabolites communicate to the host through a series of biochemical and functional links thereby affecting host homeostasis and health. In particular, the gastrointestinal tract communicates with the central nervous system through the gut-brain axis to support neuronal development and maintenance while gut dysbiosis manifests in neurological disease. There are three basic mechanisms that mediate the communication between the gut and the brain: direct neuronal communication, endocrine signaling mediators and the immune system. Together, these systems create a highly integrated molecular communication network that link systemic imbalances with the development of neurodegeneration including insulin regulation, fat metabolism, oxidative markers and immune signaling. Age is a common factor in the development of neurodegenerative disease and probiotics prevent many harmful effects of aging such as decreased neurotransmitter levels, chronic inflammation, oxidative stress and apoptosis-all factors that are proven aggravators of neurodegenerative disease. Indeed patients with Parkinson's and Alzheimer's diseases have a high rate of gastrointestinal comorbidities and it has be proposed by some the management of the gut microbiota may prevent or alleviate the symptoms of these chronic diseases.
Collapse
Affiliation(s)
- Susan Westfall
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Nikita Lomis
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Imen Kahouli
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Si Yuan Dia
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada
| | - Surya Pratap Singh
- Department of Biochemistry, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada.
- Department of Experimental Medicine, Faculty of Medicine, McGill University, 3775 University Street, Montreal, QC, H3A2B4, Canada.
| |
Collapse
|
366
|
Farias G, Netto BDM, Bettini SC, Dâmaso AR, de Freitas ACT. Neuroendocrine regulation of energy balance: Implications on the development and surgical treatment of obesity. Nutr Health 2017; 23:131-146. [PMID: 28838280 DOI: 10.1177/0260106017719369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Obesity, a serious public health problem, occurs mainly when food consumption exceeds energy expenditure. Therefore, energy balance depends on the regulation of the hunger-satiety mechanism, which involves interconnection of the central nervous system and peripheral signals from the adipose tissue, pancreas and gastrointestinal tract, generating responses in short-term food intake and long-term energy balance. Increased body fat alters the gut- and adipose-tissue-derived hormone signaling, which promotes modifications in appetite-regulating hormones, decreasing satiety and increasing hunger senses. With the failure of conventional weight loss interventions (dietary treatment, exercise, drugs and lifestyle modifications), bariatric surgeries are well-accepted tools for the treatment of severe obesity, with long-term and sustained weight loss. Bariatric surgeries may cause weight loss due to restriction/malabsorption of nutrients from the anatomical alteration of the gastrointestinal tract that decreases energy intake, but also by other physiological factors associated with better results of the surgical procedure. OBJECTIVE This review discusses the neuroendocrine regulation of energy balance, with description of the predominant hormones and peptides involved in the control of energy balance in obesity and all currently available bariatric surgeries. CONCLUSIONS According to the findings of our review, bariatric surgeries promote effective and sustained weight loss not only by reducing calorie intake, but also by precipitating changes in appetite control, satiation and satiety, and physiological changes in gut-, neuro- and adipose-tissue-derived hormone signaling.
