1
|
Yang K, Wu YT, He Y, Dai JX, Luo YL, Xie JH, Ding WJ. GLP-1 and IL-6 regulates obesity in the gut and brain. Life Sci 2025; 362:123339. [PMID: 39730038 DOI: 10.1016/j.lfs.2024.123339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/06/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Obesity is a chronic metabolic disease characterized by excessive nutrient intake leading to increased subcutaneous or visceral fat, resulting in pathological and physiological changes. The incidence rate of obesity, an important form of metabolic syndrome, is increasing worldwide. Excess appetite is a key pathogenesis of obesity, and the inflammatory response induced by obesity has received increasing attention. This review focuses on the role of appetite-regulating factor (Glucogan-like peptide 1) and inflammatory factor (Interleukin-6) in the gut and brain in individuals with obesity and draws insights from the current literature.
Collapse
Affiliation(s)
- Kun Yang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yu-Ting Wu
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yan He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jin-Xiu Dai
- Chengdu University of Traditional Chinese Medicine, 1166 Luitai Avenue, Chengdu, Sichuan 611137, China
| | - Yu-Lu Luo
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jing-Hui Xie
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei-Jun Ding
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
2
|
Yang Y, Luo J, Wang S, Yang D, Chen S, Wang Q, Zhou A. A water-soluble polysaccharide from finger citron ameliorates diabetes mellitus via gut microbiota-GLP-1 pathway in high-fat diet and streptozotocin-induced diabetic mice. Int J Biol Macromol 2025; 300:140126. [PMID: 39842590 DOI: 10.1016/j.ijbiomac.2025.140126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 01/08/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
FCP-2-1, a water-soluble polysaccharide isolated and purified from Finger Citron, demonstrated hypoglycemic effect in vitro in our previous study. However, its antidiabetic effect and underlying mechanism in vivo remain to be elucidated. In this study, the antidiabetic effect of FCP-2-1 and its effects on the gut microbiota, short-chain fatty acids (SCFAs), and glucagon-like peptide-1 (GLP-1) in high-fat diet (HFD) and streptozotocin (STZ)-induced diabetic mice were investigated. The results showed that FCP-2-1 could significantly alleviate diabetic symptoms in diabetic mice, restore the balance of the gut microbiota, and increase the content of acetic acid and propionic acid. In particular, FCP-2-1 was found to be able to promote the secretion of GLP-1, a new therapeutic target for diabetes. Moreover, propionic acid and FCP-2-1 were able to promote GLP-1 secretion in NCI-H716 cells, suggesting that FCP-2-1 could stimulate the secretion of GLP-1 through itself and propionic acid produced by the gut microbiota. These findings indicated that the antidiabetic mechanism of FCP-2-1 might be related to the gut microbiota-GLP-1 pathway.
Collapse
Affiliation(s)
- Yujie Yang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Junyun Luo
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Shuhui Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Dan Yang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Shuxi Chen
- Guangdong Zhancui Food Co. Ltd., Chaozhou 515634, China
| | - Qun Wang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| | - Aimei Zhou
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
3
|
Kim KS, Peck BC, Hung YH, Koch-Laskowski K, Wood L, Dedhia PH, Spence JR, Seeley RJ, Sethupathy P, Sandoval DA. Vertical sleeve gastrectomy induces enteroendocrine cell differentiation of intestinal stem cells through bile acid signaling. JCI Insight 2022; 7:154302. [PMID: 35503251 PMCID: PMC9220851 DOI: 10.1172/jci.insight.154302] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/26/2022] [Indexed: 11/24/2022] Open
Abstract
Vertical sleeve gastrectomy (VSG) results in an increase in the number of hormone-secreting enteroendocrine cells (EECs) in the intestinal epithelium; however, the mechanism remains unclear. Notably, the beneficial effects of VSG are lost in a mouse model lacking the nuclear bile acid receptor farnesoid X receptor (FXR). FXR is a nuclear transcription factor that has been shown to regulate intestinal stem cell (ISC) function in cancer models. Therefore, we hypothesized that the VSG-induced increase in EECs is due to changes in intestinal differentiation driven by an increase in bile acid signaling through FXR. To test this, we performed VSG in mice that express EGFP in ISC/progenitor cells and performed RNA-Seq on GFP-positive cells sorted from the intestinal epithelia. We also assessed changes in EEC number (marked by glucagon-like peptide-1, GLP-1) in mouse intestinal organoids following treatment with bile acids, an FXR agonist, and an FXR antagonist. RNA-Seq of ISCs revealed that bile acid receptors are expressed in ISCs and that VSG explicitly alters expression of several genes that regulate EEC differentiation. Mouse intestinal organoids treated with bile acids and 2 different FXR agonists increased GLP-1-positive cell numbers, and administration of an FXR antagonist blocked these effects. Taken together, these data indicate that VSG drives ISC fate toward EEC differentiation through bile acid signaling.
