1
|
Tso P, Bernier-Latmani J, Petrova TV, Liu M. Transport functions of intestinal lymphatic vessels. Nat Rev Gastroenterol Hepatol 2025; 22:127-145. [PMID: 39496888 DOI: 10.1038/s41575-024-00996-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 11/06/2024]
Abstract
Lymphatic vessels are crucial for fluid absorption and the transport of peripheral immune cells to lymph nodes. However, in the small intestine, the lymphatic fluid is rich in diet-derived lipids incorporated into chylomicrons and gut-specific immune cells. Thus, intestinal lymphatic vessels have evolved to handle these unique cargoes and are critical for systemic dietary lipid delivery and metabolism. This Review covers mechanisms of lipid absorption from epithelial cells to the lymphatics as well as unique features of the gut microenvironment that affect these functions. Moreover, we discuss details of the intestinal lymphatics in gut immune cell trafficking and insights into the role of inter-organ communication. Lastly, we highlight the particularities of fat absorption that can be harnessed for efficient lipid-soluble drug distribution for novel therapies, including the ability of chylomicron-associated drugs to bypass first-pass liver metabolism for systemic delivery. In all, this Review will help to promote an understanding of intestinal lymphatic-systemic interactions to guide future research directions.
Collapse
Affiliation(s)
- Patrick Tso
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - Jeremiah Bernier-Latmani
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Min Liu
- Department of Pathology & Laboratory Medicine, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
2
|
Cao N, Merchant W, Gautron L. Limited evidence for anatomical contacts between intestinal GLP-1 cells and vagal neurons in male mice. Sci Rep 2024; 14:23666. [PMID: 39390033 PMCID: PMC11467209 DOI: 10.1038/s41598-024-74000-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
The communication between intestinal Glucagon like peptide 1 (GLP-1)-producing cells and the peripheral nervous system has garnered renewed interest considering the availability of anti-obesity and anti-diabetic approaches targeting GLP-1 signaling. While it is well-established that intestinal GLP-1 cells can exert influence through paracrine mechanisms, recent evidence suggests the possible existence of synaptic-like connections between GLP-1 cells and peripheral neurons, including those of the vagus nerve. In this study, using a reporter Phox2b-Cre-Tomato mouse model and super-resolution confocal microscopy, we demonstrated that vagal axons made apparent contacts with less than 0.5% of GLP-1 cells. Moreover, immunohistochemistry combined with super-resolution confocal microscopy revealed abundant post-synaptic density 95 (PSD-95) immunoreactivity within the enteric plexus of the lower intestines of C57/BL6 mice, with virtually none in its mucosa. Lastly, utilizing RNAScope in situ hybridization in the lower intestines of mice, we observed that GLP-1 cells expressed generic markers of secretory cells such as Snap25 and Nefm, but neither synaptic markers such as Syn1 and Nrxn2, nor glutamatergic markers such as Slc17a7. Through theoretical considerations and a critical review of the literature, we concluded that intestinal GLP-1 cells primarily communicate with vagal neurons through paracrine mechanisms, rather than synaptic-like contacts.
Collapse
Affiliation(s)
- Newton Cao
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Warda Merchant
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Laurent Gautron
- Department of Internal Medicine, Center for Hypothalamic Research, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
3
|
Morrow NM, Morissette A, Mulvihill EE. Immunomodulation and inflammation: Role of GLP-1R and GIPR expressing cells within the gut. Peptides 2024; 176:171200. [PMID: 38555054 DOI: 10.1016/j.peptides.2024.171200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 04/02/2024]
Abstract
Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are peptide hormones produced by enteroendocrine cells in the small intestine. Despite being produced in the gut, the leveraging of their role in potentiating glucose-stimulated insulin secretion, also known as the incretin effect, has distracted from discernment of direct intestinal signaling circuits. Both preclinical and clinical evidence have highlighted a role for the incretins in inflammation. In this review, we highlight the discoveries of GLP-1 receptor (GLP-1R)+ natural (TCRαβ and TCRγδ) and induced (TCRαβ+CD4+ cells and TCRαβ+CD8αβ+) intraepithelial lymphocytes. Both endogenous signaling and pharmacological activation of GLP-1R impact local and systemic inflammation, the gut microbiota, whole-body metabolism, as well as the control of GLP-1 bioavailability. While GIPR signaling has been documented to impact hematopoiesis, the impact of these bone marrow-derived cells in gut immunology is not well understood. We uncover gaps in the literature of the evaluation of the impact of sex in these GLP-1R and GIP receptor (GIPR) signaling circuits and provide speculations of the maintenance roles these hormones play within the gut in the fasting-refeeding cycles. GLP-1R agonists and GLP-1R/GIPR agonists are widely used as treatments for diabetes and weight loss, respectively; however, their impact on gut homeostasis has not been fully explored. Advancing our understanding of the roles of GLP-1R and GIPR signaling within the gut at homeostasis as well as metabolic and inflammatory diseases may provide targets to improve disease management.
Collapse
Affiliation(s)
- Nadya M Morrow
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario KIY 4W7, Canada; Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Faculty of Medicine, 451 Smyth Rd, Ottawa, Ontario K1H 8L1, Canada
| | - Arianne Morissette
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario KIY 4W7, Canada
| | - Erin E Mulvihill
- The University of Ottawa Heart Institute, 40 Ruskin Street, H-3229A, Ottawa, Ontario KIY 4W7, Canada; Department of Biochemistry, Microbiology and Immunology, The University of Ottawa, Faculty of Medicine, 451 Smyth Rd, Ottawa, Ontario K1H 8L1, Canada; Centre for Infection, Immunity and Inflammation, Ottawa, Ontario, Canada; Montreal Diabetes Research Group, Montreal, Quebec, Canada; Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada.
| |
Collapse
|
4
|
Smits MM, Holst JJ. Endogenous glucagon-like peptide (GLP)-1 as alternative for GLP-1 receptor agonists: Could this work and how? Diabetes Metab Res Rev 2023; 39:e3699. [PMID: 37485788 DOI: 10.1002/dmrr.3699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/21/2023] [Accepted: 06/18/2023] [Indexed: 07/25/2023]
Abstract
In recent years, we have witnessed the many beneficial effects of glucagon-like peptide (GLP)-1 receptor agonists, including the reduction in cardiovascular risk in patients with type 2 diabetes, and the reduction of body weight in those with obesity. Increasing evidence suggests that these agents differ considerably from endogenous GLP-1 when it comes to their routes of action, although their clinical effects appear to be the same. Given the limitations of the GLP-1 receptor agonists, could it be useful to develop agents which stimulate GLP-1 release? Here we will discuss the differences and similarities between GLP-1 receptor agonists and endogenous GLP-1, and will detail how endogenous GLP-1-when stimulated appropriately-could have clinically relevant effects.
Collapse
Affiliation(s)
- Mark M Smits
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Amsterdam Cardiovascular Sciences, Diabetes and Metabolism, Amsterdam, The Netherlands
- Department of Internal Medicine, Diabetes Center, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
5
|
Trzaskalski NA, Vulesevic B, Nguyen MA, Jeraj N, Fadzeyeva E, Morrow NM, Locatelli CA, Travis N, Hanson AA, Nunes JR, O’Dwyer C, van der Veen JN, Lorenzen-Schmidt I, Seymour R, Pulente SM, Clément AC, Crawley AM, Jacobs RL, Doyle MA, Cooper CL, Kim KH, Fullerton MD, Mulvihill EE. Hepatocyte-derived DPP4 regulates portal GLP-1 bioactivity, modulates glucose production, and when absent influences NAFLD progression. JCI Insight 2023; 8:154314. [PMID: 36472923 PMCID: PMC9977314 DOI: 10.1172/jci.insight.154314] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Elevated circulating dipeptidyl peptidase-4 (DPP4) is a biomarker for liver disease, but its involvement in gluconeogenesis and metabolic associated fatty liver disease progression remains unclear. Here, we identified that DPP4 in hepatocytes but not TEK receptor tyrosine kinase-positive endothelial cells regulates the local bioactivity of incretin hormones and gluconeogenesis. However, the complete absence of DPP4 (Dpp4-/-) in aged mice with metabolic syndrome accelerates liver fibrosis without altering dyslipidemia and steatosis. Analysis of transcripts from the livers of Dpp4-/- mice displayed enrichment for inflammasome, p53, and senescence programs compared with littermate controls. High-fat, high-cholesterol feeding decreased Dpp4 expression in F4/80+ cells, with only minor changes in immune signaling. Moreover, in a lean mouse model of severe nonalcoholic fatty liver disease, phosphatidylethanolamine N-methyltransferase mice, we observed a 4-fold increase in circulating DPP4, in contrast with previous findings connecting DPP4 release and obesity. Last, we evaluated DPP4 levels in patients with hepatitis C infection with dysglycemia (Homeostatic Model Assessment of Insulin Resistance > 2) who underwent direct antiviral treatment (with/without ribavirin). DPP4 protein levels decreased with viral clearance; DPP4 activity levels were reduced at long-term follow-up in ribavirin-treated patients; but metabolic factors did not improve. These data suggest elevations in DPP4 during hepatitis C infection are not primarily regulated by metabolic disturbances.
Collapse
Affiliation(s)
- Natasha A. Trzaskalski
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Branka Vulesevic
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - My-Anh Nguyen
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Natasha Jeraj
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Evgenia Fadzeyeva
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Nadya M. Morrow
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Cassandra A.A. Locatelli
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Nicole Travis
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Antonio A. Hanson
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Julia R.C. Nunes
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, Ottawa, Ontario, Canada.,Centre for Catalysis Research and Innovation, Ottawa, Ontario, Canada
| | - Conor O’Dwyer
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, Ottawa, Ontario, Canada.,Centre for Catalysis Research and Innovation, Ottawa, Ontario, Canada
| | - Jelske N. van der Veen
- Li Ka Shing (LKS) Centre for Health Research Innovation, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | | | - Rick Seymour
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Serena M. Pulente
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Andrew C. Clément
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Angela M. Crawley
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, Ottawa, Ontario, Canada.,Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biology, Carleton University, Ottawa, Ontario, Canada
| | - René L. Jacobs
- Li Ka Shing (LKS) Centre for Health Research Innovation, Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Mary-Anne Doyle
- Division of Endocrinology & Metabolism, Department of Medicine
| | - Curtis L. Cooper
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Division of Infectious Diseases, Department of Medicine, and
| | - Kyoung-Han Kim
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Morgan D. Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, Ottawa, Ontario, Canada.,Centre for Catalysis Research and Innovation, Ottawa, Ontario, Canada
| | - Erin E. Mulvihill
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ontario, Canada.,University of Ottawa Heart Institute, Ottawa, Ontario, Canada.,Centre for Infection, Immunity and Inflammation, Ottawa, Ontario, Canada.,Montréal Diabetes Research Group, Montréal, Québec, Canada
| |
Collapse
|
6
|
Effect of L-Glutamine on Chylomicron Formation and Fat-Induced Activation of Intestinal Mucosal Mast Cells in Sprague-Dawley Rats. Nutrients 2022; 14:nu14091777. [PMID: 35565745 PMCID: PMC9104139 DOI: 10.3390/nu14091777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 01/27/2023] Open
Abstract
Glutamine (Gln) is required for intestinal mucosal homeostasis, and it can promote triglyceride absorption. The intestinal mucosal mast cells (MMCs) are activated during fat absorption. This study investigated the potential role of Gln on fat absorption-induced activation of MMCs in rats. Lymph fistula rats (n = 24) were studied after an overnight recovery with the infusion of saline only, saline plus 85 mM L-glutamine (L-Gln) or 85 mM D-glutamine (D-Gln), respectively. On the test day, rats (n = 8/group) were given an intraduodenal bolus of 20% Intralipid contained either saline only (vehicle group), 85 mM L-Gln (L-Gln group), or 85 mM D-Gln (D-Gln group). Lymph was collected hourly for up to 6 h for analyses. The results showed that intestinal lymph from rats given L-Gln had increased levels of apolipoprotein B (ApoB) and A-I (ApoA-I), concomitant with an increased spectrum of smaller chylomicron particles. Unexpectedly, L-Gln also increased levels of rat mucosal mast cell protease II (RMCPII), as well as histamine and prostaglandin D2 (PGD2) in response to dietary lipid. However, these effects were not observed in rats treated with 85 mM of the stereoisomer D-Gln. Our results showed that L-glutamine could specifically activate MMCs to degranulate and release MMC mediators to the lymph during fat absorption. This observation is potentially important clinically since L-glutamine is often used to promote gut health and repair leaky gut.
