1
|
Agawa S, Futagami S, Yamawaki H, Tsushima R, Higuchi K, Habiro M, Kawawa R, Kodaka Y, Ueki N, Watanabe Y, Gudis K, Ohashi R, Iwakiri K. Trypsin may be associated with duodenal eosinophils through the expression of PAR2 in early chronic pancreatitis and functional dyspepsia with pancreatic enzyme abnormalities. PLoS One 2022; 17:e0275341. [PMID: 36264979 PMCID: PMC9584419 DOI: 10.1371/journal.pone.0275341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 09/13/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Early chronic pancreatitis (ECP) has been reported to advance into chronic pancreatitis, it may be critical to differentiate the pathophysiology of ECP and functional dyspepsia (FD) in patients with pancreatic enzyme abnormalities (FD-P). This study aimed to clarify differences in the pathophysiology of ECP and FD-P and to determine whether duodenal inflammatory responses in the two diseases were associated with protease-activated receptor (PAR) 2, as the trypsin receptor. METHODS Eighty patients who presented with FD-P and ECP were enrolled. In duodenal specimens, PAR2 mRNA levels were determined using real-time PCR. Using immunostaining, CD68-, GLP-1-, PRG2-, and CCR2-positive cells, tight junction proteins, and PAR 2 were evaluated. RESULTS There were no significant differences in clinical symptoms and gastric motility between ECP and FD-P patients. The CD68-positive cells infiltrations and occludin expression levels in the duodenal mucosa of patients with FD-P were significantly (p<0.001 and p = 0.048, respectively) lower than those in patients with ECP. Although serum trypsin levels in ECP and FD-P patents were significantly (p<0.05 and p<0.001, respectively) associated with duodenal eosinophils counts, elevated trypsin levels were not significantly associated with degranulated eosinophils, occludin, claudin-1 and ZO-1 expression levels in the duodenum of either group. PAR2 mRNA levels were increased in the duodenum of patients with ECP and FD-P. PAR2 was localized in the epithelial cells of the duodenal mucosa and the surface of degranulated eosinophils in ECP and FD-P patients. CONCLUSIONS Elevated trypsin levels might be partly associated with duodenal inflammatory responses through PAR2-related degranulated eosinophils and the reduction of occludin in patients with ECP and FD-P.
Collapse
Affiliation(s)
- Shuhei Agawa
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Seiji Futagami
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
- * E-mail:
| | - Hiroshi Yamawaki
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Rina Tsushima
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | | | - Mayu Habiro
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Rie Kawawa
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Yasuhiro Kodaka
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Nobue Ueki
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Yoshiyuki Watanabe
- Department of Internal Medicine, Kawasaki Rinko General Hospital, Kawasaki, Japan
| | - Katya Gudis
- Department of Gastroenterology, Nippon Medical School, Tokyo, Japan
| | - Rhuji Ohashi
- Department of Diagnostic Pathology, Nippon Medical School, Tokyo, Japan
| | | |
Collapse
|
2
|
Mobbs JI, Belousoff MJ, Harikumar KG, Piper SJ, Xu X, Furness SGB, Venugopal H, Christopoulos A, Danev R, Wootten D, Thal DM, Miller LJ, Sexton PM. Structures of the human cholecystokinin 1 (CCK1) receptor bound to Gs and Gq mimetic proteins provide insight into mechanisms of G protein selectivity. PLoS Biol 2021; 19:e3001295. [PMID: 34086670 PMCID: PMC8208569 DOI: 10.1371/journal.pbio.3001295] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/16/2021] [Accepted: 05/19/2021] [Indexed: 01/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are critical regulators of cellular function acting via heterotrimeric G proteins as their primary transducers with individual GPCRs capable of pleiotropic coupling to multiple G proteins. Structural features governing G protein selectivity and promiscuity are currently unclear. Here, we used cryo-electron microscopy (cryo-EM) to determine structures of the cholecystokinin (CCK) type 1 receptor (CCK1R) bound to the CCK peptide agonist, CCK-8 and 2 distinct transducer proteins, its primary transducer Gq, and the more weakly coupled Gs. As seen with other Gq/11-GPCR complexes, the Gq-α5 helix (αH5) bound to a relatively narrow pocket in the CCK1R core. Surprisingly, the backbone of the CCK1R and volume of the G protein binding pocket were essentially equivalent when Gs was bound, with the Gs αH5 displaying a conformation that arises from "unwinding" of the far carboxyl-terminal residues, compared to canonically Gs coupled receptors. Thus, integrated changes in the conformations of both the receptor and G protein are likely to play critical roles in the promiscuous coupling of individual GPCRs.
Collapse
MESH Headings
- Cholecystokinin/metabolism
- Cholesterol/metabolism
- GTP-Binding Protein alpha Subunits, Gq-G11/chemistry
- GTP-Binding Protein alpha Subunits, Gq-G11/metabolism
- GTP-Binding Protein alpha Subunits, Gq-G11/ultrastructure
- GTP-Binding Protein alpha Subunits, Gs/chemistry
- GTP-Binding Protein alpha Subunits, Gs/metabolism
- GTP-Binding Protein alpha Subunits, Gs/ultrastructure
- HEK293 Cells
- Humans
- Models, Molecular
- Protein Binding
- Receptors, Cholecystokinin/chemistry
- Receptors, Cholecystokinin/metabolism
- Receptors, Cholecystokinin/ultrastructure
- Signal Transduction
Collapse
Affiliation(s)
- Jesse I. Mobbs
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Matthew J. Belousoff
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Kaleeckal G. Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Sarah J. Piper
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Sebastian G. B. Furness
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Hari Venugopal
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Victoria, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Radostin Danev
- Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Denise Wootten
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - David M. Thal
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Laurence J. Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Patrick M. Sexton
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- ARC Centre for Cryo-Electron Microscopy of Membrane Proteins, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
3
|
Association of Gut Hormones and Microbiota with Vascular Dysfunction in Obesity. Nutrients 2021; 13:nu13020613. [PMID: 33668627 PMCID: PMC7918888 DOI: 10.3390/nu13020613] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 02/08/2023] Open
Abstract
In the past few decades, obesity has reached pandemic proportions. Obesity is among the main risk factors for cardiovascular diseases, since chronic fat accumulation leads to dysfunction in vascular endothelium and to a precocious arterial stiffness. So far, not all the mechanisms linking adipose tissue and vascular reactivity have been explained. Recently, novel findings reported interesting pathological link between endothelial dysfunction with gut hormones and gut microbiota and energy homeostasis. These findings suggest an active role of gut secretome in regulating the mediators of vascular function, such as nitric oxide (NO) and endothelin-1 (ET-1) that need to be further investigated. Moreover, a central role of brain has been suggested as a main player in the regulation of the different factors and hormones beyond these complex mechanisms. The aim of the present review is to discuss the state of the art in this field, by focusing on the processes leading to endothelial dysfunction mediated by obesity and metabolic diseases, such as insulin resistance. The role of perivascular adipose tissue (PVAT), gut hormones, gut microbiota dysbiosis, and the CNS function in controlling satiety have been considered. Further understanding the crosstalk between these complex mechanisms will allow us to better design novel strategies for the prevention of obesity and its complications.
Collapse
|
4
|
Mhalhal TR, Washington MC, Heath JC, Sayegh AI. Effect of Vagotomy and Sympathectomy on the Feeding Responses Evoked by Intra-Aortic Cholecystokinin-8 in Adult Male Sprague Dawley Rats. Endocr Res 2021; 46:57-65. [PMID: 33426974 DOI: 10.1080/07435800.2020.1861621] [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] [Indexed: 10/22/2022]
Abstract
The vagus nerve and the celiaco-mesenteric ganglia (CMG) are required for reduction of meal size (MS) and prolongation of the intermeal interval (IMI) by intraperitoneal (ip) sulfated cholecystokinin-8 (CCK-8). However, recently we have shown that the gut regulates these responses. Therefore, reevaluating the role of the vagus and the CMG in the feeding responses evoked by CCK is necessary because the gut contains the highest concentration of enteric, vagal and splanchnic afferents and CCK-A receptors, which are required for reduction of food intake by this peptide, compared to other abdominal organs. To address this necessity, we injected sulfated CCK-8 (0, 0.1, 0.5, 1 and 3 nmol/kg) in the aorta, near the gastrointestinal sites of action of the peptide, in three groups of free-feeding rats (n = 10 rats per group), subdiaphragmatic vagotomy (VGX), celiaco-mesenteric ganglionectomy (CMGX) and sham-operated, and recorded seven feeding responses. In the sham group, CCK-8 reduced MS (normal chow), prolonged the intermeal interval (IMI, time between first and second meals), increased satiety ratio (SR, IMI/MS), shortened duration of first meal, reduced total (24 hrs) food intake and reduced number of meals relative to saline vehicle. Vagotomy attenuated all of the previous responses except IMI length and SR, and CMGX attenuated all of those responses. In conclusion, the feeding responses evoked by sulfated CCK-8 require, independently, the vagus nerve and the CMG.