Collapse
Affiliation(s)
- Gisele Farias
- 1 Department of Surgery, Universidade Federal do Paraná, UFPR, Curitiba-Pr, Brazil
| | | | - Solange Cravo Bettini
- 3 Federal University of Paraná (UFPR), Gastrointestinal Surgery Service of Hospital de Clínicas, Curitiba-PR, Brazil
| | | | | |
Collapse
|
367
|
Giezenaar C, Trahair LG, Luscombe-Marsh ND, Hausken T, Standfield S, Jones KL, Lange K, Horowitz M, Chapman I, Soenen S. Effects of randomized whey-protein loads on energy intake, appetite, gastric emptying, and plasma gut-hormone concentrations in older men and women. Am J Clin Nutr 2017; 106:865-877. [PMID: 28747330 DOI: 10.3945/ajcn.117.154377] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/27/2017] [Indexed: 02/05/2023] Open
Abstract
Background: Protein- and energy-rich supplements are used widely for the management of malnutrition in the elderly. Information about the effects of protein on energy intake and related gastrointestinal mechanisms and whether these differ between men and women is limited.Objective: We determined the effects of whey protein on energy intake, appetite, gastric emptying, and gut hormones in healthy older men and women.Design: Eight older women and 8 older men [mean ± SEM age: 72 ± 1 y; body mass index (in kg/m2): 25 ± 1] were studied on 3 occasions in which they received protein loads of 30 g (120 kcal) or 70 g (280 kcal) or a flavored water control drink (0 kcal). At regular intervals over 180 min, appetite (visual analog scales), gastric emptying (3-dimensional ultrasonography), and blood glucose and plasma gut-hormone concentrations [insulin, glucagon, ghrelin, cholecystokinin, gastric inhibitory polypeptide (GIP), glucagon-like peptide 1 (GLP-1), and peptide tyrosine tyrosine (PYY)] were measured, and ad libitum energy intake was quantified from a buffet meal (180-210 min; energy intake, appetite, and gastric emptying in the men have been published previously).Results: Energy intake at the buffet meal was ∼80% higher in older men than in older women (P < 0.001). Energy intake was not suppressed by protein compared with the control in men or women (P > 0.05). There was no effect of sex on gastric emptying, appetite, gastrointestinal symptoms, glucose, or gut hormones (P > 0.05). There was a protein load-dependent slowing of gastric emptying, an increase in concentrations of insulin, glucagon, cholecystokinin, GIP, GLP-1, and PYY, and an increase in total energy intake (drink plus meal: 12% increase with 30 g and 32% increase with 70 g; P < 0.001). Energy intake at the buffet meal was inversely related to the stomach volume and area under the curve of hormone concentrations (P < 0.05).Conclusion: In older men and women, whey-protein drinks load-dependently slow gastric emptying and alter gut hormone secretion compared with a control but have no suppressive effect on subsequent ad libitum energy intake. This trial was registered at www.anzctr.org.au as ACTRN12612000941864.
Collapse
Affiliation(s)
- Caroline Giezenaar
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Laurence G Trahair
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Natalie D Luscombe-Marsh
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
- The Commonwealth Scientific and Industrial Research Organisation, Animal, Food and Health Sciences, Adelaide, South Australia, Australia; and
| | - Trygve Hausken
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Scott Standfield
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Karen L Jones
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Kylie Lange
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Michael Horowitz
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Ian Chapman
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Stijn Soenen
- Discipline of Medicine and National Health and Medical Research Council of Australia Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia;
| |
Collapse
|
368
|
Abstract
PURPOSE OF REVIEW In recent years, the role of the gastrointestinal (GI) tract in energy homeostasis through modulation of the digestion and absorption of carbohydrates and the production of incretin hormones is well recognized. RECENT FINDINGS Bariatric surgery for obesity has been a very effective method in substantially improving weight, and numerous studies have focused on intestinal adaptation after bariatric procedures. A number of structural and functional changes in the GI tract have been reported postsurgery, which could be responsible for the altered hormonal responses. Furthermore, the change in food absorption rate and the intestinal regions exposed to carbohydrates may affect blood glucose response. This review hopes to give new insights into the direct role of gut hormones, by summarising the metabolic effects of bariatric surgery.
Collapse
Affiliation(s)
- Georgios K Dimitriadis
- Division of Translational and Experimental Medicine, Clinical Sciences Research Laboratories, University of Warwick Medical School, Coventry, CV2 2DX, UK.
- Academic Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, W12 0NN, UK.