Collapse
|
7
|
Morrow NM, Hanson AA, Mulvihill EE. Distinct Identity of GLP-1R, GLP-2R, and GIPR Expressing Cells and Signaling Circuits Within the Gastrointestinal Tract. Front Cell Dev Biol 2021; 9:703966. [PMID: 34660576 PMCID: PMC8511495 DOI: 10.3389/fcell.2021.703966] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 08/16/2021] [Indexed: 12/17/2022] Open
Abstract
Enteroendocrine cells directly integrate signals of nutrient content within the gut lumen with distant hormonal responses and nutrient disposal via the production and secretion of peptides, including glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide 1 (GLP-1) and glucagon-like peptide 2 (GLP-2). Given their direct and indirect control of post-prandial nutrient uptake and demonstrated translational relevance for the treatment of type 2 diabetes, malabsorption and cardiometabolic disease, there is significant interest in the locally engaged circuits mediating these metabolic effects. Although several specific populations of cells in the intestine have been identified to express endocrine receptors, including intraepithelial lymphocytes (IELs) and αβ and γδ T-cells (Glp1r+) and smooth muscle cells (Glp2r+), the definitive cellular localization and co-expression, particularly in regards to the Gipr remain elusive. Here we review the current state of the literature and evaluate the identity of Glp1r, Glp2r, and Gipr expressing cells within preclinical and clinical models. Further elaboration of our understanding of the initiating G-protein coupled receptor (GPCR) circuits engaged locally within the intestine and how they become altered with high-fat diet feeding can offer insight into the dysregulation observed in obesity and diabetes.
Collapse
Affiliation(s)
- Nadya M Morrow
- Energy Substrate Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Antonio A Hanson
- Energy Substrate Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Erin E Mulvihill
- Energy Substrate Laboratory, University of Ottawa Heart Institute, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.,Montreal Diabetes Research Center CRCHUM-Pavillion R, Montreal, QC, Canada.,Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
8
|
Watkins JD, Koumanov F, Gonzalez JT. Protein- and Calcium-Mediated GLP-1 Secretion: A Narrative Review. Adv Nutr 2021; 12:2540-2552. [PMID: 34192748 PMCID: PMC8634310 DOI: 10.1093/advances/nmab078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Glucagon-like peptide 1 (GLP-1) is an incretin hormone produced in the intestine that is secreted in response to nutrient exposure. GLP-1 potentiates glucose-dependent insulin secretion from the pancreatic β cells and promotes satiety. These important actions on glucose metabolism and appetite have led to widespread interest in GLP-1 receptor agonism. Typically, this involves pharmacological GLP-1 mimetics or targeted inhibition of dipeptidyl peptidase-IV, the enzyme responsible for GLP-1 degradation. However, nutritional strategies provide a widely available, cost-effective alternative to pharmacological strategies for enhancing hormone release. Recent advances in nutritional research have implicated the combined ingestion of protein and calcium with enhanced endogenous GLP-1 release, which is likely due to activation of receptors with high affinity and/or sensitivity for amino acids and calcium. Specifically targeting these receptors could enhance gut hormone secretion, thus providing a new therapeutic option. This narrative review provides an overview of the latest research on protein- and calcium-mediated GLP-1 release with an emphasis on human data, and a perspective on potential mechanisms that link potent GLP-1 release to the co-ingestion of protein and calcium. In light of these recent findings, potential future research directions are also presented.
Collapse
Affiliation(s)
- Jonathan D Watkins
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, United Kingdom
| | - Françoise Koumanov
- Centre for Nutrition, Exercise and Metabolism, Department for Health, University of Bath, Bath, United Kingdom
| | | |
Collapse
|
9
|
Braga TG, Graças Coelho de Souza MD, Menezes M, Nogueira Neto JF, Dellatorre-Teixeira L, Bouskela E, le Roux CW, Kraemer-Aguiar LG. Dipeptidyl peptidase-4 activity, lipopolysaccharide, C-reactive protein, glucose metabolism, and gut peptides 3 months after bariatric surgery. Surg Obes Relat Dis 2020; 17:113-120. [PMID: 33036944 DOI: 10.1016/j.soard.2020.08.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND Bariatric surgery induces weight loss, but changes in glucose metabolism, gut peptides, and inflammatory biomarkers still have conflicting results. SETTINGS University hospital. OBJECTIVES We investigated glucose metabolism, gut hormones, and inflammatory profile after bariatric surgery and medical treatment. METHODS Forty patients with obesity were recruited and were subjected to Roux-en-Y gastric bypass (n = 15; Bariatric Surgery Group - BSG) or received medical care (n = 20; MG). Sleeve gastrectomy was performed in five patients who were excluded from analysis. Glucose, insulin, homeostatic model for the assessment of insulin resistance (HOMA-IR), glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), glucagon, ghrelin, dipeptidyl peptidase-4 (DPP-4) activity, circulating lipopolysaccharide (LPS), LPS-binding protein (LPB) and high-sensitivity C-reactive protein (hs-CRP) were evaluated before and three months after each treatment. Except for HOMA-IR, hs-CRP, and LBP, all variables were assessed at fasting and 30- and 60-minutes after a standard meal. RESULTS After 3 months, both groups lost weight. However, BSG had a more extensive reduction than MG (respectively, 17.6% vs. 4.25%; P < 0.01). Except for LPS levels, higher on BSG than MG (1.38 ± 0.96 vs. 0.83 ± 0.60 EU/ml, P < 0.01), groups were similar before treatment. In respect to metabolic/hormonal changes, the BSG showed higher glucose, insulin, GLP-1, and GIP levels at 30-min and also GLP-1 at 30- and 60-minutes. DPP-4 activity, HOMA-IR, and fasting LBP did not change. LPS levels at 60-minutes decreased after surgery in the BSG. hs-CRP decreased on BSG compared to MG. CONCLUSIONS Bariatric surgery resulted in more extensive effects on glucose metabolism, gut hormones, and inflammation.
Collapse
Affiliation(s)
- Tassia Gomide Braga
- Postgraduate Program in Clinical and Experimental Physiopathology, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Das Graças Coelho de Souza
- Laboratory of Clinical and Experimental Research on Vascular Biology, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Eliete Bouskela
- Laboratory of Clinical and Experimental Research on Vascular Biology, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carel W le Roux
- Diabetes Complications Research Centre Conway Institute University College, Dublin, Ireland; Investigate Science, Imperial College, London, United Kingdom
| | - Luiz Guilherme Kraemer-Aguiar
- Laboratory of Clinical and Experimental Research on Vascular Biology, Biomedical Center, State University of Rio de Janeiro, Rio de Janeiro, Brazil; Obesity Unit, Department of Internal Medicine, Faculty of Medical Sciences, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
10
|
Mayer F, Gunawan AL, Tso P, Aponte GW. Glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide stimulate release of substance P from TRPV1- and TRPA1-expressing sensory nerves. Am J Physiol Gastrointest Liver Physiol 2020; 319:G23-G35. [PMID: 32421358 PMCID: PMC7468754 DOI: 10.1152/ajpgi.00189.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are released from enteroendocrine cells (EECs) in response to nutrient ingestion and lower blood glucose levels by stimulation of insulin secretion and thus are defined as incretins. GLP-1 receptor (GLP-1R) expression has been identified on enteric neurons that include intrinsic afferent neurons, extrinsic spinal, and vagal sensory afferents but has not been shown to have an incretin effect through these nerves. GLP-1 and GIP enter the mesenteric lymphatic fluid (MLF) after a meal via the interstitial fluid (IF) from local tissue secretion and/or blood capillaries. We tested if MLF could induce diet-dependent intransient increases in intracellular calcium ([Ca2+]i) in cultured sensory neurons. Postprandial rat MLF, collected from the superior mesenteric lymphatic duct, induced a significant twofold higher intransient increase in [Ca2+]i in primary-cultured sensory neurons than MLF from fasted rats. Inhibition of transient receptor potential vanilloid 1 (TRPV1) and TRPV1 and ankyrin 1 cation channels (TRPA1) with ruthenium red eliminated the difference. Substance P (SP) (a peptide that stimulates insulin secretion) sensor cells cocultured with sensory neurons showed both the GLP-1R agonist exendin-4 (Ex-4) and GIP induced transient increases in [Ca2+]i directly coupled to SP secretion in the sensory nerves. Ex-4-induced release of SP required expression of either TRPA1 or TRPV1. These data identify unrecognized actions of GLP-1 and GIP as incretins by acting as neurolymphocrines and suggest a mechanism for sensory nerves to respond to the postprandial state through MLF.NEW & NOTEWORTHY Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted upon eating to lower blood sugar. GLP-1 and GIP were found to induce the secretion of substance P (SP) from cultured sensory nerves. SP enhances insulin secretion. Mesenteric lymphatic fluid (MLF) also stimulates sensory neurons in a diet-dependent manner. These studies identify new actions of GLP-1 and GIP as incretins and suggest a mechanism for sensory nerves to respond to diet through MLF.
Collapse
Affiliation(s)
- Fahima Mayer
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| | - Amanda L. Gunawan
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| | - Patrick Tso
- 2Department of Pathobiology and Molecular Medicine, University of Cincinnati, Reading, Ohio
| | - Gregory W. Aponte
- 1Department of Nutritional Sciences and Toxicology, Graduate Program in Metabolic Biology, University of California, Berkeley, California
| |
Collapse
|
11
|
Davis EM, Sandoval DA. Glucagon‐Like Peptide‐1: Actions and Influence on Pancreatic Hormone Function. Compr Physiol 2020; 10:577-595. [DOI: 10.1002/cphy.c190025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
12
|
Aulinger BA, Perabo M, Seeley RJ, Parhofer KG, D'Alessio DA. Rapid hepatic metabolism blunts the endocrine action of portally infused GLP-1 in male rats. Am J Physiol Endocrinol Metab 2020; 318:E189-E197. [PMID: 31743041 PMCID: PMC7052580 DOI: 10.1152/ajpendo.00298.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) is an enteral peptide that contributes to the incretin effect. GLP-1 action is typically described as endocrine, but this mechanism has been questioned because rapid inactivation in the circulation by dipeptidylpeptidase 4 (DPP4) results in a short half-life, limiting the amount of the hormone that can reach the pancreatic islet. An alternative mechanism for GLP-1 to regulate insulin secretion through neuroendocrine signaling originating from sensors in the portal vein has been proposed. We hypothesized that portal infusion of GLP-1 would cause greater glucose-stimulated insulin secretion than equimolar administration into the jugular vein. To test this, hyperglycemic clamps with superimposed graded infusions of GLP-1 into the jugular or portal veins of male rats were performed. These experiments were repeated with pharmacologic DPP4 inhibition to determine the effect of GLP-1 metabolism in the jugular and portal venous beds. Contrary to our hypothesis, we found a higher insulinotropic effect with jugular compared with portal GLP-1, which was associated with higher plasma concentrations of intact GLP-1. The greater insulinotropic effect of jugular venous GLP-1 persisted even with pharmacological DPP4 inhibition. These findings do not support an important role of portal vein GLP-1 signaling for the incretin effect but highlight the hepatoportal bed as a major site of GLP-1 degradation that persists even with pharmacological inhibition. Together, these results support rapid inactivation of enterally released GLP-1 in the liver as limiting endocrine actions on the β-cell and raise questions about the conventional endocrine model of pharmacologic effects of DPP4 inhibitors.
Collapse
Affiliation(s)
- Benedikt A Aulinger
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Department of Medicine II, University Hospital, LMU Munich, Germany
| | - Marta Perabo
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Klaus G Parhofer
- Department of Medicine IV, University Hospital, LMU Munich, Germany
| | - David A D'Alessio
- Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
13
|
Yoshida Y, Gotoh K, Masaki T, Ozeki Y, Tokoro M, Kudo A, Ozaki T, Okamoto M, Chiba S, Watanabe K, Ohta M, Inomata M, Shibata H. Effects of Sleeve Gastrectomy on Blood Pressure and the Renal Renin-Angiotensin System in Rats with Diet-Induced Obesity. Obesity (Silver Spring) 2019; 27:785-792. [PMID: 30925198 DOI: 10.1002/oby.22443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 01/15/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Sleeve gastrectomy (SG) has been reported to decrease blood pressure (BP), although the reason has not been revealed. The present study aimed to establish the reason why SG decreases BP. METHODS Male Sprague-Dawley rats were subjected to surgical (sham operation or SG) and dietary interventions (fed a normal diet or high-fat diet ad libitum or fed by pair-feeding [PF]). Systolic BP (SBP), urinary sodium excretion, and endocrine parameters were examined 4 weeks after surgery. RESULTS Both SG and PF rats had reduced body weight compared with SO rats fed normal diet or high-fat diet ad libitum. SG rats exhibited a reduction in SBP compared with PF, which was associated with a reduction in renal renin, angiotensin II, and catechol-O-methyltransferase levels (P < 0.01 for each). SG increased plasma cholecystokinin (CCK) levels compared with PF (P < 0.0001 for each), whereas glucagon-like peptide 1 and peptide YY were not changed in fasting. Exogenous administration of CCK reduced renal catechol-O-methyltransferase (P = 0.0233), renin (P < 0.0001), and angiotensin II (P < 0.0001) levels and SBP (P = 0.0053). CONCLUSIONS SG reduced SBP, at least in part, through suppression of sympathetic nerve action by elevation of CCK, which was followed by suppression of the intrarenal renin-angiotensin system.