Collapse
Affiliation(s)
- Thaer R Mhalhal
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
- Department of Anatomy and Histology, College of Veterinary Medicine, University of Basrah, Basrah, Iraq
| | - Martha C Washington
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - John C Heath
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| | - Ayman I Sayegh
- Gastroenterology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, 36088, USA
| |
Collapse
|
5
|
Martin AM, Sun EW, Rogers GB, Keating DJ. The Influence of the Gut Microbiome on Host Metabolism Through the Regulation of Gut Hormone Release. Front Physiol 2019; 10:428. [PMID: 31057420 PMCID: PMC6477058 DOI: 10.3389/fphys.2019.00428] [Citation(s) in RCA: 233] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/27/2019] [Indexed: 12/17/2022] Open
Abstract
The microbial community of the gut conveys significant benefits to host physiology. A clear relationship has now been established between gut bacteria and host metabolism in which microbial-mediated gut hormone release plays an important role. Within the gut lumen, bacteria produce a number of metabolites and contain structural components that act as signaling molecules to a number of cell types within the mucosa. Enteroendocrine cells within the mucosal lining of the gut synthesize and secrete a number of hormones including CCK, PYY, GLP-1, GIP, and 5-HT, which have regulatory roles in key metabolic processes such as insulin sensitivity, glucose tolerance, fat storage, and appetite. Release of these hormones can be influenced by the presence of bacteria and their metabolites within the gut and as such, microbial-mediated gut hormone release is an important component of microbial regulation of host metabolism. Dietary or pharmacological interventions which alter the gut microbiome therefore pose as potential therapeutics for the treatment of human metabolic disorders. This review aims to describe the complex interaction between intestinal microbiota and their metabolites and gut enteroendocrine cells, and highlight how the gut microbiome can influence host metabolism through the regulation of gut hormone release.
Collapse
Affiliation(s)
- Alyce M Martin
- Molecular and Cellular Physiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Emily W Sun
- Molecular and Cellular Physiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Geraint B Rogers
- Microbiome Research Laboratory, Flinders University, Adelaide, SA, Australia.,Infection and Immunity, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Damien J Keating
- Molecular and Cellular Physiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia.,Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
6
|
Desai AJ, Mechin I, Nagarajan K, Valant C, Wootten D, Lam PCH, Orry A, Abagyan R, Nair A, Sexton PM, Christopoulos A, Miller LJ. Molecular Basis of Action of a Small-Molecule Positive Allosteric Modulator Agonist at the Type 1 Cholecystokinin Holoreceptor. Mol Pharmacol 2019; 95:245-259. [PMID: 30591538 DOI: 10.1124/mol.118.114082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Accepted: 12/19/2018] [Indexed: 02/14/2025] Open
Abstract
Allosteric modulation of receptors provides mechanistic safety while effectively achieving biologic endpoints otherwise difficult or impossible to obtain by other means. The theoretical case has been made for the development of a positive allosteric modulator (PAM) of the type 1 cholecystokinin receptor (CCK1R) having minimal intrinsic agonist activity to enhance meal-induced satiety for the treatment of obesity, while reducing the risk of side effects and/or toxicity. Unfortunately, such a drug does not currently exist. In this work, we have identified a PAM agonist of the CCK1R, SR146131, and determined its putative binding mode and receptor activation mechanism by combining molecular modeling, chimeric CCK1R/CCK2R constructs, and site-directed mutagenesis. We probed the structure-activity relationship of analogs of SR146131 for impact on agonism versus cooperativity of the analogs. This identified structural features that might be responsible for binding affinity and potency while retaining PAM activity. SR146131 and several of its analogs were docked into the receptor structure, which had the natural endogenous peptide agonist, cholecystokinin, already in the bound state (by docking), providing a refined structural model of the intact CCK1R holoreceptor. Both SR146131 and its analogs exhibited unique probe-dependent cooperativity with orthosteric peptide agonists and were simultaneously accommodated in this model, consistent with the derived structure-activity relationships. This provides improved understanding of the molecular basis for CCK1R-directed drug development.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Ingrid Mechin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Karthigeyan Nagarajan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Celine Valant
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Denise Wootten
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Polo C H Lam
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Andrew Orry
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Ruben Abagyan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Anil Nair
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Patrick M Sexton
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Arthur Christopoulos
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona (A.J.D., L.J.M.); In Silico Drug Discovery Department, Icagen Tucson Innovation Center, Oro Valley, Arizona (I.M., K.N., A.N.); Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Australia (C.V., D.W., P.M.S., A.C., L.J.M.); Molsoft, La Jolla, California (P.C.H.L., A.O., R.A.); Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California (R.A.); and School of Pharmacy, Fudan University, Shanghai, China (D.W., P.M.S.)
| |
Collapse
|
7
|
Martin AM, Sun EW, Rogers GB, Keating DJ. The Influence of the Gut Microbiome on Host Metabolism Through the Regulation of Gut Hormone Release. Front Physiol 2019. [PMID: 31057420 DOI: 10.3389/fphys.2019.00428/bibtex] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
The microbial community of the gut conveys significant benefits to host physiology. A clear relationship has now been established between gut bacteria and host metabolism in which microbial-mediated gut hormone release plays an important role. Within the gut lumen, bacteria produce a number of metabolites and contain structural components that act as signaling molecules to a number of cell types within the mucosa. Enteroendocrine cells within the mucosal lining of the gut synthesize and secrete a number of hormones including CCK, PYY, GLP-1, GIP, and 5-HT, which have regulatory roles in key metabolic processes such as insulin sensitivity, glucose tolerance, fat storage, and appetite. Release of these hormones can be influenced by the presence of bacteria and their metabolites within the gut and as such, microbial-mediated gut hormone release is an important component of microbial regulation of host metabolism. Dietary or pharmacological interventions which alter the gut microbiome therefore pose as potential therapeutics for the treatment of human metabolic disorders. This review aims to describe the complex interaction between intestinal microbiota and their metabolites and gut enteroendocrine cells, and highlight how the gut microbiome can influence host metabolism through the regulation of gut hormone release.
Collapse
Affiliation(s)
- Alyce M Martin
- Molecular and Cellular Physiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Emily W Sun
- Molecular and Cellular Physiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Geraint B Rogers
- Microbiome Research Laboratory, Flinders University, Adelaide, SA, Australia
- Infection and Immunity, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Damien J Keating
- Molecular and Cellular Physiology Laboratory, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| |
Collapse
|
8
|
Mussa BM, Sood S, Verberne AJM. Implication of neurohormonal-coupled mechanisms of gastric emptying and pancreatic secretory function in diabetic gastroparesis. World J Gastroenterol 2018; 24:3821-3833. [PMID: 30228777 PMCID: PMC6141338 DOI: 10.3748/wjg.v24.i34.3821] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/22/2018] [Accepted: 06/27/2018] [Indexed: 02/06/2023] Open
Abstract
Recently, diabetic gastroparesis (DGP) has received much attention as its prevalence is increasing in a dramatic fashion and management of patients with DGP represents a challenge in the clinical practice due to the limited therapeutic options. DGP highlights an interrelationship between the gastric emptying and pancreatic secretory function that regulate a wide range of digestive and metabolic functions, respectively. It well documented that both gastric emptying and pancreatic secretion are under delicate control by multiple neurohormonal mechanisms including extrinsic parasympathetic pathways and gastrointestinal (GI) hormones. Interestingly, the latter released in response to various determinants that related to the rate and quality of gastric emptying. Others and we have provided strong evidence that the central autonomic nuclei send a dual output (excitatory and inhibitory) to the stomach and the pancreas in response to a variety of hormonal signals from the abdominal viscera. Most of these hormones released upon gastric emptying to provide feedback, and control this process and simultaneously regulate pancreatic secretion and postprandial glycemia. These findings emphasize an important link between gastric emptying and pancreatic secretion and its role in maintaining homeostatic processes within the GI tract. The present review deals with the neurohormonal-coupled mechanisms of gastric emptying and pancreatic secretory function that implicated in DGP and this provides new insights in our understanding of the pathophysiology of DGP. This also enhances the process of identifying potential therapeutic targets to treat DGP and limit the complications of current management practices.
Collapse
Affiliation(s)
- Bashair M Mussa
- Department of Basic Medical Science, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Sanjay Sood
- Department of Basic Medical Science, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Anthony JM Verberne
- Department of Medicine, Austin Health, University of Melbourne, Melbourne 3084, Australia
| |
Collapse
|
9
|
Fukui H, Xu X, Miwa H. Role of Gut Microbiota-Gut Hormone Axis in the Pathophysiology of Functional Gastrointestinal Disorders. J Neurogastroenterol Motil 2018; 24:367-386. [PMID: 29969855 PMCID: PMC6034676 DOI: 10.5056/jnm18071] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/21/2018] [Indexed: 12/13/2022] Open
Abstract
Gut microbiota exert a pivotal influence on various functions including gastrointestinal (GI) motility, metabolism, nutrition, immunity, and the neuroendocrine system in the host. These effects are mediated by not only short-chain fatty acids produced by microbiota but also gut hormones and inflammatory signaling by enteroendocrine and immune cells under the influence of the microbiota. GI motility is orchestrated by the enteric nervous system and hormonal networks, and disturbance of GI motility plays an important role in the pathophysiology of functional gastrointestinal disorders (FGIDs). In this context, microbiota-associated mediators are considered to act on specific receptors, thus affecting the enteric nervous system and, subsequently, GI motility. Thus, the pathophysiology of FGIDs is based on alterations of the gut microbiota/gut hormone axis, which have crucial effects on GI motility.