Collapse
Affiliation(s)
- Ki-Suk Kim
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Bailey Ce Peck
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Yu-Han Hung
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, USA
| | | | - Landon Wood
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Priya H Dedhia
- Department of Surgery, The Ohio State University Comprehensive Cancer Center and The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Praveen Sethupathy
- Department of Biomedical Sciences, Cornell University, Ithaca, New York, USA
| | - Darleen A Sandoval
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
4
|
Lu VB, Gribble FM, Reimann F. Nutrient-Induced Cellular Mechanisms of Gut Hormone Secretion. Nutrients 2021; 13:nu13030883. [PMID: 33803183 PMCID: PMC8000029 DOI: 10.3390/nu13030883] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/27/2021] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
The gastrointestinal tract can assess the nutrient composition of ingested food. The nutrient-sensing mechanisms in specialised epithelial cells lining the gastrointestinal tract, the enteroendocrine cells, trigger the release of gut hormones that provide important local and central feedback signals to regulate nutrient utilisation and feeding behaviour. The evidence for nutrient-stimulated secretion of two of the most studied gut hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), along with the known cellular mechanisms in enteroendocrine cells recruited by nutrients, will be the focus of this review. The mechanisms involved range from electrogenic transporters, ion channel modulation and nutrient-activated G-protein coupled receptors that converge on the release machinery controlling hormone secretion. Elucidation of these mechanisms will provide much needed insight into postprandial physiology and identify tractable dietary approaches to potentially manage nutrition and satiety by altering the secreted gut hormone profile.
Collapse
|
5
|
Obaroakpo JU, Liu L, Zhang S, Lu J, Liu L, Pang X, Lv J. In vitro modulation of glucagon-like peptide release by DPP-IV inhibitory polyphenol-polysaccharide conjugates of sprouted quinoa yoghurt. Food Chem 2020; 324:126857. [DOI: 10.1016/j.foodchem.2020.126857] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 03/09/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
|
6
|
Tomaro-Duchesneau C, LeValley SL, Roeth D, Sun L, Horrigan FT, Kalkum M, Hyser JM, Britton RA. Discovery of a bacterial peptide as a modulator of GLP-1 and metabolic disease. Sci Rep 2020; 10:4922. [PMID: 32188864 PMCID: PMC7080827 DOI: 10.1038/s41598-020-61112-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
Early work in rodents highlighted the gut microbiota’s importance in metabolic disease, including Type II Diabetes Mellitus (T2DM) and obesity. Glucagon-like peptide-1 (GLP-1), an incretin secreted by L-cells lining the gastrointestinal epithelium, has important functions: promoting insulin secretion, insulin sensitivity, and β-cell mass, while inhibiting gastric emptying and appetite. We set out to identify microbial strains with GLP-1 stimulatory activity as potential metabolic disease therapeutics. Over 1500 human-derived strains were isolated from healthy individuals and screened for GLP-1 modulation by incubating bacterial cell-free supernatants with NCI H716 L-cells. Approximately 45 strains capable of increasing GLP-1 were discovered. All GLP-1 positive strains were identified as Staphylococcus epidermidis by 16S rRNA sequencing. Mass spectrometry analysis identified a 3 kDa peptide, Hld (delta-toxin), present in GLP-1 positive supernatants but absent in GLP-1 neutral supernatants. Studies in NCI-H716 cells and human jejunal enteroids engineered to make more enteroendocrine cells demonstrated that Hld alone is sufficient to enhance GLP-1 secretion. When administered in high-fat-fed mice, Hld-producing S. epidermidis significantly reduced markers associated with obesity and T2DM. Further characterization of Hld suggests GLP-1 stimulatory action of Hld occurs via calcium signaling. The presented results identify a novel host-microbe interaction which may ultimately lead to the development of a microbial peptide-based therapeutic for metabolic disease.
Collapse
Affiliation(s)
- Catherine Tomaro-Duchesneau
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Stephanie L LeValley
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daniel Roeth
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Liang Sun
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Frank T Horrigan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Markus Kalkum
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.
| |
Collapse
|
7
|
Sinagoga KL, McCauley HA, Múnera JO, Reynolds NA, Enriquez JR, Watson C, Yang HC, Helmrath MA, Wells JM. Deriving functional human enteroendocrine cells from pluripotent stem cells. Development 2018; 145:dev.165795. [PMID: 30143540 DOI: 10.1242/dev.165795] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022]
Abstract
Enteroendocrine cells (EECs) are a minor cell population in the intestine yet they play a major role in digestion, satiety and nutrient homeostasis. Recently developed human intestinal organoid models include EECs, but their rarity makes it difficult to study their formation and function. Here, we used the EEC-inducing property of the transcription factor NEUROG3 in human pluripotent stem cell-derived human intestinal organoids and colonic organoids to promote EEC development in vitro An 8-h pulse of NEUROG3 expression induced expression of known target transcription factors and after 7 days organoids contained up to 25% EECs in the epithelium. EECs expressed a broad array of human hormones at the mRNA and/or protein level, including motilin, somatostatin, neurotensin, secretin, substance P, serotonin, vasoactive intestinal peptide, oxyntomodulin, GLP-1 and INSL5. EECs secreted several hormones including gastric inhibitory polypeptide (GIP), ghrelin, GLP-1 and oxyntomodulin. Injection of glucose into the lumen of organoids caused an increase in both GIP secretion and K-cell number. Lastly, we observed formation of all known small intestinal EEC subtypes following transplantation and growth of human intestinal organoids in mice.