Collapse
Affiliation(s)
- Yuichi Yoshida
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Koro Gotoh
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Takayuki Masaki
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Yoshinori Ozeki
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Masanori Tokoro
- Department of Gastroenterology, Faculty of Medicine, Oita University, Oita, Japan
| | - Akiko Kudo
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Takashi Ozaki
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Mitsuhiro Okamoto
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| | - Seiichi Chiba
- Department of Molecular Anatomy, Faculty of Medicine, Oita University, Oita, Japan
| | - Kiminori Watanabe
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Masayuki Ohta
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Masafumi Inomata
- Department of Gastroenterological and Pediatric Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology, and Nephrology, Faculty of Medicine, Oita University, Oita, Japan
| |
Collapse
|
14
|
Finn PD, Rodriguez D, Kohler J, Jiang Z, Wan S, Blanco E, King AJ, Chen T, Bell N, Dragoli D, Jacobs JW, Jain R, Leadbetter M, Siegel M, Carreras CW, Koo-McCoy S, Shaw K, Le C, Vanegas S, Hsu IH, Kozuka K, Okamoto K, Caldwell JS, Lewis JG. Intestinal TGR5 agonism improves hepatic steatosis and insulin sensitivity in Western diet-fed mice. Am J Physiol Gastrointest Liver Physiol 2019; 316:G412-G424. [PMID: 30605011 PMCID: PMC6459286 DOI: 10.1152/ajpgi.00300.2018] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Takeda G protein-coupled receptor 5 (TGR5) agonists induce systemic release of glucagon-like peptides (GLPs) from intestinal L cells, a potentially therapeutic action against metabolic diseases such as nonalcoholic steatohepatitis (NASH), nonalcoholic fatty liver disease (NAFLD), and Type 2 diabetes. Historically, TGR5 agonist use has been hindered by side effects, including inhibition of gallbladder emptying. Here, we characterize RDX8940, a novel, orally administered TGR5 agonist designed to have minimal systemic effects and investigate its activity in mice fed a Western diet, a model of NAFLD and mild insulin resistance. Agonist activity, binding selectivity, toxicity, solubility, and permeability of RDX8940 were characterized in standard in vitro models. RDX8940 pharmacokinetics and effects on GLP secretion, insulin sensitivity, and liver steatosis were assessed in C57BL/6 mice fed normal or Western diet chow and given single or repeated doses of RDX8940 or vehicle, with or without dipeptidyl peptidase-4 (DPP4) inhibitors. Gallbladder effects were assessed in CD-1 mice fed normal chow and given RDX8940 or a systemic TGR5 agonist or vehicle. Our results showed that RDX8940 is minimally systemic, potent, and selective, and induces incretin (GLP-1, GLP-2, and peptide YY) secretion. RDX8940-induced increases in plasma active GLP-1 (aGLP-1) levels were enhanced by repeated dosing and by coadministration of DPP4 inhibitors. RDX8940 increased hepatic exposure to aGLP-1 without requiring coadministration of a DPP4 inhibitor. In mice fed a Western diet, RDX8940 improved liver steatosis and insulin sensitivity. Unlike systemic TGR5 agonists, RDX8940 did not inhibit gallbladder emptying. These results indicate that RDX8940 may have therapeutic potential in patients with NAFLD/NASH. NEW & NOTEWORTHY Takeda G protein-coupled receptor 5 (TGR5) agonists have potential as a treatment for nonalcoholic steatohepatitis and nonalcoholic fatty liver disease (NAFLD) but have until now been associated with undesirable side effects associated with systemic TGR5 agonism, including blockade of gallbladder emptying. We demonstrate that RDX8940, a potent, selective, minimally systemic oral TGR5 agonist, improves liver steatosis and insulin sensitivity in a mouse model of NAFLD and does not inhibit gallbladder emptying in mice.
Collapse
Affiliation(s)
| | | | | | | | - Sindy Wan
- Ardelyx, Incorporated, Fremont, California
| | | | | | - Tao Chen
- Ardelyx, Incorporated, Fremont, California
| | - Noah Bell
- Ardelyx, Incorporated, Fremont, California
| | | | | | | | | | | | | | | | - Karen Shaw
- Ardelyx, Incorporated, Fremont, California
| | - Cathy Le
- Ardelyx, Incorporated, Fremont, California
| | | | - I-Hsin Hsu
- Ardelyx, Incorporated, Fremont, California
| | | | | | | | | |
Collapse
|
15
|
Lee Y, Chakraborty S, Meininger CJ, Muthuchamy M. Insulin resistance disrupts cell integrity, mitochondrial function, and inflammatory signaling in lymphatic endothelium. Microcirculation 2018; 25:e12492. [PMID: 30025187 DOI: 10.1111/micc.12492] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Lymphatic vessel dysfunction and increased lymph leakage have been directly associated with several metabolic diseases. However, the underlying cellular mechanisms causing lymphatic dysfunction have not been determined. Aberrant insulin signaling affects the metabolic function of cells and consequently impairs tissue function. We hypothesized that insulin resistance in LECs decreases eNOS activity, disrupts barrier integrity increases permeability, and activates mitochondrial dysfunction and pro-inflammatory signaling pathways. METHODS LECs were treated with insulin and/or glucose to determine the mechanisms leading to insulin resistance. RESULTS Acute insulin treatment increased eNOS phosphorylation and NO production in LECs via activation of the PI3K/Akt signaling pathway. Prolonged hyperglycemia and hyperinsulinemia induced insulin resistance in LECs. Insulin-resistant LECs produced less NO due to a decrease in eNOS phosphorylation and showed a significant decrease in impedance across an LEC monolayer that was associated with disruption in the adherence junctional proteins. Additionally, insulin resistance in LECs impaired mitochondrial function by decreasing basal-, maximal-, and ATP-linked OCRs and activated NF-κB nuclear translocation coupled with increased pro-inflammatory signaling. CONCLUSION Our data provide the first evidence that insulin resistance disrupts endothelial barrier integrity, decreases eNOS phosphorylation and mitochondrial function, and activates inflammation in LECs.
Collapse
Affiliation(s)
- Yang Lee
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Cynthia J Meininger
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Mariappan Muthuchamy
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| |
Collapse
|
16
|
Jejelava N, Kaufman S, Krieger JP, Terra MM, Langhans W, Arnold M. Intestinal lymph as a readout of meal-induced GLP-1 release in an unrestrained rat model. Am J Physiol Regul Integr Comp Physiol 2018; 314:R724-R733. [PMID: 29341824 DOI: 10.1152/ajpregu.00120.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Intestinal lymph supposedly provides a readout for the secretion of intestinal peptides. We here assessed how mesenteric lymph duct (MLD) lymph levels of glucagon-like peptide (GLP-1), insulin, and metabolites [glucose and triglycerides (TG)] evolve after isocaloric high- and low-fat diet (HFD and LFD) meals and how they compare with hepatic portal vein (HPV) plasma levels. Moreover, we examined the effects of intraperitoneally administered GLP-1 (1 or 10 nmol/kg) on these parameters. At 20 min after the HFD meal onset, GLP-1 levels were higher in MLD lymph than in HPV plasma. No such difference occurred with the LFD meal. Intraperitoneal injections of 10 nmol/kg GLP-1 before meals enhanced the meal-induced increases in MLD lymph and HPV plasma GLP-1 levels except for the MLD lymph levels after the HFD meal. Intraperitoneal injection of 1 nmol/kg GLP-1 only increased HPV plasma GLP-1 levels at 60 min after the HFD meal. GLP-1 injections did not increase the MLD lymph or HPV plasma GLP-1 concentrations beyond the physiological range, suggesting that intraperitoneal GLP-1 injections can recapitulate the short-term effects of endogenous GLP-1. Dipeptidyl peptidase IV (DPP-IV) activity in MLD lymph was lower than in HPV plasma, which presumably contributed to the higher levels of GLP-1 in lymph than in plasma. Insulin and glucose showed similar profiles in MLD lymph and HPV plasma, whereas TG levels were higher in lymph than in plasma. These results indicate that intestinal lymph provides a sensitive readout of intestinal peptide release and potential action, in particular when fat-rich diets are consumed.
Collapse
Affiliation(s)
- Nino Jejelava
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology, ETH Zurich, Switzerland
| | - Sharon Kaufman
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology, ETH Zurich, Switzerland
| | - Jean-Philippe Krieger
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology, ETH Zurich, Switzerland
| | | | - Wolfgang Langhans
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology, ETH Zurich, Switzerland
| | - Myrtha Arnold
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology, ETH Zurich, Switzerland
| |
Collapse
|
17
|
Lach G, Schellekens H, Dinan TG, Cryan JF. Anxiety, Depression, and the Microbiome: A Role for Gut Peptides. Neurotherapeutics 2018; 15:36-59. [PMID: 29134359 PMCID: PMC5794698 DOI: 10.1007/s13311-017-0585-0] [Citation(s) in RCA: 353] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The complex bidirectional communication between the gut and the brain is finely orchestrated by different systems, including the endocrine, immune, autonomic, and enteric nervous systems. Moreover, increasing evidence supports the role of the microbiome and microbiota-derived molecules in regulating such interactions; however, the mechanisms underpinning such effects are only beginning to be resolved. Microbiota-gut peptide interactions are poised to be of great significance in the regulation of gut-brain signaling. Given the emerging role of the gut-brain axis in a variety of brain disorders, such as anxiety and depression, it is important to understand the contribution of bidirectional interactions between peptide hormones released from the gut and intestinal bacteria in the context of this axis. Indeed, the gastrointestinal tract is the largest endocrine organ in mammals, secreting dozens of different signaling molecules, including peptides. Gut peptides in the systemic circulation can bind cognate receptors on immune cells and vagus nerve terminals thereby enabling indirect gut-brain communication. Gut peptide concentrations are not only modulated by enteric microbiota signals, but also vary according to the composition of the intestinal microbiota. In this review, we will discuss the gut microbiota as a regulator of anxiety and depression, and explore the role of gut-derived peptides as signaling molecules in microbiome-gut-brain communication. Here, we summarize the potential interactions of the microbiota with gut hormones and endocrine peptides, including neuropeptide Y, peptide YY, pancreatic polypeptide, cholecystokinin, glucagon-like peptide, corticotropin-releasing factor, oxytocin, and ghrelin in microbiome-to-brain signaling. Together, gut peptides are important regulators of microbiota-gut-brain signaling in health and stress-related psychiatric illnesses.
Collapse
Affiliation(s)
- Gilliard Lach
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Harriet Schellekens
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- Food for Health Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland.
- Food for Health Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW We report recently published knowledge regarding gut chemosensory mechanisms focusing on nutrient-sensing G protein-coupled receptors (GPCRs) expressed on gut enteroendocrine cells (EECs), tuft cells, and in afferent nerves in the gastroduodenal mucosa and submucosa. RECENT FINDINGS Gene profiling of EECs and tuft cells have revealed expression of a variety of nutrient-sensing GPCRs. The density of EEC and tuft cells is altered by luminal environmental changes that may occur following bypass surgery or in the presence of mucosal inflammation. Some EECs and tuft cells are directly linked to sensory nerves in the subepithelial space. Vagal afferent neurons that innervate the intestinal villi express nutrient receptors, contributing to the regulation of duodenal anion secretion in response to luminal nutrients. Nutrients are also absorbed via specific epithelial transporters. SUMMARY Gastric and duodenal epithelial cells are continually exposed to submolar concentrations of nutrients that activate GPCRs expressed on EECs, tuft cells, and submucosal afferent nerves and are also absorbed through specific transporters, regulating epithelial cell proliferation, gastrointestinal physiological function, and metabolism. The chemical coding and distribution of EECs and tuft cells are keys to the development of GPCR-targeted therapies.