Collapse
Affiliation(s)
- Hirokazu Fukui
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Mukogawa, Nishinomiya,
Japan
| | - Xin Xu
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Mukogawa, Nishinomiya,
Japan
- Department of Digestive Diseases, Tianjin Medical University General Hospital, Tianjin,
China
| | - Hiroto Miwa
- Division of Gastroenterology, Department of Internal Medicine, Hyogo College of Medicine, Mukogawa, Nishinomiya,
Japan
| |
Collapse
|
10
|
Guarino D, Nannipieri M, Iervasi G, Taddei S, Bruno RM. The Role of the Autonomic Nervous System in the Pathophysiology of Obesity. Front Physiol 2017; 8:665. [PMID: 28966594 PMCID: PMC5606212 DOI: 10.3389/fphys.2017.00665] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/22/2017] [Indexed: 12/18/2022] Open
Abstract
Obesity is reaching epidemic proportions globally and represents a major cause of comorbidities, mostly related to cardiovascular disease. The autonomic nervous system (ANS) dysfunction has a two-way relationship with obesity. Indeed, alterations of the ANS might be involved in the pathogenesis of obesity, acting on different pathways. On the other hand, the excess weight induces ANS dysfunction, which may be involved in the haemodynamic and metabolic alterations that increase the cardiovascular risk of obese individuals, i.e., hypertension, insulin resistance and dyslipidemia. This article will review current evidence about the role of the ANS in short-term and long-term regulation of energy homeostasis. Furthermore, an increased sympathetic activity has been demonstrated in obese patients, particularly in the muscle vasculature and in the kidneys, possibily contributing to increased cardiovascular risk. Selective leptin resistance, obstructive sleep apnea syndrome, hyperinsulinemia and low ghrelin levels are possible mechanisms underlying sympathetic activation in obesity. Weight loss is able to reverse metabolic and autonomic alterations associated with obesity. Given the crucial role of autonomic dysfunction in the pathophysiology of obesity and its cardiovascular complications, vagal nerve modulation and sympathetic inhibition may serve as therapeutic targets in this condition.
Collapse
Affiliation(s)
- Daniela Guarino
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy.,Institute of Clinical Physiology of CNRPisa, Italy.,Scuola Superiore Sant'AnnaPisa, Italy
| | - Monica Nannipieri
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | | | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Rosa Maria Bruno
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
11
|
Desai AJ, Miller LJ. Changes in the plasma membrane in metabolic disease: impact of the membrane environment on G protein-coupled receptor structure and function. Br J Pharmacol 2017; 175:4009-4025. [PMID: 28691227 DOI: 10.1111/bph.13943] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/08/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022] Open
Abstract
Drug development targeting GPCRs often utilizes model heterologous cell expression systems, reflecting an implicit assumption that the membrane environment has little functional impact on these receptors or on their responsiveness to drugs. However, much recent data have illustrated that membrane components can have an important functional impact on intrinsic membrane proteins. This review is directed toward gaining a better understanding of the structure of the plasma membrane in health and disease, and how this organelle can influence GPCR structure, function and regulation. It is important to recognize that the membrane provides a potential mode of lateral allosteric regulation of GPCRs and can affect the effectiveness of drugs and their biological responses in various disease states, which can even vary among individuals across the population. The type 1 cholecystokinin receptor is reviewed as an exemplar of a class A GPCR that is affected in this way by changes in the plasma membrane. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
12
|
Desai AJ, Dong M, Langlais BT, Dueck AC, Miller LJ. Cholecystokinin responsiveness varies across the population dependent on metabolic phenotype. Am J Clin Nutr 2017; 106:447-456. [PMID: 28592602 PMCID: PMC5525122 DOI: 10.3945/ajcn.117.156943] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/05/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Cholecystokinin (CCK) is an important satiety factor, acting at type 1 receptors (CCK1Rs) on vagal afferent neurons; however, CCK agonists have failed clinical trials for obesity. We postulated that CCK1R function might be defective in such patients due to abnormal membrane composition, such as that observed in cholesterol gallstone disease.Objective: Due to the challenges in directly studying CCK1Rs relevant to appetite control, our goal was to develop and apply a method to determine the impact of a patient's own cellular environment on CCK stimulus-activity coupling and to determine whether CCK sensitivity correlated with the metabolic phenotype of a high-risk population.Design: Wild-type CCK1Rs were expressed on leukocytes from 112 Hispanic patients by using adenoviral transduction and 24-h culture, with quantitation of cholesterol composition and intracellular calcium responses to CCK. Results were correlated with clinical, biochemical, and morphometric characteristics.Results: Broad ranges of cellular cholesterol and CCK responsiveness were observed, with elevated cholesterol correlated with reduced CCK sensitivity. This was prominent with increasing degrees of obesity and the presence of diabetes, particularly when poorly controlled. No single standard clinical metric correlated directly with CCK responsiveness. Reduced CCK sensitivity best correlated with elevated serum triglycerides in normal-weight participants and with low HDL concentrations and elevated glycated hemoglobin in obese and diabetic patients.Conclusions: CCK responsiveness varies widely across the population, with reduced signaling in patients with obesity and diabetes. This could explain the failure of CCK agonists in previous clinical trials and supports the rationale to develop corrective modulators to reverse this defective servomechanism for appetite control. This trial was registered at www.clinicaltrials.gov as NCT03121755.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics and
| | - Maoqing Dong
- Department of Molecular Pharmacology and Experimental Therapeutics and
| | | | | | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics and
| |
Collapse
|
13
|
Martin AM, Young RL, Leong L, Rogers GB, Spencer NJ, Jessup CF, Keating DJ. The Diverse Metabolic Roles of Peripheral Serotonin. Endocrinology 2017; 158:1049-1063. [PMID: 28323941 DOI: 10.1210/en.2016-1839] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 02/23/2017] [Indexed: 02/07/2023]
Abstract
Serotonin (5-hydroxytryptamine or 5-HT) is a multifunctional bioamine with important signaling roles in a range of physiological pathways. Almost all of the 5-HT in our bodies is synthesized in specialized enteroendocrine cells within the gastrointestinal (GI) mucosa called enterochromaffin (EC) cells. These cells provide all of our circulating 5-HT. We have long appreciated the important contributions of 5-HT within the gut, including its role in modulating GI motility. However, evidence of the physiological and clinical significance of gut-derived 5-HT outside of the gut has recently emerged, implicating 5-HT in regulation of glucose homeostasis, lipid metabolism, bone density, and diseases associated with metabolic syndrome, such as obesity and type 2 diabetes. Although a new picture has developed in the last decade regarding the various metabolic roles of peripheral serotonin, so too has our understanding of the physiology of EC cells. Given that they are scattered throughout the lining of the GI tract within the epithelial cell layer, these cells are typically difficult to study. Advances in isolation procedures now allow the study of pure EC-cell cultures and single cells, enabling studies of EC-cell physiology to occur. EC cells are sensory cells that are capable of integrating cues from ingested nutrients, the enteric nervous system, and the gut microbiome. Thus, levels of peripheral 5-HT can be modulated by a multitude of factors, resulting in both local and systemic effects for the regulation of a raft of physiological pathways related to metabolism and obesity.
Collapse
Affiliation(s)
- Alyce M Martin
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide 5042, Australia
| | - Richard L Young
- Nutrition and Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5001, Australia
- Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
| | - Lex Leong
- Infection and Immunity, SAHMRI, Adelaide 5001, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University of South Australia, Adelaide 5042, Australia
| | - Geraint B Rogers
- Infection and Immunity, SAHMRI, Adelaide 5001, Australia
- SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University of South Australia, Adelaide 5042, Australia
| | - Nick J Spencer
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide 5042, Australia
| | - Claire F Jessup
- Adelaide Medical School, University of Adelaide, Adelaide 5005, Australia
- Discipline of Anatomy and Histology, Flinders University of South Australia, Adelaide 5042, Australia
| | - Damien J Keating
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University of South Australia, Adelaide 5042, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute (SAHMRI), Adelaide 5001, Australia
| |
Collapse
|
14
|
Grabauskas G, Owyang C. Plasticity of vagal afferent signaling in the gut. MEDICINA-LITHUANIA 2017; 53:73-84. [PMID: 28454890 PMCID: PMC6318799 DOI: 10.1016/j.medici.2017.03.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 03/21/2017] [Indexed: 12/13/2022]
Abstract
Vagal sensory neurons mediate the vago-vagal reflex which, in turn, regulates a wide array of gastrointestinal functions including esophageal motility, gastric accommodation and pancreatic enzyme secretion. These neurons also transmit sensory information from the gut to the central nervous system, which then mediates the sensations of nausea, fullness and satiety. Recent research indicates that vagal afferent neurons process non-uniform properties and a significant degree of plasticity. These properties are important to ensure that vagally regulated gastrointestinal functions respond rapidly and appropriately to various intrinsic and extrinsic factors. Similar plastic changes in the vagus also occur in pathophysiological conditions, such as obesity and diabetes, resulting in abnormal gastrointestinal functions. A clear understanding of the mechanisms which mediate these events may provide novel therapeutic targets for the treatment of gastrointestinal disorders due to vago-vagal pathway malfunctions.