Collapse
Affiliation(s)
- Katie L Sinagoga
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Heather A McCauley
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Jorge O Múnera
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Nichole A Reynolds
- Endocrine/Cardiovascular Division, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Jacob R Enriquez
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Carey Watson
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - Hsiu-Chiung Yang
- Endocrine/Cardiovascular Division, Eli Lilly and Company, Indianapolis, IN 46285, USA
| | - Michael A Helmrath
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA.,Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| | - James M Wells
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA .,Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave, Cincinnati, OH 45229-3039, USA
| |
Collapse
|
8
|
Ang SY, Evans BA, Poole DP, Bron R, DiCello JJ, Bathgate RAD, Kocan M, Hutchinson DS, Summers RJ. INSL5 activates multiple signalling pathways and regulates GLP-1 secretion in NCI-H716 cells. J Mol Endocrinol 2018. [PMID: 29535183 DOI: 10.1530/jme-17-0152] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Insulin-like peptide 5 (INSL5) is a newly discovered gut hormone expressed in colonic enteroendocrine L-cells but little is known about its biological function. Here, we show using RT-qPCR and in situ hybridisation that Insl5 mRNA is highly expressed in the mouse colonic mucosa, colocalised with proglucagon immunoreactivity. In comparison, mRNA for RXFP4 (the cognate receptor for INSL5) is expressed in various mouse tissues, including the intestinal tract. We show that the human enteroendocrine L-cell model NCI-H716 cell line, and goblet-like colorectal cell lines SW1463 and LS513 endogenously express RXFP4. Stimulation of NCI-H716 cells with INSL5 produced phosphorylation of ERK1/2 (Thr202/Tyr204), AKT (Thr308 and Ser473) and S6RP (Ser235/236) and inhibited cAMP production but did not stimulate Ca2+ release. Acute INSL5 treatment had no effect on GLP-1 secretion mediated by carbachol or insulin, but modestly inhibited forskolin-stimulated GLP-1 secretion in NCI-H716 cells. However, chronic INSL5 pre-treatment (18 h) increased basal GLP-1 secretion and prevented the inhibitory effect of acute INSL5 administration. LS513 cells were found to be unresponsive to INSL5 despite expressing RXFP4 Another enteroendocrine L-cell model, mouse GLUTag cells did not express detectable levels of Rxfp4 and were unresponsive to INSL5. This study provides novel insights into possible autocrine/paracrine roles of INSL5 in the intestinal tract.
Collapse
Affiliation(s)
- Sheng Y Ang
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Bronwyn A Evans
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Daniel P Poole
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Romke Bron
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jesse J DiCello
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ross A D Bathgate
- The Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular BiologyUniversity of Melbourne, Melbourne, Victoria, Australia
| | - Martina Kocan
- The Florey Institute of Neuroscience and Mental HealthUniversity of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular BiologyUniversity of Melbourne, Melbourne, Victoria, Australia
| | - Dana S Hutchinson
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Department of PharmacologyMonash University, Clayton, Victoria, Australia
| | - Roger J Summers
- Drug Discovery BiologyMonash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
9
|
Green B, Connolly L. Response to comments raised by Fernstrom and Smiga (TOXLET-D-17-00079) on our recent article Shannon M et al. [Toxicol. Lett. 265 (2017) 97]. Toxicol Lett 2017; 272:103-105. [DOI: 10.1016/j.toxlet.2017.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 11/16/2022]
|
10
|
Tian L, Jin T. The incretin hormone GLP-1 and mechanisms underlying its secretion. J Diabetes 2016; 8:753-765. [PMID: 27287542 DOI: 10.1111/1753-0407.12439] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/09/2016] [Accepted: 06/02/2016] [Indexed: 12/25/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a cell type-specific post-translational product of proglucagon. It is encoded by the proglucagon gene and released primarily from intestinal endocrine L-cells in response to hormonal, neuronal, and nutritional stimuli. In addition to serving as an incretin in mediating the effect of meal consumption on insulin secretion, GLP-1 exerts other functions in pancreatic islets, including regulation of β-cell proliferation and protection of β-cells against metabolic stresses. Furthermore, GLP-1 exerts numerous other functions in extrapancreatic organs, whereas brain GLP-1 signaling controls satiety. Herein we review the discovery of two incretins and the development of GLP-1-based drugs. We also describe the development of cellular models for studying mechanisms underlying GLP-1 secretion over the past 30 years. However, the main content of this review is a summary of studies on the exploration of mechanisms underlying GLP-1 secretion. We not only summarize studies conducted over the past three decades on elucidating the role of nutritional components and hormonal factors in regulating GLP-1 secretion, but also present a few very recent studies showing the possible role of dietary polyphenols. Finally, the emerging role of gut microbiota in metabolic homeostasis with the potential implication on GLP-1 secretion is discussed.
Collapse
Affiliation(s)
- Lili Tian
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada.