- Division of Translational and Experimental Medicine-Metabolic and Vascular Health, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| | - Manpal S Randeva
- Division of Translational and Experimental Medicine, Clinical Sciences Research Laboratories, University of Warwick Medical School, Coventry, CV2 2DX, UK
| | - Alexander D Miras
- Academic Division of Diabetes, Endocrinology and Metabolism, Imperial College London, Hammersmith Campus, London, W12 0NN, UK
| |
Collapse
|
369
|
Andermann ML, Lowell BB. Toward a Wiring Diagram Understanding of Appetite Control. Neuron 2017; 95:757-778. [PMID: 28817798 DOI: 10.1016/j.neuron.2017.06.014] [Citation(s) in RCA: 329] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 06/02/2017] [Accepted: 06/07/2017] [Indexed: 01/26/2023]
Abstract
Prior mouse genetic research has set the stage for a deep understanding of appetite regulation. This goal is now being realized through the use of recent technological advances, such as the ability to map connectivity between neurons, manipulate neural activity in real time, and measure neural activity during behavior. Indeed, major progress has been made with regard to meal-related gut control of appetite, arcuate nucleus-based hypothalamic circuits linking energy state to the motivational drive, hunger, and, finally, limbic and cognitive processes that bring about hunger-mediated increases in reward value and perception of food. Unexpected findings are also being made; for example, the rapid regulation of homeostatic neurons by cues that predict future food consumption. The aim of this review is to cover the major underpinnings of appetite regulation, describe recent advances resulting from new technologies, and synthesize these findings into an updated view of appetite regulation.
Collapse
Affiliation(s)
- Mark L Andermann
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
370
|
|
371
|
Li W, Richard D. Effects of Bariatric Surgery on Energy Homeostasis. Can J Diabetes 2017; 41:426-431. [DOI: 10.1016/j.jcjd.2017.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/11/2017] [Accepted: 05/04/2017] [Indexed: 12/11/2022]
|
372
|
The impact of gut hormones on the neural circuit of appetite and satiety: A systematic review. Neurosci Biobehav Rev 2017; 80:457-475. [PMID: 28669754 DOI: 10.1016/j.neubiorev.2017.06.013] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 06/08/2017] [Accepted: 06/27/2017] [Indexed: 01/01/2023]
Abstract
The brain-gut-axis is an interdependent system affecting neural functions and controlling our eating behaviour. In recent decades, neuroimaging techniques have facilitated its investigation. We systematically looked into functional and neurochemical brain imaging studies investigating how key molecules such as ghrelin, glucagon-like peptide-1 (GLP-1), peptide tyrosine-tyrosine (PYY), cholecystokinin (CCK), leptin, glucose and insulin influence the function of brain regions regulating appetite and satiety. Of the 349 studies published before July 2016 identified in the database search, 40 were included (27 on healthy and 13 on obese subjects). Our systematic review suggests that the plasma level of ghrelin, the gut hormone promoting appetite, is positively correlated with activation in the pre-frontal cortex (PFC), amygdala and insula and negatively correlated with activation in subcortical areas such as the hypothalamus. In contrast, the plasma levels of glucose, insulin, leptin, PYY, GLP-1 affect the same brain regions conversely. Our study integrates previous investigations of the gut-brain matrix during food-intake and homeostatic regulation and may be of use for future meta-analyses of brain-gut interactions.
Collapse
|
373
|
Rock CL, Flatt SW, Barkai HS, Pakiz B, Heath DD. A walnut-containing meal had similar effects on early satiety, CCK, and PYY, but attenuated the postprandial GLP-1 and insulin response compared to a nut-free control meal. Appetite 2017; 117:51-57. [PMID: 28610906 PMCID: PMC5822690 DOI: 10.1016/j.appet.2017.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/05/2017] [Accepted: 06/09/2017] [Indexed: 11/02/2022]
Abstract
Regular nut consumption is associated with lower adiposity and reduced weight gain in adulthood. Walnut feeding studies have observed minimal effect on body weight despite potential additional energy intake. Several mechanisms may explain why consuming nuts promotes weight control, including increased early phase satiety, possibly reflected in postprandial response of gastrointestinal and pancreatic peptides hypothesized to affect appetite. The purpose of this study was to compare postprandial insulin, glucagon and gastrointestinal peptide response and satiety following a meal with ∼54% of energy from walnuts or cream cheese, using a within-subject crossover study design in overweight/obese adults (N = 28). Sixty minutes after the walnut-containing meal, glucagon-like peptide-1 was lower than after the reference meal (p=0.0433), and peptide YY, cholecystokinin and ghrelin did not differ after the two meals. Sixty and 120 min after the walnut-containing meal, pancreatic polypeptide (p = 0.0014 and p = 0.0002) and glucose-dependent insulinotropic peptide (p < 0.0001 and p = 0.0079) were lower than after the reference meal, and 120 min after the walnut-containing meal, glucagon was higher (p=0.0069). Insulin and C-peptide increased at 60 min in response to both meals but were lower at 120 min after the walnut-containing meal (p=0.0349 and 0.0237, respectively). Satiety measures were similar after both meals. These findings fail to support the hypothesis that acute postprandial gastrointestinal peptide response to a walnut-containing meal contributes to increased satiety. However, inclusion of walnuts attenuated the postprandial insulin response, which may contribute to the more favorable lipid profile observed in association with regular walnut consumption.