Collapse
Affiliation(s)
- Gintautas Grabauskas
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48019, USA.
| | - Chung Owyang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48019, USA
| |
Collapse
|
15
|
Desai AJ, Dong M, Harikumar KG, Miller LJ. Cholecystokinin-induced satiety, a key gut servomechanism that is affected by the membrane microenvironment of this receptor. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2016; 6:S22-S27. [PMID: 28685026 DOI: 10.1038/ijosup.2016.5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The gastrointestinal (GI) tract has a central role in nutritional homeostasis, as location for food ingestion, digestion and absorption, with the gut endocrine system responding to and regulating these events, as well as influencing appetite. One key GI hormone with the full spectrum of these activities is cholecystokinin (CCK), a peptide released from neuroendocrine I cells scattered through the proximal intestine in response to fat and protein, with effects to stimulate gall bladder contraction and pancreatic exocrine secretion, to regulate gastric emptying and intestinal transit, and to induce satiety. There has been interest in targeting the type 1 CCK receptor (CCK1R) for drug development to provide non-caloric satiation as an aid to dieting and weight loss; however, there have been concerns about CCK1R agonists related to side effects and potential trophic impact on the pancreas. A positive allosteric modulator (PAM) of CCK action at this receptor without intrinsic agonist activity could provide a safer and more effective approach to long-term administration. In addition, CCK1R stimulus-activity coupling has been shown to be negatively affected by excess membrane cholesterol, a condition described in the metabolic syndrome, thereby potentially interfering with an important servomechanism regulating appetite. A PAM targeting this receptor could also potentially correct the negative impact of cholesterol on CCK1R function. We will review the molecular basis for binding natural peptide agonist, binding and action of small molecules within the allosteric pocket, and the impact of cholesterol. Novel strategies for taking advantage of this receptor for the prevention and management of obesity will be reviewed.
Collapse
Affiliation(s)
- A J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - M Dong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - K G Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| | - L J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
16
|
Abstract
A large body of research has been dedicated to the effects of gastrointestinal peptides on vagal afferent fibres, yet multiple lines of evidence indicate that gastrointestinal peptides also modulate brainstem vagal neurocircuitry, and that this modulation has a fundamental role in the physiology and pathophysiology of the upper gastrointestinal tract. In fact, brainstem vagovagal neurocircuits comprise highly plastic neurons and synapses connecting afferent vagal fibres, second order neurons of the nucleus tractus solitarius (NTS), and efferent fibres originating in the dorsal motor nucleus of the vagus (DMV). Neuronal communication between the NTS and DMV is regulated by the presence of a variety of inputs, both from within the brainstem itself as well as from higher centres, which utilize an array of neurotransmitters and neuromodulators. Because of the circumventricular nature of these brainstem areas, circulating hormones can also modulate the vagal output to the upper gastrointestinal tract. This Review summarizes the organization and function of vagovagal reflex control of the upper gastrointestinal tract, presents data on the plasticity within these neurocircuits after stress, and discusses the gastrointestinal dysfunctions observed in Parkinson disease as examples of physiological adjustment and maladaptation of these reflexes.
Collapse
|
17
|
Desai AJ, Dong M, Miller LJ. Beneficial effects of β-sitosterol on type 1 cholecystokinin receptor dysfunction induced by elevated membrane cholesterol. Clin Nutr 2016; 35:1374-1379. [PMID: 27016394 DOI: 10.1016/j.clnu.2016.03.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 02/09/2016] [Accepted: 03/07/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS The type 1 cholecystokinin receptor (CCK1R) mediates the actions of CCK to support nutritional homeostasis, including post-cibal satiety. However, elevated levels of membrane cholesterol, such as have been observed in metabolic syndrome, interfere with CCK stimulus-activity coupling at the CCK1R, thereby disrupting this important servomechanism. We hypothesize that reversal of the negative impact of cholesterol on this receptor could be useful in the management of obesity. METHODS We have studied the effects of β-sitosterol, a phytosterol structurally related to cholesterol, on CCK receptor function. This included CCK binding and biological activity at wild type CCK1R and CCK2R, as well as at CCK1R in a high cholesterol environment, and at a CCK1R mutant, Y140A, which mimics the behavior of wild type receptor in high cholesterol. RESULTS β-sitosterol (100 μM and 10 μM) significantly improved the defective signaling of the CCK1R present in high cholesterol (p < 0.05), without affecting CCK binding affinity. This effect was absent at the CCK1R present in a normal cholesterol environment, as well as at the structurally-related CCK2R. Furthermore, the cholesterol-insensitive Y140A mutant of CCK1R was resistant to the effects of β-sitosterol. CONCLUSION These data suggest that β-sitosterol affects CCK1R function in high cholesterol by competing with cholesterol at a receptor cholesterol-binding site and may shift its conformation toward normal. This phytosterol extends our understanding of the structure-activity relationships for developing a drug that can target the external surface of CCK1R. Since the concentrations of β-sitosterol shown to be effective in this study are similar to serum levels of this compound achievable during oral administration, it may be worthwhile to study possible beneficial effects of β-sitosterol in metabolic syndrome.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Maoqing Dong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, AZ 85259, USA.
| |
Collapse
|
18
|
Desai AJ, Lam PCH, Orry A, Abagyan R, Christopoulos A, Sexton PM, Miller LJ. Molecular Mechanism of Action of Triazolobenzodiazepinone Agonists of the Type 1 Cholecystokinin Receptor. Possible Cooperativity across the Receptor Homodimeric Complex. J Med Chem 2015; 58:9562-77. [PMID: 26654202 DOI: 10.1021/acs.jmedchem.5b01110] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The type 1 cholecystokinin receptor (CCK1R) has multiple physiologic roles relating to nutrient homeostasis, including mediation of postcibal satiety. This effect has been central in efforts to develop agonists of this receptor as part of a program to manage and/or prevent obesity. While a number of small molecule CCK1R agonists have been developed, none have yet been approved for clinical use, based on inadequate efficacy, side effects, or the potential for toxicity. Understanding the molecular details of docking and mechanism of action of these ligands can be helpful in the rational refinement and enhancement of small molecule drug candidates. In the current work, we have defined the mechanism of binding and activity of two triazolobenzodiazepinones, CE-326597 and PF-04756956, which are reported to be full agonist ligands. To achieve this, we utilized receptor binding with a series of allosteric and orthosteric radioligands at structurally related CCK1R and CCK2R, as well as chimeric CCK1R/CCK2R constructs exchanging residues in the allosteric pocket, and assessment of biological activity. These triazolobenzodiazepinones docked within the intramembranous small molecule allosteric ligand pocket, with higher affinity binding to CCK2R than CCK1R, yet with biological activity exclusive to or greatly enhanced at CCK1R. These ligands exhibited cooperativity with benzodiazepine binding across the CCK1R homodimeric complex, resulting in their ability to inhibit only a fraction of the saturable binding of a benzodiazepine radioligand, unlike other small molecule antagonists and agonists of this receptor. This may contribute to the understanding of the unique short duration and reversible gallbladder contraction observed in vivo upon administration of these drugs.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic , Scottsdale, Arizona 85259, United States
| | - Polo C H Lam
- Molsoft LLC , La Jolla, California 92037, United States
| | - Andrew Orry
- Molsoft LLC , La Jolla, California 92037, United States
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego , La Jolla, California 92037, United States
| | - Arthur Christopoulos
- Department of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University , Parkville, Victoria 3052, Australia
| | - Patrick M Sexton
- Department of Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University , Parkville, Victoria 3052, Australia
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic , Scottsdale, Arizona 85259, United States
| |
Collapse
|
19
|
Desai AJ, Dong M, Harikumar KG, Miller LJ. Impact of ursodeoxycholic acid on a CCK1R cholesterol-binding site may contribute to its positive effects in digestive function. Am J Physiol Gastrointest Liver Physiol 2015; 309:G377-86. [PMID: 26138469 PMCID: PMC4556949 DOI: 10.1152/ajpgi.00173.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/26/2015] [Indexed: 01/31/2023]
Abstract
Dysfunction of the type 1 cholecystokinin (CCK) receptor (CCK1R) as a result of increased gallbladder muscularis membrane cholesterol has been implicated in the pathogenesis of cholesterol gallstones. Administration of ursodeoxycholic acid, which is structurally related to cholesterol, has been shown to have beneficial effects on gallstone formation. Our aims were to explore the possible direct effects and mechanism of action of bile acids on CCK receptor function. We studied the effects of structurally related hydrophobic chenodeoxycholic acid and hydrophilic ursodeoxycholic acid in vitro on CCK receptor function in the setting of normal and elevated membrane cholesterol. We also examined their effects on a cholesterol-insensitive CCK1R mutant (Y140A) disrupting a key site of cholesterol action. The results show that, similar to the impact of cholesterol on CCK receptors, bile acid effects were limited to CCK1R, with no effects on CCK2R. Chenodeoxycholic acid had a negative impact on CCK1R function, while ursodeoxycholic acid had no effect on CCK1R function in normal membranes but was protective against the negative impact of elevated cholesterol on this receptor. The cholesterol-insensitive CCK1R mutant Y140A was resistant to effects of both bile acids. These data suggest that bile acids compete with the action of cholesterol on CCK1R, probably by interacting at the same site, although the conformational impact of each bile acid appears to be different, with ursodeoxycholic acid capable of correcting the abnormal conformation of CCK1R in a high-cholesterol environment. This mechanism may contribute to the beneficial effect of ursodeoxycholic acid in reducing cholesterol gallstone formation.