- Banting & Best Diabetes Centre, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Protein digestion and energy homeostasis: How generated peptides may impact intestinal hormones? Food Res Int 2016. [DOI: 10.1016/j.foodres.2015.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
12
|
Kuhre RE, Wewer Albrechtsen NJ, Deacon CF, Balk-Møller E, Rehfeld JF, Reimann F, Gribble FM, Holst JJ. Peptide production and secretion in GLUTag, NCI-H716, and STC-1 cells: a comparison to native L-cells. J Mol Endocrinol 2016; 56:201-11. [PMID: 26819328 PMCID: PMC7212058 DOI: 10.1530/jme-15-0293] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
GLUTag, NCI-H716, and STC-1 are cell lines that are widely used to study mechanisms underlying secretion of glucagon-like peptide-1 (GLP-1), but the extent to which they resemble native L-cells is unknown. We used validated immunoassays for 14 different hormones to analyze peptide content (lysis samples; n = 9 from different passage numbers) or peptide secretion in response to buffer (baseline), and after stimulation with 50 mM KCl or 10 mM glucose + 10 µM forskolin/3-isobutyl-1-methylxanthine (n = 6 also different passage numbers). All cell lines produced and processed proglucagon into GLP-1, GLP-2, glicentin, and oxyntomodulin in a pattern (prohormone convertase (PC)1/3 dependent) similar to that described for human gut. All three cell lines showed basal secretion of GLP-1 and GLP-2, which increased after stimulation. In contrast to freshly isolated murine L-cells, all cell lines also expressed PC2 and secreted large amounts of pancreatic glucagon. Neurotensin and somatostatin storage was low and secretion was not consistently increased by stimulation. STC-1 cells released more glucose-dependent insulinotropic polypeptide than GLP-1 at baseline (P < 0.01) and KCl elevated its secretion (P < 0.05). Peptide YY, which normally co-localizes with GLP-1 in distal L-cells, was not detected in any of the cell lines. GLUTag and STC-1 cells also expressed vasoactive intestinal peptide, but none expressed pancreatic polypeptide or insulin. GLUTag contained and secreted large amounts of CCK, while NCI-H716 did not store this peptide and STC-1 contained low amounts. Our results show that hormone production in cell line models of the L-cell has limited similarity to the natural L-cells.
Collapse
Affiliation(s)
- Rune Ehrenreich Kuhre
- Department of Biomedical Sciences and NNF Center for Basic Metabolic Researchthe Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Nicolai Jacob Wewer Albrechtsen
- Department of Biomedical Sciences and NNF Center for Basic Metabolic Researchthe Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Carolyn Fiona Deacon
- Department of Biomedical Sciences and NNF Center for Basic Metabolic Researchthe Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Emilie Balk-Møller
- Department of Biomedical Sciences and NNF Center for Basic Metabolic Researchthe Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jens Frederik Rehfeld
- Department of Clinical Biochemistry RigshospitaletUniversity of Copenhagen, Copenhagen, Denmark
| | - Frank Reimann
- Cambridge Institute for Medical Research and MRC Metabolic Diseases UnitUniversity of Cambridge, Cambridge, United Kingdom
| | - Fiona Mary Gribble
- Cambridge Institute for Medical Research and MRC Metabolic Diseases UnitUniversity of Cambridge, Cambridge, United Kingdom
| | - Jens Juul Holst
- Department of Biomedical Sciences and NNF Center for Basic Metabolic Researchthe Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Characterisation of insulin-producing cells differentiated from tonsil derived mesenchymal stem cells. Differentiation 2015; 90:27-39. [DOI: 10.1016/j.diff.2015.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 07/25/2015] [Accepted: 08/30/2015] [Indexed: 01/22/2023]
|
14
|
El-Jamal N, Erdual E, Neunlist M, Koriche D, Dubuquoy C, Maggiotto F, Chevalier J, Berrebi D, Dubuquoy L, Boulanger E, Cortot A, Desreumaux P. Glugacon-like peptide-2: broad receptor expression, limited therapeutic effect on intestinal inflammation and novel role in liver regeneration. Am J Physiol Gastrointest Liver Physiol 2014; 307:G274-85. [PMID: 24875097 DOI: 10.1152/ajpgi.00389.2012] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The glucagon-like peptide 2 (GLP-2) is an intestinotrophic hormone with growth promoting and anti-inflammatory actions. However, the full biological functions of GLP-2 and the localization of its receptor (GLP-2R) remain controversial. Among cell lines tested, the expression of GLP-2R transcript was detected in human colonic myofibroblasts (CCD-18Co) and in primary culture of rat enteric nervous system but not in intestinal epithelial cell lines, lymphocytes, monocytes, or endothelial cells. Surprisingly, GLP-2R was expressed in murine (GLUTag), but not human (NCI-H716) enteroendocrine cells. The screening of GLP-2R mRNA in mice organs revealed an increasing gradient of GLP-2R toward the distal gut. An unexpected expression was detected in the mesenteric fat, mesenteric lymph nodes, bladder, spleen, and liver, particularly in hepatocytes. In two mice models of trinitrobenzene sulfonic acid (TNBS)- and dextran sulfate sodium (DSS)-induced colitis, the colonic expression of GLP-2R mRNA was decreased by 60% compared with control mice. Also, GLP-2R mRNA was significantly downregulated in intestinal tissues of inflammatory bowel disease patients. Therapeutically, GLP-2 showed a weak restorative effect on intestinal inflammation during TNBS-induced colitis as assessed by macroscopic score and inflammatory markers. Finally, GLP-2 treatment accelerated mouse liver regeneration following partial hepatectomy as assessed by histological and molecular analyses. In conclusion, the limited therapeutic effect of GLP-2 on colonic inflammation dampens its utility in the management of severe inflammatory intestinal disorders. However, the role of GLP-2 in liver regeneration is a novelty that might introduce GLP-2 into the management of liver diseases and emphasizes on the importance of elucidating other extraintestinal functions of GLP-2.