Collapse
|
19
|
Abstract
Intestinal glucagon-like peptide-1 (GLP-1) and pancreatic insulin, released postprandially, commonly regulate glucose metabolism. Recent clinical experience indicates that the GLP-1R agonist and insulin in combination, compared to insulin alone, results in better glycemic and weight controls in type 2 diabetic patients. These observations suggest possible interactive effect of these hormones. These hormones, in addition to peripherally controlling glycemia, exert central regulation of food intake and glucose metabolism, the effect at least partly mediated by signaling to the brain via the vagal afferents. However, whether the vagal afferents are involved in the interactive effects of GLP-1 and insulin remains unknown. The present study explored possible cooperative effect of GLP-1 and insulin on vagal afferent neurons isolated from nodose ganglion (NG) of mice, while monitoring the neuronal activity by measuring cytosolic Ca2+ concentration ([Ca2+]i) with fura-2. GLP-1 at 10-8M increased [Ca2+]i in 8-11% of single NG neurons. GLP-1-induced [Ca2+]i increases were inhibited by GLP-1 receptor antagonist exendin (9-39). Majority (92%) of GLP-1-responseive NG neurons also responded to 10-7M insulin with [Ca2+]i increases. Both GLP-1 and insulin at lower concentration of 10-9M induced [Ca2+]i increases with smaller amplitude in lesser NG neuron population (4-7%). These hormones at 10-9M in combination recruited the unresponsive neurons to [Ca2+]i increases, and induced [Ca2+]i increases with greater amplitude in the responsive neurons. The results demonstrate that GLP-1 and insulin synergistically and additively activate vagal afferent neurons. This interaction may be linked to the postprandial functions mediated commonly by GLP-1 and insulin and in the beneficial outcome of the therapy with GLP-1 receptor agonist and insulin in combination.
Collapse
Affiliation(s)
- Yusaku Iwasaki
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 320-0498, Japan
| | - Chayon Goswami
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 320-0498, Japan
| | - Toshihiko Yada
- Division of Integrative Physiology, Department of Physiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 320-0498, Japan.
| |
Collapse
|
20
|
Bernier-Latmani J, Petrova TV. Intestinal lymphatic vasculature: structure, mechanisms and functions. Nat Rev Gastroenterol Hepatol 2017; 14:510-526. [PMID: 28655884 DOI: 10.1038/nrgastro.2017.79] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The mammalian intestine is richly supplied with lymphatic vasculature, which has functions ranging from maintenance of interstitial fluid balance to transport of antigens, antigen-presenting cells, dietary lipids and fat-soluble vitamins. In this Review, we provide in-depth information concerning the organization and structure of intestinal lymphatics, the current view of their developmental origins, as well as molecular mechanisms of intestinal lymphatic patterning and maintenance. We will also discuss physiological aspects of intestinal lymph flow regulation and the known and emerging roles of intestinal lymphatic vessels in human diseases, such as IBD, infection and cancer.
Collapse
Affiliation(s)
- Jeremiah Bernier-Latmani
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Fundamental Oncology, Ludwig Institute for Cancer Research and Institute of Pathology, Centre Hospitalier Universitaire Vaudois and University of Lausanne (UNIL), Chemin des Boveresses 155, Epalinges, Switzerland.,Swiss Institute for Experimental Cancer Research, School of Life Sciences, Swiss Federal Institute of Technology Lausanne, Route Cantonale 1015, Lausanne, Switzerland
| |
Collapse
|
21
|
Steinert RE, Feinle-Bisset C, Asarian L, Horowitz M, Beglinger C, Geary N. Ghrelin, CCK, GLP-1, and PYY(3-36): Secretory Controls and Physiological Roles in Eating and Glycemia in Health, Obesity, and After RYGB. Physiol Rev 2017; 97:411-463. [PMID: 28003328 PMCID: PMC6151490 DOI: 10.1152/physrev.00031.2014] [Citation(s) in RCA: 402] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The efficacy of Roux-en-Y gastric-bypass (RYGB) and other bariatric surgeries in the management of obesity and type 2 diabetes mellitus and novel developments in gastrointestinal (GI) endocrinology have renewed interest in the roles of GI hormones in the control of eating, meal-related glycemia, and obesity. Here we review the nutrient-sensing mechanisms that control the secretion of four of these hormones, ghrelin, cholecystokinin (CCK), glucagon-like peptide-1 (GLP-1), and peptide tyrosine tyrosine [PYY(3-36)], and their contributions to the controls of GI motor function, food intake, and meal-related increases in glycemia in healthy-weight and obese persons, as well as in RYGB patients. Their physiological roles as classical endocrine and as locally acting signals are discussed. Gastric emptying, the detection of specific digestive products by small intestinal enteroendocrine cells, and synergistic interactions among different GI loci all contribute to the secretion of ghrelin, CCK, GLP-1, and PYY(3-36). While CCK has been fully established as an endogenous endocrine control of eating in healthy-weight persons, the roles of all four hormones in eating in obese persons and following RYGB are uncertain. Similarly, only GLP-1 clearly contributes to the endocrine control of meal-related glycemia. It is likely that local signaling is involved in these hormones' actions, but methods to determine the physiological status of local signaling effects are lacking. Further research and fresh approaches are required to better understand ghrelin, CCK, GLP-1, and PYY(3-36) physiology; their roles in obesity and bariatric surgery; and their therapeutic potentials.
Collapse
Affiliation(s)
- Robert E Steinert
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christine Feinle-Bisset
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Lori Asarian
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Michael Horowitz
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Christoph Beglinger
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| | - Nori Geary
- University of Adelaide Discipline of Medicine and National Health and Medical Research Council of Australia Centre of Research Excellence in Translating Nutritional Science to Good Health, Adelaide, Australia; DSM Nutritional Products, R&D Human Nutrition and Health, Basel, Switzerland; Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland; Department of Biomedicine and Division of Gastroenterology, University Hospital Basel, Basel, Switzerland; and Department of Psychiatry, Weill Medical College of Cornell University, New York, New York
| |
Collapse
|
22
|
Sato H, Zhang LS, Martinez K, Chang EB, Yang Q, Wang F, Howles PN, Hokari R, Miura S, Tso P. Antibiotics Suppress Activation of Intestinal Mucosal Mast Cells and Reduce Dietary Lipid Absorption in Sprague-Dawley Rats. Gastroenterology 2016; 151:923-932. [PMID: 27436071 PMCID: PMC5391873 DOI: 10.1053/j.gastro.2016.07.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 06/28/2016] [Accepted: 07/08/2016] [Indexed: 01/04/2023]
Abstract
BACKGROUND & AIMS The gut microbiota affects intestinal permeability and mucosal mast cells (MMCs) responses. Activation of MMCs has been associated with absorption of dietary fat. We investigated whether the gut microbiota contributes to the fat-induced activation of MMCs in rats, and how antibiotics might affect this process. METHODS Adult male Sprague-Dawley rats were given streptomycin and penicillin for 4 days (n = 6-8) to reduce the abundance of their gut flora, or normal drinking water (controls, n = 6-8). They underwent lymph fistula surgery and after an overnight recovery were given an intraduodenal bolus of intralipid. We collected intestinal tissues and lymph fluid and assessed activation of MMCs, intestinal permeability, and fat transport parameters. RESULTS Compared with controls, intestinal lymph from rats given antibiotics had reduced levels of mucosal mast cell protease II (produced by MMCs) and decreased activity of diamine oxidase (produced by enterocytes) (P < .05). Rats given antibiotics had reduced intestinal permeability in response to dietary lipid compared with controls (P < .01). Unexpectedly, antibiotics also reduced lymphatic transport of triacylglycerol and phospholipid (P < .01), concomitant with decreased levels of mucosal apolipoproteins B, A-I, and A-IV (P < .01). No differences were found in intestinal motility or luminal pancreatic lipase activity between rats given antibiotics and controls. These effects were not seen with an acute dose of antibiotics or 4 weeks after the antibiotic regimen ended. CONCLUSIONS The intestinal microbiota appears to activate MMCs after the ingestion of fat in rats; this contributes to fat-induced intestinal permeability. We found that the gut microbiome promotes absorption of lipid, probably by intestinal production of apolipoproteins and secretion of chylomicrons.
Collapse
Affiliation(s)
- Hirokazu Sato
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio; Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Linda S Zhang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Kristina Martinez
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Eugene B Chang
- Department of Medicine, Section of Gastroenterology, Hepatology, and Nutrition, University of Chicago, Chicago, Illinois
| | - Qing Yang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Fei Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Philip N Howles
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Ryota Hokari
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Soichiro Miura
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
23
|
Bauer PV, Hamr SC, Duca FA. Regulation of energy balance by a gut-brain axis and involvement of the gut microbiota. Cell Mol Life Sci 2016; 73:737-55. [PMID: 26542800 PMCID: PMC11108299 DOI: 10.1007/s00018-015-2083-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 10/22/2015] [Accepted: 10/26/2015] [Indexed: 12/11/2022]
Abstract
Despite significant progress in understanding the homeostatic regulation of energy balance, successful therapeutic options for curbing obesity remain elusive. One potential target for the treatment of obesity is via manipulation of the gut-brain axis, a complex bidirectional communication system that is crucial in maintaining energy homeostasis. Indeed, ingested nutrients induce secretion of gut peptides that act either via paracrine signaling through vagal and non-vagal neuronal relays, or in an endocrine fashion via entry into circulation, to ultimately signal to the central nervous system where appropriate responses are generated. We review here the current hypotheses of nutrient sensing mechanisms of enteroendocrine cells, including the release of gut peptides, mainly cholecystokinin, glucagon-like peptide-1, and peptide YY, and subsequent gut-to-brain signaling pathways promoting a reduction of food intake and an increase in energy expenditure. Furthermore, this review highlights recent research suggesting this energy regulating gut-brain axis can be influenced by gut microbiota, potentially contributing to the development of obesity.
Collapse
Affiliation(s)
- Paige V Bauer
- Department of Medicine, Toronto General Research Institute, UHN, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Sophie C Hamr
- Department of Medicine, Toronto General Research Institute, UHN, Toronto, ON, M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Frank A Duca
- Department of Medicine, Toronto General Research Institute, UHN, Toronto, ON, M5G 1L7, Canada.
- MaRS Centre, Toronto Medical Discovery Tower, Room 10-701H, 101 College Street, Toronto, ON, M5G 1L7, Canada.
| |
Collapse
|
24
|
Amouyal C, Andreelli F. Increasing GLP-1 Circulating Levels by Bariatric Surgery or by GLP-1 Receptor Agonists Therapy: Why Are the Clinical Consequences so Different? J Diabetes Res 2016; 2016:5908656. [PMID: 27382574 PMCID: PMC4921140 DOI: 10.1155/2016/5908656] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/18/2016] [Indexed: 12/13/2022] Open
Abstract
The "incretin effect" is used to describe the observation that more insulin is secreted after the oral administration of glucose compared to that after the intravenous administration of the same amount of glucose. During the absorption of meals, the gut is thought to regulate insulin secretion by secreting a specific factor that targets pancreatic beta cells. Additional research confirmed this hypothesis with the discovery of two hormones called incretins: gastric inhibitory peptide (GIP) and glucagon-like peptide 1 (GLP-1). During meals, specific cells in the gut (L and K enteroendocrine cells) secrete incretins, causing an increase in the blood concentrations of, respectively, GLP-1 and GIP. Bariatric surgery is now proposed during the therapeutic management of type 2 diabetes in obese or overweight populations. It has been hypothesized that restoration of endogenous GLP-1 secretion after the surgery may contribute to the postsurgical resolution of diabetes. In 2005, the commercialization of GLP-1 receptor agonists gave the possibility to test this hypothesis. A few years later, it is now accepted that GLP-1 receptor agonists and bariatric surgery differently improve type 2 diabetes. These differences between endogenous and exogenous GLP-1 on glucose homeostasis emphasized the dual properties of GLP-1 as a peptide hormone and as a neurotransmitter.