Collapse
Affiliation(s)
- Aditya J. Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona
| | - Maoqing Dong
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona
| | - Kaleeckal G. Harikumar
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona
| | - Laurence J. Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Scottsdale, Arizona
| |
Collapse
|
20
|
Desai AJ, Henke BR, Miller LJ. Elimination of a cholecystokinin receptor agonist 'trigger' in an effort to develop positive allosteric modulators without intrinsic agonist activity. Bioorg Med Chem Lett 2015; 25:1849-55. [PMID: 25862198 DOI: 10.1016/j.bmcl.2015.03.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 01/25/2023]
Abstract
Cholecystokinin (CCK) acts at the type 1 cholecystokinin receptor (CCK1R) to elicit satiety and is a well-established drug target for obesity. To date, small molecule agonists have been developed, but have failed to demonstrate adequate efficacy in clinical trials, and concerns about side effects and potential toxicity have limited further development of full agonists. The use of positive allosteric modulators (PAMs) without intrinsic agonist activity that are active only for a brief period of time after a meal might represent a safer alternative. Here, we propose a possible novel strategy to develop such compounds by modifying the agonist 'trigger' of an existing small molecule agonist. We have studied analogues of the 1,5-benzodiazepine agonist, GI181771X, in which the N1-isopropyl agonist 'trigger' was modified. While agonist activity was greatly reduced in these compounds, they acted as negative, rather than positive modulators. The parent drug was also found to exhibit no positive modulation of CCK action. Receptor structure-activity relationship studies demonstrated that the mode of docking these derivatives was distinct from that of the parent compound, perhaps explaining their action as negative allosteric modulators. We conclude that this outcome is likely characteristic of the parental agonist, and that this strategy may be more successfully utilized with a parental ago-PAM, possessing intrinsic positive modulatory activity.
Collapse
Affiliation(s)
- Aditya J Desai
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ 85259, United States
| | - Brad R Henke
- Metabolic Pathways and Cardiovascular Therapy Area Unit, GlaxoSmithKline Inc., Research Triangle Park, NC 27709, United States
| | - Laurence J Miller
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, 13400 East Shea Blvd, Scottsdale, AZ 85259, United States.
| |
Collapse
|
21
|
Browning KN, Babic T, Holmes GM, Swartz E, Travagli RA. A critical re-evaluation of the specificity of action of perivagal capsaicin. J Physiol 2013; 591:1563-80. [PMID: 23297311 DOI: 10.1113/jphysiol.2012.246827] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Perivagal application of capsaicin (1% solution) is considered to cause a selective degeneration of vagal afferent C fibres and has been used extensively to examine the site of action of many gastrointestinal (GI) neuropeptides. The actions of both capsaicin and GI neuropeptides may not be restricted to vagal afferent fibres, however, as other non-sensory neurones have displayed sensitivity to capsaicin and brainstem microinjections of these neuropeptides induce GI effects similar to those obtained upon systemic application. The aim of the present study was to test the hypothesis that perivagal capsaicin induces degeneration of vagal efferents controlling GI functions. Experiments were conducted 7-14 days after 30 min unilateral perivagal application of 0.1-1% capsaicin. Immunohistochemical analyses demonstrated that, as following vagotomy, capsaicin induced dendritic degeneration, decreased choline acetyltransferase but increased nitric oxide synthase immunoreactivity in rat dorsal motor nucleus of the vagus (DMV) neurones. Electrophysiological recordings showed a decreased DMV input resistance and excitability due, in part, to the expression of a large conductance calcium-dependent potassium current and the opening of a transient outward potassium window current at resting potential. Furthermore, the number of DMV neurones excited by thyrotrophin-releasing hormone and the gastric motility response to DMV microinjections of TRH were decreased significantly. Our data indicate that perivagal application of capsaicin induced DMV neuronal degeneration and decreased vagal motor responses. Treatment with perivagal capsaicin cannot therefore be considered selective for vagal afferent C fibres and, consequently, care is needed when using perivagal capsaicin to assess the mechanism of action of GI neuropeptides.
Collapse
Affiliation(s)
- K N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, MC H109, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
22
|
Yamamoto M, Wei L, Otani M, Harada M, Otsuki M. Valsartan, a specific angiotensin II receptor blocker, inhibits pancreatic fluid secretion via vagal afferent pathway in conscious rats. ACTA ACUST UNITED AC 2012; 178:80-5. [DOI: 10.1016/j.regpep.2012.06.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 03/24/2012] [Accepted: 06/22/2012] [Indexed: 02/01/2023]
|
23
|
Babic T, Browning KN, Kawaguchi Y, Tang X, Travagli RA. Pancreatic insulin and exocrine secretion are under the modulatory control of distinct subpopulations of vagal motoneurones in the rat. J Physiol 2012; 590:3611-22. [PMID: 22711959 DOI: 10.1113/jphysiol.2012.234955] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Brainstem vago-vagal neurocircuits modulate upper gastrointestinal functions. Derangement of these sensory-motor circuits is implicated in several pathophysiological states, such as gastroesophageal reflux disease (GERD), functional dyspepsia and, possibly, pancreatitis. While vagal circuits controlling the stomach have received more attention, the organization of brainstem pancreatic neurocircuits is still largely unknown. We aimed to investigate the in vitro and in vivo modulation of brainstem vagal circuits controlling pancreatic secretion. Using patch clamp techniques on identified vagal pancreas-projecting neurones, we studied the effects of metabotropic glutamate receptor (mGluR) agents in relation to the effects of exendin-4, a glucagon-like peptide 1 analogue, cholecystokinin (CCK) and pancreatic polypeptide (PP). An in vivo anaesthetized rat preparation was used to measure pancreatic exocrine secretion (PES) and plasma insulin following microinjection of metabotropic glutamate receptor (mGluR) agonists and exendin-4 in the brainstem. Group II and III mGluR agonists (2R,4R-4-aminopyrrolidine-2,4-dicarboxylate (APDC) and L(+)-2-amino-4-phosphonobutyric acid (L-AP4), respectively) decreased the frequency of miniature inhibitory and excitatory postsynaptic currents (mIPSCs and mEPSCs, respectively) in the majority of the neurones tested. All neurones responsive to L-AP4 were also responsive to APDC, but not vice versa. Further, in neurones where L-AP4 decreased mIPSC frequency, exendin-4 increased, while PP had no effect upon, mIPSC frequency. Brainstem microinjection of APDC or L-AP4 decreased plasma insulin secretion, whereas only APDC microinjections increased PES. Exendin-4 microinjections increased plasma insulin. Our results indicate a discrete organization of vagal circuits, which opens up promising avenues of research aimed at investigating the physiology of homeostatic autonomic neurocircuits.
Collapse
Affiliation(s)
- Tanja Babic
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, 500 University Drive, MC H109, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
24
|
Cao B, Zhang X, Yan N, Chen S, Li Y. Cholecystokinin enhances visceral pain-related affective memory via vagal afferent pathway in rats. Mol Brain 2012; 5:19. [PMID: 22681758 PMCID: PMC3407758 DOI: 10.1186/1756-6606-5-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 06/09/2012] [Indexed: 11/20/2022] Open
Abstract
Background Pain contains both sensory and affective dimensions. Using a rodent visceral pain assay that combines the colorectal distension (CRD) model with the conditioned place avoidance (CPA) paradigms, we measured a learned behavior that directly reflects the affective component of visceral pain, and showed that perigenual anterior cingulate cortex (pACC) activation is critical for memory processing involved in long-term visceral affective state and prediction of aversive stimuli by contextual cue. Progress has been made and suggested that activation of vagal afferents plays a role in the behavioral control nociception and memory storage processes. In human patients, electrical vagus nerve stimulation enhanced retention of verbal learning performance. Cholecystokinin-octapeptide (CCK), which is a gastrointestinal hormone released during feeding, has been shown to enhance memory retention. Mice access to food immediately after training session enhanced memory retention. It has been well demonstrated that CCK acting on vagal afferent fibers mediates various physiological functions. We hypothesize that CCK activation of vagal afferent enhances visceral pain-related affective memory. Results In the presented study, infusion of CCK-8 at physiological concentration combining with conditional training significantly increased the CRD-induced CPA scores, and enhanced the pain affective memory retention. In contrast, CCK had no effect on CPA induced by non-nociceptive aversive stimulus (U69,593). The physiological implications were further strengthened by the similar effects observed in the rats with duodenal infusion of 5% peptone, which has been shown to induce increases in plasma CCK levels. CCK-8 receptor antagonist CR-1409 or perivagal application of capsaicin abolished the effect of CCK on aversive visceral pain memory, which was consistent with the notion that vagal afferent modulates affective aspects of visceral pain. CCK does not change the nociceptive response (visceral pain sensitivity) and anterior cingulate cortex neuronal responses to CRD. Conclusion CCK activating vagal afferent C fibers enhances memory consolidation and retention involved in long-term visceral negative affective state. Thus, in a number of gastrointestinal disorders, such as irritable bowel syndrome, nutrient content may contribute to painful visceral perception by enhancing visceral aversive memory via acts on vagal afferent pathway.