Collapse
Affiliation(s)
- Cheryl L Rock
- Department of Family Medicine and Public Health, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0901, USA.
| | - Shirley W Flatt
- Department of Family Medicine and Public Health, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0901, USA
| | - Hava-Shoshana Barkai
- Department of Family Medicine and Public Health, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0901, USA
| | - Bilge Pakiz
- Department of Family Medicine and Public Health, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0901, USA
| | - Dennis D Heath
- Department of Family Medicine and Public Health, University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093-0901, USA
| |
Collapse
|
374
|
Luo QQ, Zhou YF, Chen MYJ, Liu L, Ma J, Zhang MW, Zhang FL, Ke Y, Qian ZM. Fasting up-regulates ferroportin 1 expression via a Ghrelin/GHSR/MAPK signaling pathway. J Cell Physiol 2017; 233:30-37. [PMID: 28338217 DOI: 10.1002/jcp.25931] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023]
Abstract
The significant positive correlation between ghrelin and iron and hepcidin levels in the plasma of children with iron deficiency anemia prompted us to hypothesize that ghrelin may affect iron metabolism. Here, we investigated the effects of fasting or ghrelin on the expression of hepcidin, ferroportin 1 (Fpn1), transferrin receptor 1 (TfR1), ferritin light chain (Ft-L) proteins, and ghrelin, and also hormone secretagogue receptor 1 alpha (GHSR1α) and ghrelin O-acyltransferase (GOAT) mRNAs in the spleen and/or macrophage. We demonstrated that fasting induces a significant increase in the expression of ghrelin, GHSR1α, GOAT, and hepcidin mRNAs, as well as Ft-L and Fpn1 but not TfR1 proteins in the spleens of mice in vivo. Similar to the effects of fasting on the spleen, ghrelin induced a significant increase in the expression of Ft-L and Fpn1 but not TfR1 proteins in macrophages in vitro. In addition, ghrelin was found to induce a significant enhancement in phosphorylation of ERK as well as translocation of pERK from the cytosol to nuclei. Furthermore, the increased pERK and Fpn1 induced by ghrelin was demonstrated to be preventable by pre-treatment with either GHSR1α antagonist or pERK inhibitor. Our findings support the hypothesis that fasting upregulates Fpn1 expression, probably via a ghrelin/GHSR/MAPK signaling pathway.
Collapse
Affiliation(s)
- Qian-Qian Luo
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China.,Pharmacological Evaluation and Research Center, Shanghai Institute of PharmaceuticalIndustry, Shanghai, China.,Department of Biochemistry, Institute for Nautical Medicine, Nantong University, Nantong, China
| | - Yu-Fu Zhou
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Mesona Yung-Jin Chen
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Li Liu
- Pharmacological Evaluation and Research Center, Shanghai Institute of PharmaceuticalIndustry, Shanghai, China
| | - Juan Ma
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Meng-Wan Zhang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Fa-Li Zhang
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Ya Ke
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Zhong-Ming Qian
- Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai, China
| |
Collapse
|
375
|
Affiliation(s)
- Nori Geary
- Department of Psychiatry, Weill Cornell Medical College, New York, NY
| |
Collapse
|