Collapse
Affiliation(s)
- Chloé Amouyal
- Cardiometabolism and Nutrition Institute (ICAN), Heart and Metabolism Department, Pitié-Salpêtrière Hospital (APHP), 75013 Paris, France
- INSERM UMRS U1166 (Eq 6) Nutriomics, UPMC, Pierre et Marie Curie Faculty Paris 6, Sorbonne University, 75013 Paris, France
| | - Fabrizio Andreelli
- Cardiometabolism and Nutrition Institute (ICAN), Heart and Metabolism Department, Pitié-Salpêtrière Hospital (APHP), 75013 Paris, France
- INSERM UMRS U1166 (Eq 6) Nutriomics, UPMC, Pierre et Marie Curie Faculty Paris 6, Sorbonne University, 75013 Paris, France
- *Fabrizio Andreelli:
| |
Collapse
|
25
|
Wang F, Yoder SM, Yang Q, Kohan AB, Kindel TL, Wang J, Tso P. Chronic high-fat feeding increases GIP and GLP-1 secretion without altering body weight. Am J Physiol Gastrointest Liver Physiol 2015; 309:G807-15. [PMID: 26336929 PMCID: PMC4652142 DOI: 10.1152/ajpgi.00351.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 08/20/2015] [Indexed: 01/31/2023]
Abstract
The incretin hormones, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), enhance postprandial insulin secretion, promote adipogenesis, and regulate gastrointestinal motility and food intake. To date, a consensus on how the incretin response is altered in obesity is lacking. We investigated the effects of chronic high-fat (HF) feeding on incretin secretion in the lymph fistula rat model. Male Sprague-Dawley rats (8 wk) were provided a semipurified AIN93M HF or low-fat (LF) diet ad libitum for 3 or 13 wk; a HF pair-fed (HF-PF) group was included as a control during the 3-wk feeding trial. Energy intake, body weight, and body composition were regularly monitored. At the culmination of the feeding period, an intestinal lymphatic duct cannula and duodenal infusion tube were installed. All animals were challenged with a 3-ml Ensure bolus (3.125 kcal/animal) to measure lymphatic incretin secretion. Despite a significantly higher energy intake, both the 3-wk and 13-wk HF-fed animals did not have an increase in body weight and only a slight increase in body fat compared with LF-fed rats. Following the duodenal Ensure challenge, the 3-wk and 13-wk HF-fed rats had significantly greater lymphatic GIP and GLP-1 secretion than the LF-fed animals. Additionally, the HF-PF group displayed a secretion profile similar to the HF-fed animals for GIP but a similar pattern to the LF-fed animals for GLP-1. The HF-PF data suggest that the increased GIP secretion is driven by the greater percentage of fat intake, whereas the increased GLP-1 secretion is driven by the excess caloric intake.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Patrick Tso
- Department of Pathology and Laboratory, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
26
|
Wang F, Yang Q, Huesman S, Xu M, Li X, Lou D, Woods SC, Marziano C, Tso P. The role of apolipoprotein A-IV in regulating glucagon-like peptide-1 secretion. Am J Physiol Gastrointest Liver Physiol 2015; 309:G680-7. [PMID: 26294669 PMCID: PMC4609932 DOI: 10.1152/ajpgi.00075.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 08/14/2015] [Indexed: 01/31/2023]
Abstract
Both glucagon-like peptide-1 (GLP-1) and apolipoprotein A-IV (apoA-IV) are produced from the gut and enhance postprandial insulin secretion. This study investigated whether apoA-IV regulates nutrient-induced GLP-1 secretion and whether apoA-IV knockout causes compensatory GLP-1 release. Using lymph-fistula-mice, we first determined lymphatic GLP-1 secretion by administering apoA-IV before an intraduodenal Ensure infusion. apoA-IV changed neither basal nor Ensure-induced GLP-1 secretion relative to saline administration. We then assessed GLP-1 in apoA-IV-/- and wild-type (WT) mice administered intraduodenal Ensure. apoA-IV-/- mice had comparable lymph flow, lymphatic triglyceride, glucose, and protein outputs as WT mice. Intriguingly, apoA-IV-/- mice had higher lymphatic GLP-1 concentration and output than WT mice 30 min after Ensure administration. Increased GLP-1 was also observed in plasma of apoA-IV-/- mice at 30 min. apoA-IV-/- mice had comparable total gut GLP-1 content relative to WT mice under fasting, but a lower GLP-1 content 30 min after Ensure administration, suggesting that more GLP-1 was secreted. Moreover, an injection of apoA-IV protein did not reverse the increased GLP-1 secretion in apoA-IV-/- mice. Finally, we assessed gene expression of GLUT-2 and the lipid receptors, including G protein-coupled receptor (GPR) 40, GPR119, and GPR120 in intestinal segments. GLUT-2, GPR40 and GPR120 mRNAs were unaltered by apoA-IV knockout. However, ileal GPR119 mRNA was significantly increased in apoA-IV-/- mice. GPR119 colocalizes with GLP-1 in ileum and stimulates GLP-1 secretion by sensing OEA, lysophosphatidylcholine, and 2-monoacylglycerols. We suggest that increased ileal GPR119 is a potential mechanism by which GLP-1 secretion is enhanced in apoA-IV-/- mice.
Collapse
Affiliation(s)
- Fei Wang
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Qing Yang
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Sarah Huesman
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Min Xu
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Xiaoming Li
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Danwen Lou
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Stephen C. Woods
- 2Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, Ohio
| | - Corina Marziano
- 1Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio; and
| |
Collapse
|
27
|
Hansen M, Hjøllund KR, Hartmann B, Plamboeck A, Deacon CF, Wewer Albrechtsen NJ, Holst JJ. Important species differences regarding lymph contribution to gut hormone responses. Peptides 2015; 71:28-31. [PMID: 26048091 DOI: 10.1016/j.peptides.2015.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/21/2015] [Accepted: 05/25/2015] [Indexed: 02/06/2023]
Abstract
INTRODUCTION GLP-1 is secreted from the gut upon nutrient intake and stimulates insulin secretion. The lymph draining the intestine may transport high levels of GLP-1 to the systemic circulation before it is metabolized by DPP-4. The aims of this study were to investigate to what extent the lymphatic system might contribute to the final level(s) of systemic circulating intact GLP-1 and, in addition, whether secretory profiles in intestinal lymph might reflect lamina propria levels of GLP-1 i.e. before capillary uptake and degradation by endothelial dipeptidyl peptidase-4 (DPP-4). METHOD 7 pigs of the YDL-strain were catheterized in the portal vein, carotid artery and cisterna chyli (lymph). Neuromedin C (NC) was infused through an ear vein catheter, before and after injection of a selective DPP-4 inhibitor (vildagliptin). Total and intact GLP-1 levels were measured throughout the 150min experiments using specific sandwich ELISAs. DPP-4 activity was measured spectrophotometrically. RESULTS Concentrations of both total and intact GLP-1 were markedly lower in lymph compared to plasma samples, and did not increase significantly in response to stimulation with NC in the absence/presence of vildagliptin. In contrast, total and intact GLP-1 levels increased significantly in the portal vein and carotid artery. DPP-4 activity was lower in lymph than plasma, and was reduced further by vildagliptin. CONCLUSION Our observations indicate that the lymphatic system does not transport high levels of intact GLP-1 to the systemic circulation, and that GLP-1 levels in cisternal lymph do not reflect the hormone levels in the intestinal lamina propria.
Collapse
Affiliation(s)
- Marie Hansen
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Section for Translational Metabolism, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Karina R Hjøllund
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Section for Translational Metabolism, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Bolette Hartmann
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Section for Translational Metabolism, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Astrid Plamboeck
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Section for Translational Metabolism, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Carolyn F Deacon
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Section for Translational Metabolism, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Nicolai J Wewer Albrechtsen
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Section for Translational Metabolism, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Jens J Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Section for Translational Metabolism, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
28
|
Intestinal GLP-1 and satiation: from man to rodents and back. Int J Obes (Lond) 2015; 40:198-205. [PMID: 26315842 DOI: 10.1038/ijo.2015.172] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 08/27/2015] [Accepted: 08/12/2015] [Indexed: 02/06/2023]
Abstract
In response to luminal food stimuli during meals, enteroendocrine cells release gastrointestinal (GI) peptides that have long been known to control secretory and motor functions of the gut, pancreas and liver. Glucagon-like peptide-1 (GLP-1) has emerged as one of the most important GI peptides because of a combination of functions not previously ascribed to any other molecule. GLP-1 potentiates glucose-induced insulin secretion, suppresses glucagon release, slows gastric emptying and may serve as a satiation signal, although the physiological status of the latter function has not been fully established yet. Here we review the available evidence for intestinal GLP-1 to fulfill a number of established empirical criteria for assessing whether a hormone inhibits eating by eliciting physiological satiation in man and rodents.
Collapse
|
29
|
Sandoval DA, D'Alessio DA. Physiology of proglucagon peptides: role of glucagon and GLP-1 in health and disease. Physiol Rev 2015; 95:513-48. [PMID: 25834231 DOI: 10.1152/physrev.00013.2014] [Citation(s) in RCA: 342] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preproglucagon gene (Gcg) is expressed by specific enteroendocrine cells (L-cells) of the intestinal mucosa, pancreatic islet α-cells, and a discrete set of neurons within the nucleus of the solitary tract. Gcg encodes multiple peptides including glucagon, glucagon-like peptide-1, glucagon-like peptide-2, oxyntomodulin, and glicentin. Of these, glucagon and GLP-1 have received the most attention because of important roles in glucose metabolism, involvement in diabetes and other disorders, and application to therapeutics. The generally accepted model is that GLP-1 improves glucose homeostasis indirectly via stimulation of nutrient-induced insulin release and by reducing glucagon secretion. Yet the body of literature surrounding GLP-1 physiology reveals an incompletely understood and complex system that includes peripheral and central GLP-1 actions to regulate energy and glucose homeostasis. On the other hand, glucagon is established principally as a counterregulatory hormone, increasing in response to physiological challenges that threaten adequate blood glucose levels and driving glucose production to restore euglycemia. However, there also exists a potential role for glucagon in regulating energy expenditure that has recently been suggested in pharmacological studies. It is also becoming apparent that there is cross-talk between the proglucagon derived-peptides, e.g., GLP-1 inhibits glucagon secretion, and some additive or synergistic pharmacological interaction between GLP-1 and glucagon, e.g., dual glucagon/GLP-1 agonists cause more weight loss than single agonists. In this review, we discuss the physiological functions of both glucagon and GLP-1 by comparing and contrasting how these peptides function, variably in concert and opposition, to regulate glucose and energy homeostasis.
Collapse
Affiliation(s)
- Darleen A Sandoval
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David A D'Alessio
- Division of Endocrinology and Metabolism, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
30
|
Ronveaux CC, Tomé D, Raybould HE. Glucagon-like peptide 1 interacts with ghrelin and leptin to regulate glucose metabolism and food intake through vagal afferent neuron signaling. J Nutr 2015; 145:672-80. [PMID: 25833771 PMCID: PMC4381768 DOI: 10.3945/jn.114.206029] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 01/16/2015] [Indexed: 12/25/2022] Open
Abstract
Emerging evidence has suggested a possible physiologic role for peripheral glucagon-like peptide 1 (GLP-1) in regulating glucose metabolism and food intake. The likely site of action of GLP-1 is on vagal afferent neurons (VANs). The vagal afferent pathway is the major neural pathway by which information about ingested nutrients reaches the central nervous system and influences feeding behavior. Peripheral GLP-1 acts on VANs to inhibit food intake. The mechanism of the GLP-1 receptor (GLP-1R) is unlike other gut-derived receptors; GLP-1Rs change their cellular localization according to feeding status rather than their protein concentrations. It is possible that several gut peptides are involved in mediating GLP-1R translocation. The mechanism of peripheral GLP-1R translocation still needs to be elucidated. We review data supporting the role of peripheral GLP-1 acting on VANs in influencing glucose homeostasis and feeding behavior. We highlight evidence demonstrating that GLP-1 interacts with ghrelin and leptin to induce satiation. Our aim was to understand the mechanism of peripheral GLP-1 in the development of noninvasive antiobesity treatments.