Collapse
Affiliation(s)
- Bing Cao
- Neuroscience Laboratory, Department of Biology and Chemistry, City University of Hong Kong, Kowloon, Hong Kong, China
| | | | | | | | | |
Collapse
|
25
|
Browning KN, Wan S, Baptista V, Travagli RA. Vanilloid, purinergic, and CCK receptors activate glutamate release on single neurons of the nucleus tractus solitarius centralis. Am J Physiol Regul Integr Comp Physiol 2011; 301:R394-401. [PMID: 21543639 DOI: 10.1152/ajpregu.00054.2011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Baroreceptor inputs to nucleus of the tractus solitarius medialis (mNTS) neurons can be differentiated, among other features, by their response to vanilloid or purinergic agonists, active only on C- or A-fibers, respectively. A major aim of this study was to examine whether neurons of NTS centralis (cNTS), a subnucleus dominated by esophageal inputs, exhibit a similar dichotomy. Since it has been suggested that cholecystokinin (CCK), exerts its gastrointestinal (GI)-related effects via paracrine activation of vagal afferent C-fibers, we tested whether CCK-sensitive fibers impinging upon cNTS neurons are responsive to vanilloid but not purinergic agonists. Using whole cell patch-clamp recordings from cNTS, we recorded miniature excitatory postsynaptic currents (mEPSCs) to test the effects of the vanilloid agonist capsaicin, the purinergic agonist α,β-methylene-ATP (α,β-Met-ATP), and/or CCK-octapeptide (CCK-8s). α,β-Met-ATP, capsaicin; and CCK-8s increased EPSC frequency in 37, 71, and 46% of cNTS neurons, respectively. Approximately 30% of cNTS neurons were responsive to both CCK-8s and α,β-Met-ATP, to CCK-8s and capsaicin, or to α,β-Met-ATP and capsaicin, while 32% of neurons were responsive to all three agonists. All neurons responding to either α,β-Met-ATP or CCK-8s were also responsive to capsaicin. Perivagal capsaicin, which is supposed to induce a selective degeneration of C-fibers, decreased the number of cNTS neurons responding to capsaicin or CCK-8s but not those responding to α,β-Met-ATP. In summary, GI inputs to cNTS neurons cannot be distinguished on the basis of their selective responses to α,β-Met-ATP or capsaicin. Our data also indicate that CCK-8s increases glutamate release from purinergic and vanilloid responsive fibers impinging on cNTS neurons.
Collapse
Affiliation(s)
- Kirsteen N Browning
- Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania 17033-0850, USA
| | | | | | | |
Collapse
|
26
|
Liou AP, Chavez DI, Espero E, Hao S, Wank SA, Raybould HE. Protein hydrolysate-induced cholecystokinin secretion from enteroendocrine cells is indirectly mediated by the intestinal oligopeptide transporter PepT1. Am J Physiol Gastrointest Liver Physiol 2011; 300:G895-902. [PMID: 21311026 PMCID: PMC3094145 DOI: 10.1152/ajpgi.00521.2010] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Dietary protein is a major stimulant for cholecystokinin (CCK) secretion by the intestinal I cell, however, the mechanism by which protein is detected is unknown. Indirect functional evidence suggests that PepT1 may play a role in CCK-mediated changes in gastric motor function. However, it is unclear whether this oligopeptide transporter directly or indirectly activates the I cell. Using both the CCK-expressing enteroendocrine STC-1 cell and acutely isolated native I cells from CCK-enhanced green fluorescent protein (eGFP) mice, we aimed to determine whether PepT1 directly activates the enteroendocrine cell to elicit CCK secretion in response to oligopeptides. Both STC-1 cells and isolated CCK-eGFP cells expressed PepT1 transcripts. STC-1 cells were activated, as measured by ERK(1/2) phosphorylation, by both peptone and the PepT1 substrate Cefaclor; however, the PepT1 inhibitor 4-aminomethyl benzoic acid (AMBA) had no effect on STC-1 cell activity. The PepT1-transportable substrate glycyl-sarcosine dose-dependently decreased gastric motility in anesthetized rats but had no affect on activation of STC-1 cells or on CCK secretion by CCK-eGFP cells. CCK secretion was significantly increased in response to peptone but not to Cefaclor, cephalexin, or Phe-Ala in CCK-eGFP cells. Taken together, the data suggest that PepT1 does not directly mediate CCK secretion in response to PepT1 specific substrates. PepT1, instead, may have an indirect role in protein sensing in the intestine.
Collapse
Affiliation(s)
- Alice P. Liou
- 1Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, California; and ,2Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Diana I. Chavez
- 1Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, California; and
| | - Elvis Espero
- 1Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, California; and
| | - Shuzhen Hao
- 1Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, California; and
| | - Stephen A. Wank
- 2Digestive Diseases Branch, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Helen E. Raybould
- 1Department of Anatomy, Physiology, and Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, California; and
| |
Collapse
|
27
|
Okano-Matsumoto S, McRoberts JA, Taché Y, Adelson DW. Electrophysiological evidence for distinct vagal pathways mediating CCK-evoked motor effects in the proximal versus distal stomach. J Physiol 2011; 589:371-93. [PMID: 21078593 PMCID: PMC3043539 DOI: 10.1113/jphysiol.2010.196832] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2010] [Accepted: 11/09/2010] [Indexed: 12/26/2022] Open
Abstract
Intravenous cholecystokinin octapeptide (CCK-8) elicits vago-vagal reflexes that inhibit phasic gastric contractions and reduce gastric tone in urethane-anaesthetized rats. A discrete proximal subdivision of the ventral gastric vagus nerve (pVGV) innervates the proximal stomach, but the fibre populations within it have not been characterized previously.We hypothesized that I.V. CCK-8 injection would excite inhibitory efferent outflow in the pVGV, in contrast to its inhibitory effect on excitatory efferent outflow in the distal subdivision (dVGV), which supplies the distal stomach. In each VGV subdivision, a dual-recording technique was used to record afferent and efferent activity simultaneously, while also monitoring intragastric pressure (IGP). CCK-8 dose dependently (100-1000 pmol kg(-1), I.V.) reduced gastric tone, gastric contractile activity and multi-unit dVGV efferent discharge, but increased pVGV efferent firing. Single-unit analysis revealed a minority of efferent fibres in each branch whose response differed in direction from the bulk response. Unexpectedly, efferent excitation in the pVGV was significantly shorter lived and had a significantly shorter decay half-time than did efferent inhibition in the dVGV, indicating that distinct pathways drive CCK-evoked outflow to the proximal vs. the distal stomach. Efferent inhibition in the dVGV began several seconds before, and persisted significantly longer than, simultaneously recorded dVGV afferent excitation.Thus, dVGV afferent excitation could not account for the pattern of dVGV efferent inhibition. However, the time course of dVGV afferent excitation paralleled that of pVGV efferent excitation. Similarly, the duration of CCK-8-evoked afferent responses recorded in the accessory celiac branch of the vagus (ACV) matched the duration of dVGV efferent responses. The observed temporal relationships suggest that postprandial effects on gastric complicance of CCK released from intestinal endocrine cells may require circulating concentrations to rise to levels capable of exciting distal gastric afferent fibres, in contrast to more immediate effects on distal gastric contractile activity mediated via vago-vagal reflexes initiated by paracrine excitation of intestinal afferents.