Collapse
Affiliation(s)
- Noura El-Jamal
- INSERM U995, Lille, France; Université Lille Nord de France, Lille, France; Intestinal Biotech Development, Lille, France
| | - Edmone Erdual
- INSERM U995, Lille, France; Université Lille Nord de France, Lille, France
| | | | - Dine Koriche
- CHU Lille, Service des maladies de l'appareil digestif et de la nutrition, Hôpital Claude Huriez, Lille, France
| | - Caroline Dubuquoy
- INSERM U995, Lille, France; Intestinal Biotech Development, Lille, France
| | - Francois Maggiotto
- INSERM U995, Lille, France; Université Lille Nord de France, Lille, France
| | | | - Dominique Berrebi
- Université Paris-Sud, Laboratoire "Cytokines, Chimiokines et Immunopathologie," Unité Mixte de Recherche S996, Clamart, France; INSERM, Laboratoire d'Excellence en Recherche sur le Médicament et l'Innovation Thérapeutique, Clamart, France; Service d'Anatomie et de Cytologie Pathologique, Unité Propre de Recherche de l'Enseignement Supérieur Associé 1320, Hôpital Robert Debré, France; Université Denis Diderot, Université Paris 7, Paris, France; and
| | - Laurent Dubuquoy
- INSERM U995, Lille, France; Université Lille Nord de France, Lille, France
| | - Eric Boulanger
- Université Lille Nord de France, Lille, France; EA 2693, Lille, France
| | - Antoine Cortot
- INSERM U995, Lille, France; Université Lille Nord de France, Lille, France; CHU Lille, Service des maladies de l'appareil digestif et de la nutrition, Hôpital Claude Huriez, Lille, France
| | - Pierre Desreumaux
- INSERM U995, Lille, France; Université Lille Nord de France, Lille, France; CHU Lille, Service des maladies de l'appareil digestif et de la nutrition, Hôpital Claude Huriez, Lille, France;
| |
Collapse
|
15
|
Gagnon J, Anini Y. Insulin and norepinephrine regulate ghrelin secretion from a rat primary stomach cell culture. Endocrinology 2012; 153:3646-56. [PMID: 22691550 DOI: 10.1210/en.2012-1040] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Ghrelin is a peptide hormone primarily produced in the previously unidentified X/A endocrine cells of the stomach. Extensive studies have focused on the effects of ghrelin on growth hormone release and appetite regulation. However, the mechanisms regulating ghrelin secretion are less understood. In the present study, we developed a primary culture of newborn rat stomach cells to investigate the mechanisms regulating ghrelin synthesis and secretion. We demonstrated that this cell preparation secretes ghrelin in a regulated manner through the increase of cAMP, intracellular calcium, and activation of protein kinase C. Norepinephrine (NE) (0.1-10 μm) stimulated ghrelin secretion through the β1-adrenergic receptor via increased cAMP and protein kinase A activity, whereas acetylcholine had no effect. Because circulating ghrelin levels were previously shown to be inversely correlated with insulin levels, we investigated the effect of insulin on ghrelin secretion. We first demonstrated that ghrelin cells express the insulin receptor α- and β-subunits. Next, we determined that insulin (1-10 nm) inhibited both basal and NE-stimulated ghrelin secretion, caused an increase in phosphorylated serine-threonine kinase (AKT) and a reduction in intracellular cAMP, but did not alter proghrelin mRNA levels. The inhibitory effect of insulin was blocked by inhibiting phospho-inositol-3 kinase and AKT but not MAPK. Higher dose insulin (100 nm) did not suppress ghrelin secretion, which prompted the investigation of cellular insulin resistance by pretreating the cells with 100 nm insulin for 24 h. This caused a reduction in insulin receptor expression and prevented the insulin-mediated AKT activation and the suppression of ghrelin secretion with no impact on NE-stimulated ghrelin secretion. Our findings highlight the role of the sympathetic nervous system, insulin, and insulin resistance in the regulation of ghrelin secretion.