Collapse
Affiliation(s)
- Charlotte C Ronveaux
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA; and,Department of Nutrition and Physiology and Ingestive Behavior, AgroParisTech, Paris, France
| | - Daniel Tomé
- Department of Nutrition and Physiology and Ingestive Behavior, AgroParisTech, Paris, France
| | - Helen E Raybould
- Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California, Davis, Davis, CA; and
| |
Collapse
|
31
|
Punjabi M, Arnold M, Rüttimann E, Graber M, Geary N, Pacheco-López G, Langhans W. Circulating glucagon-like peptide-1 (GLP-1) inhibits eating in male rats by acting in the hindbrain and without inducing avoidance. Endocrinology 2014; 155:1690-9. [PMID: 24601880 DOI: 10.1210/en.2013-1447] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To address the neural mediation of the eating-inhibitory effect of circulating glucagon-like peptide-1 (GLP-1), we investigated the effects of 1) intra-fourth ventricular infusion of the GLP-1 receptor antagonist exendin-9 or 2) area postrema lesion on the eating-inhibitory effect of intrameal hepatic portal vein (HPV) GLP-1 infusion in adult male rats. To evaluate the physiological relevance of the observed effect we examined 3) the influence of GLP-1 on flavor acceptance in a 2-bottle conditioned flavor avoidance test, and 4) measured active GLP-1 in the HPV and vena cava (VC) in relation to a meal and in the VC after HPV GLP-1 infusion. Intrameal HPV GLP-1 infusion (1 nmol/kg body weight-5 min) specifically reduced ongoing meal size by almost 40% (P < .05). Intra-fourth ventricular exendin-9 (10 μg/rat) itself did not affect eating, but attenuated (P < .05) the satiating effect of HPV GLP-1. Area postrema lesion also blocked (P < .05) the eating-inhibitory effect of HPV GLP-1. Pairing consumption of flavored saccharin solutions with HPV GLP-1 infusion did not alter flavor acceptance, indicating that HPV GLP-1 can inhibit eating without inducing malaise. A regular chow meal transiently increased (P < .05) HPV, but not VC, plasma active GLP-1 levels, whereas HPV GLP-1 infusion caused a transient supraphysiological increase (P < .01) in VC GLP-1 concentration 3 minutes after infusion onset. The results implicate hindbrain GLP-1 receptors and the area postrema in the eating-inhibitory effect of circulating GLP-1, but question the physiological relevance of the eating-inhibitory effect of iv infused GLP-1 under our conditions.
Collapse
Affiliation(s)
- Mukesh Punjabi
- Physiology and Behavior Laboratory, Institute of Food, Nutrition, and Health, Swiss Federal Institute of Technology Zurich (ETH Zurich), 8603 Schwerzenbach, Switzerland
| | | | | | | | | | | | | |
Collapse
|
32
|
Poole DP, Lee M, Tso P, Bunnett NW, Yo SJ, Lieu T, Shiu A, Wang JC, Nomura DK, Aponte GW. Feeding-dependent activation of enteric cells and sensory neurons by lymphatic fluid: evidence for a neurolymphocrine system. Am J Physiol Gastrointest Liver Physiol 2014; 306:G686-98. [PMID: 24578341 PMCID: PMC3989702 DOI: 10.1152/ajpgi.00433.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lymphatic fluid is a plasma filtrate that can be viewed as having biological activity through the passive accumulation of molecules from the interstitial fluid. The possibility that lymphatic fluid is part of an active self-contained signaling process that parallels the endocrine system, through the activation of G-protein coupled receptors (GPCR), has remained unexplored. We show that the GPCR lysophosphatidic acid 5 (LPA5) is found in sensory nerve fibers expressing calcitonin gene-related peptide (CGRP) that innervate the lumen of lymphatic lacteals and enteric nerves. Using LPA5 as a model for nutrient-responsive GPCRs present on sensory nerves, we demonstrate that dietary protein hydrolysate (peptone) can induce c-Fos expression in enterocytes and nerves that express LPA5. Mesenteric lymphatic fluid (MLF) mobilizes intracellular calcium in cell models expressing LPA5 upon feeding in a time- and dose-dependent manner. Primary cultured neurons of the dorsal root ganglia expressing CGRP are activated by MLF, which is enhanced upon LPA5 overexpression. Activation is independent of the known LPA5 agonists, lysophosphatidic acid and farnesyl pyrophosphate. These data bring forth a pathway for the direct stimulation of sensory nerves by luminal contents and interstitial fluid. Thus, by activating LPA5 on sensory nerves, MLF provides a means for known and yet to be identified constituents of the interstitial fluid to act as signals to comprise a "neurolymphocrine" system.
Collapse
Affiliation(s)
- Daniel P. Poole
- 1Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia; ,2Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia;
| | - Mike Lee
- 4Department of Pathology, Stanford University, Palo Alto, California;
| | - Patrick Tso
- 6Department of Pathobiology and Molecular Medicine, University of Cincinnati, Reading, Ohio
| | - Nigel W. Bunnett
- 1Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia; ,3Department of Pharmacology, The University of Melbourne, Parkville, Victoria, Australia;
| | - Sek Jin Yo
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - TinaMarie Lieu
- 1Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia;
| | - Amy Shiu
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - Jen-Chywan Wang
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - Daniel K. Nomura
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| | - Gregory W. Aponte
- 5Department of Nutritional Sciences and Toxicology, University of California at Berkeley, Berkeley, California;
| |
Collapse
|
33
|
Ohlsson L, Kohan AB, Tso P, Ahrén B. GLP-1 released to the mesenteric lymph duct in mice: effects of glucose and fat. ACTA ACUST UNITED AC 2014; 189:40-5. [PMID: 24583245 DOI: 10.1016/j.regpep.2014.02.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 02/13/2014] [Accepted: 02/19/2014] [Indexed: 11/19/2022]
Abstract
Using a newly developed in vivo model measuring glucagon-like peptide-1 (GLP-1) in gut lymphatics in mice, we quantified GLP-1 secretion in vivo after glucose versus fat ingestion with and without concomitant DPP-4 inhibition. The mesenteric lymphatic duct was cannulated in anesthetized C57BL6/J mice and lymph was collected in 30 min intervals. Glucose or fat emulsion (Intralipid®) (0.03, 0.1 or 0.3 kcal) with or without DPP-4-inhibition (NVP DPP728; 10 μmol/kg) was administered by gastric gavage. Basal intact GLP-1 levels were 0.37±0.04 pmol/l (n=61) in lymph compared to 0.07±0.03 in plasma (n=6; P=0.04) and basal DPP-4 activity was 4.7±0.3 pmol/min/μl in lymph (n=23) compared to 22.3±0.9 pmol/min/μl in plasma (n=8; P<0.001). Lymph flow increased from 1.2±0.1 μl/min to 2.3±02μl/min at 30 min after glucose and fat administration, with no difference between type of challenge or dose (n=81). Lymph GLP-1 levels increased calorie-dependently after both glucose and fat but with different time courses in that glucose induced a transient increase which had returned to baseline after 90 min whereas the lipid induced a sustained increase which was still elevated above baseline after 210 min. Lymph GLP-1 appearance during 210 min was two to three-fold higher after glucose (7.4±2.3 fmol at 0.3 kcal) than after isocaloric fat (2.9±0.8 fmol at 0.3 kcal; P<0.001). The slope between caloric load and lymph GLP-1 appearance was, however, identical after glucose and fat. We conclude that lymph GLP-1 is higher than plasma GLP-1 whereas lymph DPP-4 activity is lower than plasma DPP-4 activity and that both glucose and fat clearly stimulate GLP-1 secretion calorie-dependently in vivo but with different time courses.
Collapse
Affiliation(s)
- Lena Ohlsson
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Alison B Kohan
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Patrick Tso
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Bo Ahrén
- Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
| |
Collapse
|
34
|
Young RL, Chia B, Isaacs NJ, Ma J, Khoo J, Wu T, Horowitz M, Rayner CK. Disordered control of intestinal sweet taste receptor expression and glucose absorption in type 2 diabetes. Diabetes 2013; 62:3532-3541. [PMID: 23761104 PMCID: PMC3781477 DOI: 10.2337/db13-0581] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Accepted: 06/05/2013] [Indexed: 02/05/2023]
Abstract
We previously established that the intestinal sweet taste receptors (STRs), T1R2 and T1R3, were expressed in distinct epithelial cells in the human proximal intestine and that their transcript levels varied with glycemic status in patients with type 2 diabetes. Here we determined whether STR expression was 1) acutely regulated by changes in luminal and systemic glucose levels, 2) disordered in type 2 diabetes, and 3) linked to glucose absorption. Fourteen healthy subjects and 13 patients with type 2 diabetes were studied twice, at euglycemia (5.2 ± 0.2 mmol/L) or hyperglycemia (12.3 ± 0.2 mmol/L). Endoscopic biopsy specimens were collected from the duodenum at baseline and after a 30-min intraduodenal glucose infusion of 30 g/150 mL water plus 3 g 3-O-methylglucose (3-OMG). STR transcripts were quantified by RT-PCR, and plasma was assayed for 3-OMG concentration. Intestinal STR transcript levels at baseline were unaffected by acute variations in glycemia in healthy subjects and in type 2 diabetic patients. T1R2 transcript levels increased after luminal glucose infusion in both groups during euglycemia (+5.8 × 10(4) and +5.8 × 10(4) copies, respectively) but decreased in healthy subjects during hyperglycemia (-1.4 × 10(4) copies). T1R2 levels increased significantly in type 2 diabetic patients under the same conditions (+6.9 × 10(5) copies). Plasma 3-OMG concentrations were significantly higher in type 2 diabetic patients than in healthy control subjects during acute hyperglycemia. Intestinal T1R2 expression is reciprocally regulated by luminal glucose in health according to glycemic status but is disordered in type 2 diabetes during acute hyperglycemia. This defect may enhance glucose absorption in type 2 diabetic patients and exacerbate postprandial hyperglycemia.
Collapse
Affiliation(s)
- Richard L. Young
- Nerve-Gut Research Laboratory, University of Adelaide, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Bridgette Chia
- Nerve-Gut Research Laboratory, University of Adelaide, Adelaide, South Australia, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Nicole J. Isaacs
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Jing Ma
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
- Department of Endocrinology and Metabolism, Shanghai Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Joan Khoo
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
- Department of Endocrinology, Changi General Hospital, Singapore
| | - Tongzhi Wu
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Michael Horowitz
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
| | - Christopher K. Rayner
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health, University of Adelaide, Adelaide, South Australia, Australia
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| |
Collapse
|
35
|
Sato S, Hokari R, Kurihara C, Sato H, Narimatsu K, Hozumi H, Ueda T, Higashiyama M, Okada Y, Watanabe C, Komoto S, Tomita K, Kawaguchi A, Nagao S, Miura S. Dietary lipids and sweeteners regulate glucagon-like peptide-2 secretion. Am J Physiol Gastrointest Liver Physiol 2013; 304:G708-14. [PMID: 23370677 DOI: 10.1152/ajpgi.00282.2012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glucagon-like peptide-2 (GLP-2) is a potent intestinal growth factor derived from enteroendocrine L cells. Although food intake is known to increase GLP-2 secretion, its regulatory mechanisms are largely unknown as a result of its very short half-life in venules. The aims of this study were to compare the effects of luminal nutrients on the stimulation of GLP-2 secretion in vivo using lymph samples and to clarify the involvement of the sweet taste receptor in this process in vitro. Lymph samples were collected from the thoracic duct after bolus administration of dietary lipids or sweetening agents into the duodenum of rats. Human enteroendocrine NCI-H716 cells were also used to compare the effects of various nutrients on GLP-2 secretion. GLP-2 concentrations were measured by ELISA in vivo and in vitro. GLP-2 secretion was enhanced by polyunsaturated fatty acid- and monounsaturated fatty acid-rich dietary oils, dietary carbohydrates, and some kinds of sweeteners in rats; this effect was reproduced in NCI-H716 cells using α-linolenic acid (αLA), glucose, and sweeteners. GLP-2 secretion induced by sweetening agents was inhibited by lactisole, a sweetness-antagonizing inhibitor of T1R3. In contrast, lactisole was unable to inhibit GLP-2 secretion induced by αLA alone. Our results suggested that fatty acid- and sweetener-induced GLP-2 secretion may be mediated by two different pathways, with the sweet taste receptor involved in the regulation of the latter.
Collapse
Affiliation(s)
- Shingo Sato
- Department of Internal Medicine, National Defense Medical College, Saitama, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Chandarana K, Gelegen C, Irvine EE, Choudhury AI, Amouyal C, Andreelli F, Withers DJ, Batterham RL. Peripheral activation of the Y2-receptor promotes secretion of GLP-1 and improves glucose tolerance. Mol Metab 2013; 2:142-52. [PMID: 24049729 DOI: 10.1016/j.molmet.2013.03.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/04/2013] [Accepted: 03/05/2013] [Indexed: 12/15/2022] Open
Abstract
The effect of peptide tyrosine-tyrosine (PYY) on feeding is well established but currently its role in glucose homeostasis is poorly defined. Here we show in mice, that intraperitoneal (ip) injection of PYY3-36 or Y2R agonist improves nutrient-stimulated glucose tolerance and enhances insulin secretion; an effect blocked by peripheral, but not central, Y2R antagonist administration. Studies on isolated mouse islets revealed no direct effect of PYY3-36 on insulin secretion. Bariatric surgery in mice, enterogastric anastomosis (EGA), improved glucose tolerance in wild-type mice and increased circulating PYY and active GLP-1. In contrast, in Pyy-null mice, post-operative glucose tolerance and active GLP-1 levels were similar in EGA and sham-operated groups. PYY3-36 ip increased hepato-portal active GLP-1 plasma levels, an effect blocked by ip Y2R antagonist. Collectively, these data suggest that PYY3-36 therefore acting via peripheral Y2R increases hepato-portal active GLP-1 plasma levels and improves nutrient-stimulated glucose tolerance.