Collapse
|
28
|
de Lartigue G, Dimaline R, Varro A, Raybould H, de la Serre CB, Dockray GJ. Cocaine- and amphetamine-regulated transcript mediates the actions of cholecystokinin on rat vagal afferent neurons. Gastroenterology 2010; 138:1479-90. [PMID: 19854189 PMCID: PMC2847060 DOI: 10.1053/j.gastro.2009.10.034] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Revised: 10/01/2009] [Accepted: 10/13/2009] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Cholecystokinin (CCK) acts on vagal afferent neurons to inhibit food intake and gastric emptying; it also increases expression of the neuropeptide cocaine- and amphetamine-regulated transcript (CART), but the significance of this is unknown. We investigated the role of CARTp in vagal afferent neurons. METHODS Release of CART peptide (CARTp) from cultured vagal afferent neurons was determined by enzyme-linked immunosorbent assay. Expression of receptors and neuropeptides in rat vagal afferent neurons in response to CARTp was studied using immunohistochemistry and luciferase promoter reporter constructs. Effects of CARTp and CCK were studied on food intake. RESULTS CCK stimulated CARTp release from cultured nodose neurons. CARTp replicated the effect of CCK in stimulating expression of Y2R and of CART itself in these neurons in vivo and in vitro, but not in inhibiting cannabinoid-1, melanin-concentrating hormone, and melanin-concentrating hormone-1 receptor expression. Effects of CCK on Y2R and CART expression were reduced by CART small interfering RNA or brefeldin A. Exposure of rats to CARTp increased the inhibitory action of CCK on food intake after short-, but not long-duration, fasting. CONCLUSIONS The actions of CCK in stimulating CART and Y2R expression in vagal afferent neurons and in inhibiting food intake are augmented by CARTp; CARTp is released by CCK from these neurons, indicating that it acts as an autocrine excitatory mediator.
Collapse
Affiliation(s)
- Guillaume de Lartigue
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown St, Liverpool, L69 3BX, UK
| | - Rod Dimaline
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown St, Liverpool, L69 3BX, UK
| | - Andrea Varro
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown St, Liverpool, L69 3BX, UK
| | - Helen Raybould
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, UC Davis, California, USA
| | - Claire Barbier de la Serre
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, UC Davis, California, USA
| | - Graham J. Dockray
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown St, Liverpool, L69 3BX, UK
| |
Collapse
|
29
|
|
30
|
Abstract
The aim of the present review is to synthesise and summarise our recent knowledge on the involvement of cholecystokinin (CCK) and gastrin peptides and their receptors in the control of digestive functions and more generally their role in the field of nutrition in mammals. First, we examined the release of these peptides from the gut, focusing on their molecular forms, the factors regulating their release and the signalling pathways mediating their effects. Second, general physiological effects of CCK and gastrin peptides are described with regard to their specific receptors and the role of CCK on vagal mucosal afferent nerve activities. Local effects of CCK and gastrin in the gut are also reported, including gut development, gastrointestinal motility and control of pancreatic functions through vagal afferent pathways, including NO. Third, some examples of the intervention of the CCK and gastrin peptides are exposed in diseases, taking into account intervention of the classical receptor subtypes (CCK1 and CCK2 receptors) and their heterodimerisation as well as CCK-C receptor subtype. Finally, applications and future challenges are suggested in the nutritional field (performances) and in therapy with regards to the molecular forms or in relation with the type of receptor as well as new techniques to be utilised in detection or in therapy of disease. In conclusion, the present review underlines recent developments in this field: CCK and gastrin peptides and their receptors are the key factor of nutritional aspects; a better understanding of the mechanisms involved may increase the efficiency of the nutritional functions and the treatment of abnormalities under pathological conditions.
Collapse
|
31
|
Dockray GJ. Cholecystokinin and gut-brain signalling. ACTA ACUST UNITED AC 2009; 155:6-10. [PMID: 19345244 DOI: 10.1016/j.regpep.2009.03.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Accepted: 03/25/2009] [Indexed: 01/04/2023]
Abstract
Enteroendocrine cells of the gastrointestinal tract act as a luminal surveillance system responding to either the presence or absence of food in the gut lumen. Collectively, their secretory products regulate the course of digestion and determine the delivery of nutrient to the gut by controlling food intake. Afferent neurons of the vagus nerve are an important target of gut hormones, particularly for control of food intake. The intestinal hormone cholecystokinin (CCK) stimulates vagal afferent neuron discharge and also controls the expression of both G-protein coupled receptors and peptide neurotransmitters in these neurons. When plasma CCK concentrations are low, for example in fasting, vagal afferent neurons express cannabinoid CB1 and melanin concentrating hormone (MCH)-1 receptors, both of which are associated with stimulation of food intake. Post-prandial release of CCK rapidly down-regulates the expression of both receptors but stimulates the expression of Y2 receptors in neurons projecting to the stomach. In fasting, there is also increased expression in these neurons of the appetite-stimulating neuropeptide transmitter MCH, and depressed expression of the satiety-peptide cocaine and amphetamine regulated transcript (CART). Secretion of CCK decreases expression of MCH and increases expression of CART. The neurochemical phenotype of vagal afferent neurons therefore encodes whether or not there has been nutrient ingestion over the previous period. At low plasma concentrations of CCK vagal afferent neurons exhibit increased capacity for appetite-stimulation, while post-prandial concentrations of CCK lead to enhanced capacity for satiety signalling. A gatekeeper function can therefore be attributed to CCK in that its presence or absence influences the capacity of vagal afferent neurons to respond to other neurohormonal signals.
Collapse
Affiliation(s)
- Graham J Dockray
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Crown St, Liverpool L69 3BX, UK.
| |
Collapse
|
32
|
Dockray GJ. The versatility of the vagus. Physiol Behav 2009; 97:531-6. [PMID: 19419683 DOI: 10.1016/j.physbeh.2009.01.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2008] [Revised: 01/13/2009] [Accepted: 01/14/2009] [Indexed: 12/24/2022]
Abstract
The gut is one of several organs contributing to the peripheral signalling network that controls food intake. Afferent neurons of the vagus nerve provide an important pathway for gut signals that act by triggering ascending pathways from the brain stem to hypothalamus. Recent work indicates the existence of mechanisms operating at the level of vagal afferent neurons to modulate the effect of gastrointestinal satiety signals. Thus, the well known satiety hormone cholecystokinin (CCK) not only stimulates the discharge of these neurons but also controls their expression of both G-protein coupled receptors and peptide neurotransmitters known to influence food intake. When plasma CCK concentrations are low e.g. in fasting, the expression by vagal afferent neurons of cannabinoid (CB)-1 and melanin concentrating hormone (MCH)-1 receptors is increased. Release of CCK by feeding leads to a rapid down-regulation of expression of both receptors and to increased expression of Y2 receptors. In fasting, there is also increased expression in these neurons of the appetite-stimulating neuropeptide transmitter MCH, and depressed expression of the satiety-peptide cocaine and amphetamine regulated transcript (CARTp); endogenous CCK decreases MCH expression and stimulates CART expression. The gastric orexigenic hormone ghrelin blocks these actions of CCK at least in part by excluding phosphoCREB from the nucleus. The data suggest that CCK acts as a gatekeeper to determine the capacity of other neuroendocrine signals to act via vagal afferent neurons to influence food intake.
Collapse
Affiliation(s)
- Graham J Dockray
- Physiological Laboratory, School of Biomedical Sciences, University of Liverpool, Liverpool, UK.
| |
Collapse
|
33
|
Mussa BM, Sartor DM, Verberne AJM. Activation of cholecystokinin (CCK 1) and serotonin (5-HT 3) receptors increases the discharge of pancreatic vagal afferents. Eur J Pharmacol 2008; 601:198-206. [PMID: 19026634 DOI: 10.1016/j.ejphar.2008.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2008] [Accepted: 11/03/2008] [Indexed: 01/13/2023]
Abstract
Cholecystokinin and serotonin are released from the gastrointestinal tract in response to the products of digestion and play critical roles in mediating pancreatic secretion via vago-vagal reflex pathways. This study was designed to investigate the effects of activation of cholecystokinin CCK(1) and serotonin (5-hydroxytryptamine, 5-HT) 5-HT(3) receptors on pancreatic vagal afferent discharge and to determine whether there is an interaction between these receptors. Male Sprague Dawley rats anaesthetised with isoflurane (1.5%/100% O(2)) were used in all experiments. The effects of systemic administration of cholecystokinin and the serotonin 5-HT(3) receptor agonist phenylbiguanide on pancreatic vagal afferent discharge were recorded before and after administration of cholecystokinin CCK(1) and serotonin 5-HT(3) receptor antagonists. Cholecystokinin (0.1-10 microg/kg, i.v.) and phenylbiguanide (1 and 10 microg/kg, i.v.) increased pancreatic vagal afferent discharge dose-dependently. Cholecystokinin CCK(1) receptor antagonists, lorglumide (10 mg/kg, i.v.) and devazepide (0.5 mg/kg, i.v.), reduced cholecystokinin- and phenylbiguanide-induced increases in pancreatic vagal afferent discharge significantly (n=5, P<0.05). On the other hand, serotonin 5-HT(3) receptor blockade with granisetron (1 mg/kg, i.v.) or MDL72222 ([(1S,5R)-8-methyl-8-azabicyclo[3.2.1]octan-3-yl] 3,5-dichlorobenzoate; 0.1 mg/kg, i.v.) inhibited the pancreatic vagal afferent discharge responses to phenylbiguanide but not those to cholecystokinin. This study has confirmed that cholecystokinin and phenylbiguanide activate pancreatic vagal afferent discharge via activation of cholecystokinin CCK(1) and serotonin 5-HT(3) receptors, respectively. In addition, it has demonstrated that (i) the serotonin 5-HT(3) agonist phenylbiguanide acts partly via an interaction with cholecystokinin CCK(1) receptors, and (ii) the actions of cholecystokinin are not dependent on serotonin 5-HT(3) receptor activation.