Collapse
Affiliation(s)
- Jeffrey Gagnon
- Department of Obstetrics, Faculty of Medicine,Dalhousie University, Halifax, Nova Scotia, Canada
| | | |
Collapse
|
16
|
Le Nevé B, Daniel H. Selected tetrapeptides lead to a GLP-1 release from the human enteroendocrine cell line NCI-H716. ACTA ACUST UNITED AC 2010; 167:14-20. [PMID: 21070823 DOI: 10.1016/j.regpep.2010.10.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Revised: 09/17/2010] [Accepted: 10/29/2010] [Indexed: 10/18/2022]
Abstract
Enteroendocrine cells in the intestine sense the luminal contents and have been shown to respond to not only fatty acids, proteins, and monosaccharides but also artificial sweeteners and bitter compounds. Secretion of hormones such as CCK and GLP-1 from these cells is often associated with a rise in intracellular calcium concentration [Ca²+](i). The human NCI-H716 enteroendocrine cell line has been described as a proper model to study GLP-1 secretion in response to amino acids and protein hydrolysates. Here, we describe that NCI-H716 cells specifically respond to selective tetrapeptides such as tetra-glycine, tetra-alanine and Gly-Trp-Gly-Gly with a dose-dependent [Ca²+](i) response and a GLP-1 secretion, whereas selected free amino acids, dipeptides, tripeptides and pentapeptides failed to elicit such a response. Hormone secretion was not associated with changes in cAMP levels in the cells. The calcium-dependence of hormone secretion appears to involve store-operated calcium channels (SOCCs), since the SOCC blocker 2-APB abolished both the [Ca²+](i) response and GLP-1 release upon tetra-glycine stimulation. The nature of the sensor currently remains elusive, and no obvious common structural pattern in tetrapeptides eliciting GLP-1 secretion was identified. This tetrapeptide sensing in NCI-H716 cells may be underlying the effective stimulation of hormone secretion shown for various protein hydrolysates, and could involve a novel G-protein-coupled receptor (GPCR).
Collapse
Affiliation(s)
- Boris Le Nevé
- Molecular Nutrition Unit, Technical University of Munich, Freising-Weihenstephan, Germany
| | | |
Collapse
|
17
|
Nikoulina SE, Andon NL, McCowen KM, Hendricks MD, Lowe C, Taylor SW. A primary colonic crypt model enriched in enteroendocrine cells facilitates a peptidomic survey of regulated hormone secretion. Mol Cell Proteomics 2010; 9:728-41. [PMID: 20081152 DOI: 10.1074/mcp.m900529-mcp200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
To enable the first physiologically relevant peptidomic survey of gastrointestinal tissue, we have developed a primary mouse colonic crypt model enriched for enteroendocrine L-cells. The cells in this model were phenotypically profiled using PCR-based techniques and showed peptide hormone and secretory and processing marker expression at mRNA levels that were increased relative to the parent tissue. Co-localization of glucagon-like peptide-1 and peptide YY, a characteristic feature of L-cells, was demonstrated by double label immunocytochemistry. The L-cells displayed regulated hormone secretion in response to physiological and pharmacological stimuli as measured by immunoassay. Using a high resolution mass spectrometry-based platform, more than 50 endogenous peptides (<16 kDa), including all known major hormones, were identified a priori. The influence of culture conditions on peptide relative abundance and post-translational modification was characterized. The relative abundance of secreted peptides in the presence/absence of the stimulant forskolin was measured by label-free quantification. All peptides exhibiting a statistically significant increase in relative concentration in the culture media were derived from prohormones, consistent with a cAMP-coupled response. The only peptides that exhibited a statistically significant decrease in secretion on forskolin stimulation were derived from annexin A1 and calcyclin. Biophysical interactions between annexin A1 and calcyclin have been reported very recently and may have functional consequences. This work represents the first step in characterizing physiologically relevant peptidomic secretion of gastrointestinally derived primary cells and will aid in elucidating new endocrine function.
Collapse
|
18
|
Parker HE, Reimann F, Gribble FM. Molecular mechanisms underlying nutrient-stimulated incretin secretion. Expert Rev Mol Med 2010; 12:e1. [PMID: 20047700 DOI: 10.1017/s146239940900132x] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The incretin hormones glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are released from enteroendocrine cells in the intestinal epithelium in response to nutrient ingestion. The actions of GLP-1 and GIP - not only on local gut physiology but also on glucose homeostasis, appetite control and fat metabolism - have made these hormones an attractive area for drug discovery programmes. The potential range of strategies to target the secretion of these hormones therapeutically has been limited by an incomplete understanding of the mechanisms underlying their release. The use of organ and whole-animal perfusion techniques, cell line models and primary L- and K-cells has led to the identification of a variety of pathways involved in the sensing of carbohydrate, fat and protein in the gut lumen. This review focuses on our current understanding of these signalling mechanisms that might underlie nutrient responsiveness of L- and K-cells.