Collapse
Key Words
- AUC, area under the curve
- CNS, central nervous system
- DPP-4, di-peptidyl peptidase-4
- EGA, entero-gastric anastomosis
- GLP-1
- Glucose homeostasis
- HFD, high-fat diet
- ICV, intracerebroventricular
- IPGTT, intraperitoneal glucose tolerance test
- PYY
- PYY, peptide tyrosine–tyrosine
- T2DM, type 2 diabetes mellitus
- WT, wild-type
- Y2-receptor
- Y2R, Y2-receptor
- aCSF, artificial cerebrospinal fluid
- active GLP-1, glucagon-like peptide-1(7-36)amide
- ip, intraperitoneal
Collapse
Affiliation(s)
- Keval Chandarana
- Centre for Obesity Research, Department of Medicine, University College London, Rayne Institute, 5 University Street, WC1E 6JJ, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Dailey MJ, Moran TH. Glucagon-like peptide 1 and appetite. Trends Endocrinol Metab 2013; 24:85-91. [PMID: 23332584 PMCID: PMC3594872 DOI: 10.1016/j.tem.2012.11.008] [Citation(s) in RCA: 134] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 11/19/2012] [Accepted: 11/23/2012] [Indexed: 01/05/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) and GLP-1 analogs have received much recent attention due to the success of GLP-1 mimetics in treating type II diabetes mellitus (T2DM), but these compounds may also have the potential to treat obesity. The satiety effect of GLP-1 may involve both within-meal enteroenteric reflexes, and across-meal central signaling mechanisms, that mediate changes in appetite and promote satiety. Here, we review data supporting the role of both peripheral and central GLP-1 signaling in the control of gastrointestinal motility and food intake. Understanding the mechanisms underlying the appetite-suppressive effects of GLP-1 may help in developing targeted treatments for obesity.
Collapse
Affiliation(s)
- Megan J Dailey
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | |
Collapse
|
38
|
Jessen L, Aulinger BA, Hassel JL, Roy KJ, Smith EP, Greer TM, Woods SC, Seeley RJ, D'Alessio DA. Suppression of food intake by glucagon-like peptide-1 receptor agonists: relative potencies and role of dipeptidyl peptidase-4. Endocrinology 2012; 153:5735-45. [PMID: 23033273 PMCID: PMC3512077 DOI: 10.1210/en.2012-1358] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Administration of the glucagon-like peptide-1 (GLP-1) receptor agonists GLP-1 and exendin-4 (Ex-4) directly into the central nervous system decreases food intake. But although Ex-4 potently suppresses food intake after peripheral administration, the effects of parenteral GLP-1 are variable and not as strong. A plausible explanation for these effects is the rapid inactivation of circulating GLP-1 by dipeptidyl peptidase-4 (DPP-4), an enzyme that does not alter Ex-4 activity. To test this hypothesis, we assessed the relative potency of Ex-4 and GLP-1 under conditions in which DPP-4 activity was reduced. Outbred rats, wild-type mice, and mice with a targeted deletion of DPP-4 (Dpp4(-/-)) were treated with GLP-1 alone or in combination with the DPP-4 inhibitor vildagliptin, Ex-4, or saline, and food intake was measured. GLP-1 alone, even at high doses, did not affect feeding in wild-type mice or rats but did reduce food intake when combined with vildagliptin or given to Dpp4(-/-) mice. Despite plasma clearance similar to DPP-4-protected GLP-1, equimolar Ex-4 caused greater anorexia than vildagliptin plus GLP-1. To determine whether supraphysiological levels of endogenous GLP-1 would suppress food intake if protected from DPP-4, rats with Roux-en-Y gastric bypass and significantly elevated postprandial plasma GLP-1 received vildagliptin or saline. Despite 5-fold greater postprandial GLP-1 in these animals, vildagliptin did not affect food intake in Roux-en-Y gastric bypass rats. Thus, in both mice and rats, peripheral GLP-1 reduces food intake significantly less than Ex-4, even when protected from DPP-4. These findings suggest distinct potencies of GLP-1 receptor agonists on food intake that cannot be explained by plasma pharmacokinetics.
Collapse
Affiliation(s)
- Lene Jessen
- Division of Endocrinology, University of Cincinnati, Cincinnati, OH 45237, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Thomson SC, Kashkouli A, Singh P. Glucagon-like peptide-1 receptor stimulation increases GFR and suppresses proximal reabsorption in the rat. Am J Physiol Renal Physiol 2012; 304:F137-44. [PMID: 23019232 DOI: 10.1152/ajprenal.00064.2012] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The incretin hormone glucagon-like peptide-1 (GLP-1) is released from the gut in response to fat or carbohydrate and contributes to negative feedback control of blood glucose by stimulating insulin secretion, inhibiting glucagon, and slowing gastric emptying. GLP-1 receptors (GLP-1R) are also expressed in the proximal tubule, and possibly elsewhere in the kidney. Presently, we examined the effect of a GLP-1R agonist on single-nephron glomerular filtration rate (GFR; SNGFR), proximal reabsorption (Jprox), tubuloglomerular feedback (TGF) responses, and urine flow rate in hydropenic male Wistar and Wistar-Froemter rats. Micropuncture and whole-kidney data were obtained before and during infusion of the GLP-1 agonist exenatide (1 nmol/h iv). SNGFR and Jprox were measured by late proximal collection at both extremes of TGF activation, which was achieved by perfusing Henle's loop at 0 or 50 nl/min. Primary changes in Jprox were revealed by analysis of covariance for Jprox with SNGFR as a covariate. Effects on TGF activation were determined in a separate set of experiments by comparing early distal and late proximal collections. Exenatide increased SNGFR by 33-50%, suppressed proximal tubular reabsorption by 20-40%, doubled early distal flow rate, and increased urine flow rate sixfold without altering the efficiency of glomerulotubular balance, TGF responsiveness, or the tonic influence of TGF. This implies that exenatide is both a proximal diuretic and a renal vasodilator. Since the natural agonist for the GLP-1R is regulated by intake of fat and carbohydrate, but not by salt or fluid, the control of salt excretion by the GLP-1R system departs from the usual negative-feedback paradigm for regulating salt balance.
Collapse
Affiliation(s)
- Scott C Thomson
- Department of Medicine, University of California and Veterans Affairs San Diego Healthcare System, San Diego, California 92161, USA.
| | | | | |
Collapse
|
40
|
Lu WJ, Yang Q, Yang L, Lee D, D'Alessio D, Tso P. Chylomicron formation and secretion is required for lipid-stimulated release of incretins GLP-1 and GIP. Lipids 2012; 47:571-80. [PMID: 22297815 DOI: 10.1007/s11745-011-3650-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2009] [Accepted: 12/15/2011] [Indexed: 11/25/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are incretins produced in the intestine that play a central role in glucose metabolism and insulin secretion. Circulating concentrations of GLP-1 and GIP are low and can be difficult to assay in rodents. These studies utilized the novel intestinal lymph fistula model we have established to investigate the mechanism of lipid-stimulated incretin secretion. Peak concentrations of GLP-1 and GIP following an enteral lipid stimulus (Liposyn) were significantly higher in intestinal lymph than portal venous plasma. To determine whether lipid-stimulated incretin secretion was related to chylomicron formation Pluronic L-81 (L-81), a surfactant inhibiting chylomicron synthesis, was given concurrently with Liposyn. The presence of L-81 almost completely abolished the increase in lymph triglyceride seen with Liposyn alone (P < 0.001). Inhibition of chylomicron formation with L-81 reduced GLP-1 secretion into lymph compared to Liposyn stimulation alone (P = 0.034). The effect of L-81 relative to Liposyn alone had an even greater effect on GIP secretion, which was completely abolished (P = 0.004). These findings of a dramatic effect of L-81 on lymph levels of GLP-1 and GIP support a strong link between intestinal lipid absorption and incretin secretion. The relative difference in the effect of L-81 on the two incretins provides further support that nutrient-stimulation of GIP and GLP-1 is via distinct mechanisms.
Collapse
Affiliation(s)
- Wendell J Lu
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, OH 45267, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Nagino K, Yokozawa J, Sasaki Y, Matsuda A, Takeda H, Kawata S. Increased secretion of insulin and proliferation of islet β-cells in rats with mesenteric lymph duct ligation. Biochem Biophys Res Commun 2012; 425:266-72. [PMID: 22842578 DOI: 10.1016/j.bbrc.2012.07.079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 07/17/2012] [Indexed: 11/18/2022]
Abstract
BACKGROUND & AIMS It has been suggested that intestinal lymph flow plays an important role in insulin secretion and glucose metabolism after meals. In this study, we investigated the influence of ligation of the mesenteric lymph duct on glucose metabolism and islet β-cells in rats. METHODS Male Sprague-Dawley rats (10 weeks old) were divided into two groups: one underwent ligation of the mesenteric lymph duct above the cistern (ligation group), and the other underwent a sham operation (sham group). After 1 and 2 weeks, fasting plasma concentrations of glucose, insulin, triglyceride, glucose-dependent insulinotropic polypeptide (GIP), and the active form of glucagon-like peptide-1 (GLP-1) were measured. At 2 weeks after the operation, the oral glucose tolerance test (OGTT) and intravenous glucose tolerance test (IVGTT) were performed. After the rats had been sacrificed, the insulin content of the pancreas was measured and the proliferation of β-cells was assessed immunohistochemically using antibodies against insulin and Ki-67. RESULTS During the OGTT, the ligation group showed a significant decrease in the plasma glucose concentration at 120 min (p<0.05) and a significant increase in the plasma insulin concentration by more than 2-fold at 15 min (p<0.01). On the other hand, the plasma GIP concentration was significantly decreased at 60 min (p<0.01) in the ligated group, while the active form of GLP-1 showed a significantly higher level at 90 min (1.7-fold; p<0.05) and 120 min (2.5-fold; p<0.01). During the IVGTT, the plasma insulin concentration in the ligation group was significantly higher at 2 min (more than 1.4-fold; p<0.05). Immunohistochemistry showed that the ratios of β-cell area/acinar cell area and β-cell area/islet area, and also β-cell proliferation, were significantly higher in the ligation group than in the sham group (p<0.05, p<0.01 and p<0.01, respectively). The insulin content per unit wet weight of pancreas was also significantly increased in the ligation group (p<0.05). CONCLUSIONS In rats with ligation of the mesenteric lymph duct, insulin secretion during the OGTT or IVGTT was higher, and the insulin content and β-cell proliferation in the pancreas were also increased. Our data show that mesenteric lymph duct flow has a role in glucose metabolism.
Collapse
Affiliation(s)
- Ko Nagino
- Department of Gastroenterology, Faculty of Medicine, Yamagata University, Yamagata 990-9585, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Ji Y, Sakata Y, Yang Q, Li X, Xu M, Yoder S, Langhans W, Tso P. Activation of rat intestinal mucosal mast cells by fat absorption. Am J Physiol Gastrointest Liver Physiol 2012; 302:G1292-300. [PMID: 22461027 PMCID: PMC3378165 DOI: 10.1152/ajpgi.00011.2012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Previous studies have linked certain types of gut mucosal immune cells with fat intake. We determined whether fat absorption activates intestinal mucosal mast cells (MMC), a key component of the gut mucosal immune system. Conscious intestinal lymph fistula rats were used. The mesenteric lymph ducts were cannulated, and the intraduodenal (i.d.) tubes were installed for the infusion of Liposyn II 20% (an intralipid emulsion). Lymphatic concentrations of histamine, rat MMC protease II (RMCPII), a specific marker of rat intestinal MMC degranulation, and prostaglandin D(2) (PGD(2)) were measured by ELISA. Intestinal MMC degranulation was visualized by immunofluorescent microscopy of jejunum sections taken at 1 h after Liposyn II gavage. Intraduodenal bolus infusion of Liposyn II 20% (4.4 kcal/3 ml) induced approximately a onefold increase in lymphatic histamine and PGD(2), ∼20-fold increase in lymphatic RMCPII, but only onefold increase in peripheral serum RMCPII concentrations. Release of RMCPII into lymph increased dose dependently with the amount of lipid fed. In addition, i.d. infusion of long-chain triacylglycerol trilinolein (C18:2 n-6, the major composite in Liposyn II) significantly increased the lymphatic RMCPII concentration, whereas medium-chain triacylglycerol tricaprylin (C8:0) did not alter lymph RMCPII secretion. Immunohistochemistry image revealed the degranulation of MMC into lamina propria after lipid feeding. These novel findings indicate that intestinal MMC are activated and degranulate to release MMC mediators to the circulation during fat absorption. This action of fatty acid is dose and chain length dependent.
Collapse
Affiliation(s)
- Yong Ji
- Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Ohio 45237, USA.
| | - Yasuhisa Sakata
- 1Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio;
| | - Qing Yang
- 1Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio;
| | - Xiaoming Li
- 1Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio;
| | - Min Xu
- 1Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio;
| | - Stephanie Yoder
- 1Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio;
| | - Wolfgang Langhans
- 2Institute of Animal Science, Swiss Federal Institute of Technology, Schwerzenbach, Switzerland
| | - Patrick Tso
- 1Department of Pathology and Laboratory Medicine, Metabolic Diseases Institute, University of Cincinnati, Cincinnati, Ohio;
| |
Collapse
|
43
|
Current and emerging concepts on the role of peripheral signals in the control of food intake and development of obesity. Br J Nutr 2012; 108:778-93. [PMID: 22409929 DOI: 10.1017/s0007114512000529] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The gastrointestinal peptides are classically known as short-term signals, primarily inducing satiation and/or satiety. However, accumulating evidence has broadened this view, and their role in long-term energy homeostasis and the development of obesity has been increasingly recognised. In the present review, the recent research involving the role of satiation signals, especially ghrelin, cholecystokinin, glucagon-like peptide 1 and peptide YY, in the development and treatment of obesity will be discussed. Their activity, interactions and release profile vary constantly with changes in dietary and energy influences, intestinal luminal environment, body weight and metabolic status. Manipulation of gut peptides and nutrient sensors in the oral and postoral compartments through diet and/or changes in gut microflora or using multi-hormone 'cocktail' therapy are among promising approaches aimed at reducing excess food consumption and body-weight gain.
Collapse
|
44
|
Cao Y, Gao W, Jusko WJ. Pharmacokinetic/pharmacodynamic modeling of GLP-1 in healthy rats. Pharm Res 2011; 29:1078-86. [PMID: 22179928 DOI: 10.1007/s11095-011-0652-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 12/06/2011] [Indexed: 10/14/2022]
Abstract
PURPOSE To provide a mechanism-based model to quantitatively describe GLP-1 pharmacokinetics (PK) and pharmacodynamics (PD) in rats. METHODS Intravenous (IV), infusion (IF), subcutaneous (SC), and intraperitoneal (IP) doses of GLP-1 were administered after glucose challenge in healthy Sprague-Dawley rats. Blood was analyzed for GLP-1, glucose, and insulin. The PK-PD modeling was performed with ADAPT 5. The concentration-response curve was generated and analyzed in comparison with other incretin-related therapeutics. RESULTS The PK of GLP-1 was described using a two-compartment model with a zero-order input accounting for endogenous GLP-1 synthesis. For SC and IP dosing, sequential zero-order and first-order absorption models reasonably described the rapid absorption process and flip-flop kinetics. In dynamics, GLP-1 showed insulinotropic effects (3-fold increase) after IV glucose challenge in a dose-dependent manner. The concentration-response curve was bell-shaped, which was captured using a biphasic two-binding site Adair model. Receptor binding of GLP-1 exhibited high capacity and low affinity kinetics for both binding sites (K(D) = 9.94 × 10(3) pM, K(2) = 1.56 × 10(-4) pM(-1)). CONCLUSIONS The PK of GLP-1 was linear and bi-exponential and its PD showed glucose-dependent insulinotropic effects. All profiles were captured by the present mechanistic model and the dynamic analysis yields several implications for incretin-related therapies.
Collapse
Affiliation(s)
- Yanguang Cao
- Department of Pharmaceutical Sciences School of Pharmacy & Pharmaceutical Sciences, State University of New York at Buffalo, 565 Hochstetter Hall, Buffalo, New York 14260, USA
| | | | | |
Collapse
|
45
|
Berthoud HR, Shin AC, Zheng H. Obesity surgery and gut-brain communication. Physiol Behav 2011; 105:106-19. [PMID: 21315095 PMCID: PMC3118403 DOI: 10.1016/j.physbeh.2011.01.023] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 12/15/2022]
Abstract
The prevalence of obesity, and the cluster of serious metabolic diseases it is associated with, continues to rise globally, and hopes for effective treatment with drugs have been considerably set back. Thus, success with bariatric surgeries to induce sustained body weight loss and effectively cure most of the associated co-morbidities appears almost "miraculous" and systematic investigation of the mechanisms at work has gained momentum. Here, we will discuss the basic organization of gut-brain communication and review clinical and pre-clinical investigations on the potential mechanisms by which gastric bypass surgery leads to its beneficial effects on energy balance and glucose homeostasis. Although a lot has been learned regarding changes in energy intake and expenditure, secretion of gut hormones, and improvement in glucose homeostasis, there has not yet been the "breakthrough observation" of identifying a key signaling component common to the beneficial effects of the surgery. However, given the complexity and redundancy of gut-brain signaling and gut signaling to other relevant organs, it is perhaps more realistic to expect a number of key signaling changes that act in concert to bring about the "miracle".
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, USA.
| | | | | |
Collapse
|
46
|
Kohan AB, Yoder SM, Tso P. Using the lymphatics to study nutrient absorption and the secretion of gastrointestinal hormones. Physiol Behav 2011; 105:82-8. [PMID: 21605578 PMCID: PMC3179774 DOI: 10.1016/j.physbeh.2011.04.056] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 04/25/2011] [Accepted: 04/27/2011] [Indexed: 12/31/2022]
Abstract
The lymph fistula rat model has traditionally been used to study the intestinal absorption of nutrients, especially lipids, but recently this model has also been used for studying the secretion of incretin hormones by the small intestine. The small intestine is not only responsible for the digestion and transport of dietary triacylglycerol, through the formation of chylomicrons, but it also secretes the incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) from enteroendocrine cells. Ultimately, both chylomicrons and incretins are found in lymph. Advantages of the lymph fistula rat model in studying chylomicron and incretin secretion are numerous and include: 1) the concentrations of incretin hormones are higher in lymph than in peripheral or portal plasma; 2) there is reduced degradation of incretin hormones by DPP-IV in the lymph compartment; 3) less dilution by the circulating fluid; 4) this model allows the continuous collection of lymph from conscious animals, eliminating any potential side effects on lymph flow and gastrointestinal function due to anesthesia; and finally, and perhaps most importantly, and 5) the concentration in the intestinal lymph provides a physiologically accurate representation of the hormonal milieu within the intestinal mucosa where incretins may interact with enteroendocrine and/or dendritic cells and signal through the enteric or autonomic neurons. The importance of GIP and GLP-1 in health and disease is becoming more apparent, especially as the prevalence of type 2 diabetes and other metabolic disorders increases. This review focuses on the use of the lymph fistula rat as a model to study the secretion of incretins, as well as dietary lipid.
Collapse
Affiliation(s)
- Alison B. Kohan
- Department of Pathology, College of Medicine Metabolic Diseases Institute University of Cincinnati 2180 E. Galbraith Road, ML 0507, Cincinnati, OH 45237
| | - Stephanie M. Yoder
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research Indiana University School of Medicine 635 Barnhill Drive, MS 2055 Indianapolis, IN 46202
| | - Patrick Tso
- Department of Pathology, College of Medicine Metabolic Diseases Institute University of Cincinnati 2180 E. Galbraith Road, ML 0507, Cincinnati, OH 45237
| |
Collapse
|
47
|
Steinert RE, Beglinger C. Nutrient sensing in the gut: interactions between chemosensory cells, visceral afferents and the secretion of satiation peptides. Physiol Behav 2011; 105:62-70. [DOI: 10.1016/j.physbeh.2011.02.039] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Revised: 02/24/2011] [Accepted: 02/25/2011] [Indexed: 01/01/2023]
|
48
|
Kitahara Y, Miura K, Yasuda R, Kawanabe H, Ogawa S, Eto Y. Nateglinide stimulates glucagon-like peptide-1 release by human intestinal L cells via a K(ATP) channel-independent mechanism. Biol Pharm Bull 2011; 34:671-6. [PMID: 21532155 DOI: 10.1248/bpb.34.671] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A reduced incretin effect is one of the well-known characteristics of patients with type 2 diabetes, and impaired release of glucagon-like peptide-1 (GLP-1) has been reported to be at least partly involved. In this study, we investigated the effect of nateglinide on GLP-1 release in vivo and in vitro. The GLP-1 level in the portal blood at 20 min after oral administration of nateglinide to Wistar rats was about twice that in vehicle-treated rats. To clarify whether this effect of nateglinide was related to direct stimulation of intestinal cells, in vitro studies were performed using human intestinal L cells (NCI-H716). Nateglinide stimulated GLP-1 release in a concentration-dependent manner from 500 µM, along with transient elevation of the intracellular calcium level. However, diazoxide, nitrendipine, and dantrolene did not block this effect of nateglinide. In addition, the major metabolite of nateglinide, tolbutamide, and mitiglinide, all of which augment insulin secretion by the pancreatic islets, had no effect on GLP-1 release by this cell line. On the other hand, capsazepine significantly inhibited the promotion of GLP-1 release by nateglinide in a concentration-dependent manner. These findings indicate that nateglinide directly stimulates GLP-1 release by intestinal L cells in a K(ATP) channel-independent manner. A novel target of nateglinide may be involved in increasing intracellular calcium to stimulate GLP-1 release, e.g., the transient receptor potential channels. Taken together, the present findings indicate that promotion of GLP-1 release from intestinal L cells may be another important mechanism by which nateglinide restores early-phase insulin secretion and regulates postprandial glucose metabolism.
Collapse
Affiliation(s)
- Yoshiro Kitahara
- Exploratory Research Laboratories, Ajinomoto Pharmaceuticals Co., Ltd, Kawasaki, Japan.
| | | | | | | | | | | |
Collapse
|
49
|
D'Alessio DA. What if gut hormones aren't really hormones: DPP-4 inhibition and local action of GLP-1 in the gastrointestinal tract. Endocrinology 2011; 152:2925-6. [PMID: 21785013 PMCID: PMC3138230 DOI: 10.1210/en.2011-1385] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- David A D'Alessio
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, ML 0547, University of Cincinnati, Cincinnati Ohio 45267-0547, USA.
| |
Collapse
|
50
|
K-cells and glucose-dependent insulinotropic polypeptide in health and disease. VITAMINS AND HORMONES 2011; 84:111-50. [PMID: 21094898 DOI: 10.1016/b978-0-12-381517-0.00004-7] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In the 1970s, glucose-dependent insulinotropic polypeptide (GIP, formerly gastric inhibitory polypeptide), a 42-amino acid peptide hormone, was discovered through a search for enterogastrones and subsequently identified as an incretin, or an insulinotropic hormone secreted in response to intraluminal nutrients. Independent of the discovery of GIP, the K-cell was identified in small intestine by characteristic ultrastructural features. Subsequently, it was realized that K-cells are the predominant source of circulating GIP. The density of K-cells may increase under conditions including high-fat diet and obesity, and generally correlates with plasma GIP levels. In addition to GIP, K-cells secrete xenin, a peptide with as of yet poorly understood physiological functions, and GIP is often colocalized with the other incretin hormone glucagon-like peptide-1 (GLP-1). Differential posttranslational processing of proGIP produces 30 and 42 amino acid versions of GIP. Its secretion is elicited by intraluminal nutrients, especially carbohydrate and fat, through the action of SGLT1, GPR40, GPR120, and GPR119. There is also evidence of regulation of GIP secretion via neural pathways and somatostatin. Intracellular signaling mechanisms of GIP secretion are still elusive but include activation of adenylyl cyclase, protein kinase A (PKA), and protein kinase C (PKC). GIP has extrapancreatic actions on adipogenesis, neural progenitor cell proliferation, and bone metabolism. However, the clinical or physiological relevance of these extrapancreatic actions remain to be defined in humans. The application of GIP as a glucose-lowering drug is limited due to reduced efficacy in humans with type 2 diabetes and its potential obesogenic effects demonstrated by rodent studies. There is some evidence to suggest that a reduction in GIP production or action may be a strategy to reduce obesity. The meal-dependent nature of GIP release makes K-cells a potential target for genetically engineered production of satiety factors or glucose-lowering agents, for example, insulin. Transgenic mice engineered to produce insulin from intestinal K-cells are resistant to diabetes induced by a beta-cell toxin. Collectively, K-cells and GIP play important roles in health and disease, and both may be targets for novel therapies.
Collapse
|