Collapse
Affiliation(s)
- Helen E Parker
- Cambridge Institute for Medical Research and Department of Clinical Biochemistry, Addenbrooke's Hospital, Cambridge, UK
| | | | | |
Collapse
|
19
|
Affiliation(s)
- M Laakso
- Department of Medicine, University of Kuopio, Kuopio University Hospital, 70210, Kuopio, Finland.
| |
Collapse
|
20
|
Wen JH, Chen YY, Song SJ, Ding J, Gao Y, Hu QK, Feng RP, Liu YZ, Ren GC, Zhang CY, Hong TP, Gao X, Li LS. Paired box 6 (PAX6) regulates glucose metabolism via proinsulin processing mediated by prohormone convertase 1/3 (PC1/3). Diabetologia 2009; 52:504-13. [PMID: 19034419 DOI: 10.1007/s00125-008-1210-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Accepted: 08/20/2008] [Indexed: 11/30/2022]
Abstract
AIMS/HYPOTHESIS Human patients with aniridia caused by heterozygous PAX6 mutations display abnormal glucose metabolism, but the underlying molecular mechanism is largely unknown. Disturbed islet architecture has been proposed as the reason why mice with complete inactivation of paired box 6 (PAX6) in the pancreas develop diabetes. This is not, however, the case in human aniridia patients with heterozygous PAX6 deficiency and no apparent defects in pancreatic development. We investigated the molecular mechanism underlying the development of abnormal glucose metabolism in these patients. METHODS A human aniridia pedigree with a PAX6 R240Stop mutation was examined for abnormal glucose metabolism using an OGTT. The underlying mechanism was further investigated using Pax6 R266Stop mutant small-eye mice, which also have abnormal glucose metabolism similar to that in PAX6 R240Stop mutation human aniridia patients. RESULTS Paired box 6 (PAX6) deficiency, both in aniridia patients with a heterozygous PAX6 R240Stop mutation and in mice with a heterozygous Pax6 R266Stop mutation, causes defective proinsulin processing and abnormal glucose metabolism. PAX6 can bind to the promoter and directly upregulate production of prohormone convertase (PC)1/3, an enzyme essential for conversion of proinsulin to insulin. Pax6 mutations lead to PC1/3 deficiency, resulting in defective proinsulin processing and abnormal glucose metabolism. CONCLUSIONS/INTERPRETATION This study indicates a novel function for PAX6 in the regulation of proinsulin processing and glucose metabolism via modulation of PC1/3 production. It also provides an insight into the abnormal glucose metabolism caused by heterozygous PAX6 mutations in humans and mice.
Collapse
Affiliation(s)
- J H Wen
- Peking University Stem Cell Research Center, China-Australian Center of Excellence for Stem Cell Sciences, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Scicchitano MS, McFarland DC, Tierney LA, Boyce RW, Frazier KS, Schwartz LW, Thomas HC. Role of p38 in regulation of hematopoiesis: Effect of p38 inhibition on cytokine production and transcription factor activity in human bone marrow stromal cells. Blood Cells Mol Dis 2008; 40:370-80. [DOI: 10.1016/j.bcmd.2007.10.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 10/30/2007] [Indexed: 10/22/2022]
|
22
|
Ruhnke M, Ungefroren H, Nussler A, Martin F, Brulport M, Schormann W, Hengstler JG, Klapper W, Ulrichs K, Hutchinson JA, Soria B, Parwaresch RM, Heeckt P, Kremer B, Fändrich F. Differentiation of in vitro-modified human peripheral blood monocytes into hepatocyte-like and pancreatic islet-like cells. Gastroenterology 2005; 128:1774-86. [PMID: 15940611 DOI: 10.1053/j.gastro.2005.03.029] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Adult stem cells provide a promising alternative for the treatment of diabetes mellitus and end-stage liver diseases. We evaluated the differentiation potential of human peripheral blood monocytes into hepatocyte-like and pancreatic islet-like cells. METHODS Monocytes were treated with macrophage colony-stimulating factor and interleukin 3 for 6 days, followed by incubation with hepatocyte and pancreatic islet-specific differentiation media. Cells were characterized by flow cytometry, gene-expression analysis, metabolic assays, and transplantation for their state of differentiation and tissue-specific functions. RESULTS In response to macrophage colony-stimulating factor and interleukin 3, monocytes resumed cell division in a CD115-dependent fashion, which was associated with a down-regulation of the PRDM1 and ICSBP genes. These programmable cells of monocytic origin were capable of differentiating into neohepatocytes, which closely resemble primary human hepatocytes with respect to morphology, expression of hepatocyte markers, and specific metabolic functions. After transplantation into the liver of severe combined immunodeficiency disease/nonobese diabetic mice, neohepatocytes integrated well into the liver tissue and showed a morphology and albumin expression similar to that of primary human hepatocytes transplanted under identical conditions. Programmable cells of monocytic origin-derived pancreatic neoislets expressed beta cell-specific transcription factors, secreted insulin and C peptide in a glucose-dependent manner, and normalized blood glucose levels when xenotransplanted into immunocompetent, streptozotocin-treated diabetic mice. Programmable cells of monocytic origin retained monocytic characteristics, notably CD14 expression, a monocyte-specific methylation pattern of the CD115 gene, and expression of the transcription factor PU.1. CONCLUSIONS The ability to reprogram, expand, and differentiate peripheral blood monocytes in large quantities opens the real possibility of the clinical application of programmable cells of monocytic origin in tissue repair and organ regeneration.
Collapse
Affiliation(s)
- Maren Ruhnke
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Flock G, Cao X, Drucker DJ. Pdx-1 is not sufficient for repression of proglucagon gene transcription in islet or enteroendocrine cells. Endocrinology 2005; 146:441-9. [PMID: 15471960 DOI: 10.1210/en.2004-0495] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pdx-1 plays a key role in the development of the pancreas and the control of islet gene transcription and has also been proposed as a dominant regulator of the alpha- vs. beta-cell phenotype via extinction of proglucagon expression. To ascertain the relationship between Pdx-1 and proglucagon gene expression, we examined the effect of enhanced pdx-1 expression on proglucagon gene expression in murine islet alphaTC-1 and GLUTag enteroendocrine cells. Although adenoviral transduction increased the levels of pdx-1 mRNA transcripts and nuclear Pdx-1 protein, overexpression of pdx-1 did not repress endogenous proglucagon gene expression in alphaTC-1 or GLUTag cells or murine islets. Immunohistochemical analysis of cells transduced with Ad-pdx-1 demonstrated multiple individual islet or enteroendocrine cells exhibiting both nuclear Pdx-1 and cytoplasmic glucagon-like peptide-1 immunopositivity. The failure of pdx-1 to inhibit endogenous proglucagon gene expression was not attributable to defects in Pdx-1 nuclear translocation or DNA binding as demonstrated using Western blotting and EMSA analyses. Furthermore, Ad-pdx-1 transduction did not repress proglucagon promoter activity in alphaTC-1 or GLUTag cells. Taken together, these findings demonstrate that pdx-1 alone is not sufficient for specification of the hormonal phenotype or extinction of proglucagon gene expression in islet or enteroendocrine cells.
Collapse
Affiliation(s)
- Grace Flock
- Department of Medicine, Toronto General Hospital, Banting and Best Diabetes Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | |
Collapse
|
24
|
Hirasawa A, Tsumaya K, Awaji T, Katsuma S, Adachi T, Yamada M, Sugimoto Y, Miyazaki S, Tsujimoto G. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med 2004; 11:90-4. [PMID: 15619630 DOI: 10.1038/nm1168] [Citation(s) in RCA: 1149] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2004] [Accepted: 11/22/2004] [Indexed: 12/11/2022]
Abstract
Diabetes, a disease in which the body does not produce or use insulin properly, is a serious global health problem. Gut polypeptides secreted in response to food intake, such as glucagon-like peptide-1 (GLP-1), are potent incretin hormones that enhance the glucose-dependent secretion of insulin from pancreatic beta cells. Free fatty acids (FFAs) provide an important energy source and also act as signaling molecules in various cellular processes, including the secretion of gut incretin peptides. Here we show that a G-protein-coupled receptor, GPR120, which is abundantly expressed in intestine, functions as a receptor for unsaturated long-chain FFAs. Furthermore, we show that the stimulation of GPR120 by FFAs promotes the secretion of GLP-1 in vitro and in vivo, and increases circulating insulin. Because GLP-1 is the most potent insulinotropic incretin, our results indicate that GPR120-mediated GLP-1 secretion induced by dietary FFAs is important in the treatment of diabetes.
Collapse
Affiliation(s)
- Akira Hirasawa
- Department of Molecular, Cell Pharmacology, National Research Institute for Child Health and Development, 3-35-31, Taishi-do, Setagaya-ku, Tokyo 154-8567, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cani PD, Dewever C, Delzenne NM. Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats. Br J Nutr 2004; 92:521-6. [PMID: 15469657 DOI: 10.1079/bjn20041225] [Citation(s) in RCA: 299] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The hypothesis tested in the present study is that dietary fructans are able to modulate gastrointestinal peptides involved in the control of food intake, namely glucagon-like peptide (GLP)-1 (7-36) amide and ghrelin. After 3 weeks of treatment with a standard diet (control) or the same diet enriched with 100 g fructans varying in their degrees of polymerization (oligofructose (OFS), Synergy 1 (Syn) or long chain inulin)/kg, male Wistar rats were deprived of food for 8 h before sample collection. Dietary energy intake throughout the experiment was significantly lower (P<0.05) in fructans-fed rats than in control rats, leading to a significant decrease (P<0.01) in epidydimal fat mass at the end of the treatment in OFS- and Syn-treated rats. GLP-1 (7-36) amide concentration in portal vein serum was higher in OFS- and Syn-fed than in control rats. Both GLP-1 (7-36) amide concentration and proglucagon mRNA concentrations were significantly greater (P<0.05) in the proximal colonic mucosa of fructans-fed rats v. controls. Normally active ghrelin concentration in plasma increases during food deprivation and rapidly falls during a meal. In the present study, after 8 h of food deprivation, active ghrelin in the plasma remained significantly lower (P<0.05) in OFS and Syn-fed than in control rats. These results are in accordance with the modifications of dietary intake and fat-mass development in short-chain fructans-treated rats and demonstrate the potential modulation of GLP-1 (7-36) amide and ghrelin by fermentable fibres such as fructans, which are rapidly and extensively fermented in the proximal part of the colon.
Collapse
Affiliation(s)
- Patrice D Cani
- Unit of Pharmacokinetics, Metabolism, Nutrition and Toxicology, Department of Pharmaceutical Sciences, Université Catholique de Louvain, B-1200 Brussels, Belgium
| | | | | |
Collapse
|