Collapse
Affiliation(s)
- Bashair M Mussa
- University of Melbourne, Department of Medicine, Clinical Pharmacology and Therapeutics Unit, Austin Health, Heidelberg 3084, Victoria, Australia
| | | | | |
Collapse
|
34
|
Sartor DM, Verberne AJ. Abdominal vagal signalling: A novel role for cholecystokinin in circulatory control? ACTA ACUST UNITED AC 2008; 59:140-54. [DOI: 10.1016/j.brainresrev.2008.07.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2008] [Revised: 06/24/2008] [Accepted: 07/07/2008] [Indexed: 02/07/2023]
|
35
|
Stearns AT, Balakrishnan A, Rounds J, Rhoads DB, Ashley SW, Tavakkolizadeh A. Capsaicin-sensitive vagal afferents modulate posttranscriptional regulation of the rat Na+/glucose cotransporter SGLT1. Am J Physiol Gastrointest Liver Physiol 2008; 294:G1078-83. [PMID: 18308853 DOI: 10.1152/ajpgi.00591.2007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION the intestinal Na(+)/glucose cotransporter (SGLT1) displays rapid anticipatory diurnal rhythms in mRNA and protein expression. The vagus nerve has been implicated in the entrainment of some transporters. We aimed to clarify the influence of the vagus nerve on the diurnal entrainment pathway for SGLT1 and examine the role of vagal afferent fibers. METHODS male Sprague-Dawley rats were randomized to three groups, total subdiaphragmatic vagotomy, selective deafferentation of the vagus with capsaicin, or sham laparotomy. Postoperatively, animals were maintained in a 12-h light-dark cycle with food access limited to night. On the ninth postoperative day, animals were euthanized to harvest jejunal mucosa at 6-h intervals starting at 10 AM. Whole cell SGLT1 protein was measured by semiquantitative densitometry of immunoblots. Sglt1 and regulatory subunit RS1 mRNA was assessed by quantitative PCR. Fluorogold tracer technique was used to confirm adequacy of the vagotomy. RESULTS the diurnal rhythm in intestinal SGLT1, with a 5.3-fold increase in Sglt1 mRNA at 4 PM, was preserved in both vagotomy and capsaicin groups. However, the rhythmicity in SGLT1 protein expression (2.3-fold peak at 10 PM; P = 0.041) was abolished following either total vagotomy or deafferentation. Lack of change in RS1 mRNA suggests this is independent of the RS1 regulatory pathway. CONCLUSION SGLT1 transcription is independent of the vagus. However, dissociation of the protein rhythm from the underlying mRNA signal by vagotomy suggests the vagus may be involved in posttranscriptional regulation of SGLT1 in an RS1 independent pathway. Disruption following afferent ablation by capsaicin suggests this limb is specifically necessary.
Collapse
Affiliation(s)
- Adam T Stearns
- Dept. of Surgery, Brigham & Women's Hospital, 75 Francis St., Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
36
|
Hao S, Sternini C, Raybould HE. Role of CCK1 and Y2 receptors in activation of hindbrain neurons induced by intragastric administration of bitter taste receptor ligands. Am J Physiol Regul Integr Comp Physiol 2008; 294:R33-8. [DOI: 10.1152/ajpregu.00675.2007] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
G-protein-coupled receptors signaling bitter taste (T2Rs) in the oral gustatory system and the α-subunit of the taste-specific G-protein gustducin are expressed in the gastrointestinal (GI) tract. α-Subunit of the taste-specific G-protein gustducin colocalizes with markers of enteroendocrine cells in human and mouse GI mucosa, including peptide YY. Activation of T2Rs increases cholecystokinin (CCK) release from the enteroendocrine cell line, STC-1. The aim of this study was to determine whether T2R agonists in the GI tract activate neurons in the nucleus of the solitary tract (NTS) and whether this activation is mediated by CCK and peptide YY acting at CCK1 and Y2 receptors. Immunocytochemistry for the protooncogene c-Fos protein, a marker for neuronal activation, was used to determine activation of neurons in the midregion of the NTS, the region where vagal afferents from the GI tract terminate. Intragastric administration of the T2R agonist denatonium benzoate (DB), or phenylthiocarbamide (PTC), or a combination of T2R agonists significantly increased the number of Fos-positive neurons in the mid-NTS; subdiaphragmatic vagotomy abolished the NTS response to the mixture of T2R agonists. Deletion of CCK1 receptor gene or blockade of CCK1 receptors with devazepide abolishes the activation of NTS neurons in response to DB, but had no effect on the response to PTC. Administration of the Y2 receptor antagonist BIIE0246 blocks the activation of NTS neurons to DB, but not PTC. These findings suggest that activation of neurons in the NTS following administration of T2R agonists to the GI tract involves CCK1 and Y2 receptors located on vagal afferent terminals in the gut wall. T2Rs may regulate GI function via release of regulatory peptides and activation of the vagal reflex pathway.
Collapse
|
37
|
Sabbatini ME, Rodríguez MR, Dabas P, Vatta MS, Bianciotti LG. C-type natriuretic peptide stimulates pancreatic exocrine secretion in the rat: role of vagal afferent and efferent pathways. Eur J Pharmacol 2007; 577:192-202. [PMID: 17900562 DOI: 10.1016/j.ejphar.2007.08.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Revised: 08/28/2007] [Accepted: 08/29/2007] [Indexed: 11/28/2022]
Abstract
We previously reported that C-type natriuretic peptide (CNP) increases amylase release in isolated pancreatic acini through natriuretic peptide receptor C activation and enhances pancreatic exocrine secretion via vagal pathways when applied to the brain. In the present study we sought to establish whether CNP was involved in the peripheral regulation of pancreatic secretion. Anesthetized rats were prepared with pancreatic duct cannulation, pyloric ligation and bile diversion into the duodenum. CNP dose-dependently enhanced pancreatic flow, chloride and protein excretion but did not modify bicarbonate output. A selective natriuretic peptide receptor C agonist enhanced pancreatic flow and mimicked CNP-evoked protein output but failed to modify chloride secretion. Truncal vagotomy, perivagal application of capsaicin and hexamethonium reduced CNP-evoked pancreatic flow and abolished chloride excretion but did not affect protein output. Furthermore, pre-treatment with atropine reduced both CNP-stimulated pancreatic flow and chloride excretion but failed to modify protein excretion. Partial muscarinic blockade of CNP-evoked chloride output suggested that mediators other than acetylcholine were involved. However, CNP response was unaltered by cholecystokinin and vasoactive intestinal peptide receptor blockade or by nitric oxide synthase inhibition. In conclusion, CNP-stimulated pancreatic flow through the activation of the natriuretic peptide receptor C and the vago-vagal reflex but it increased protein output only by natriuretic peptide receptor C activation and chloride excretion by vago-vagal reflexes. Present results suggest that CNP may play a role as a local regulator of the exocrine pancreas.
Collapse
Affiliation(s)
- María E Sabbatini
- Cátedra de Fisiopatología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
38
|
Viard E, Zheng Z, Wan S, Travagli RA. Vagally mediated, nonparacrine effects of cholecystokinin-8s on rat pancreatic exocrine secretion. Am J Physiol Gastrointest Liver Physiol 2007; 293:G493-500. [PMID: 17569741 DOI: 10.1152/ajpgi.00118.2007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cholecystokinin (CCK) has been proposed to act in a vagally dependent manner to increase pancreatic exocrine secretion via actions exclusively at peripheral vagal afferent fibers. Recent evidence, however, suggests the CCK-8s may also affect brain stem structures directly. We used an in vivo preparation with the aims of 1) investigating whether the actions of intraduodenal casein perfusion to increase pancreatic protein secretion also involved direct actions of CCK at the level of the brain stem and, if so, 2) determining whether, in the absence of vagal afferent inputs, CCK-8s applied to the dorsal vagal complex (DVC) can also modulate pancreatic exocrine secretion (PES). Sprague-Dawley rats (250-400 g) were anesthetized and the common bile-pancreatic duct was cannulated to collect PES. Both vagal deafferentation and pretreatment with the CCK-A antagonist lorglumide on the floor of the fourth ventricle decreased the casein-induced increase in PES output. CCK-8s microinjection (450 pmol) in the DVC significantly increased PES; the increase was larger when CCK-8s was injected in the left side of the DVC. Protein secretion returned to baseline levels within 30 min. Microinjection of CCK-8s increased PES (although to a lower extent) also in rats that underwent complete vagal deafferentation. These data indicate that, as well as activating peripheral vagal afferents, CCK-8s increases pancreatic exocrine secretion via an action in the DVC. Our data suggest that the CCK-8s-induced increases in PES are due mainly to a paracrine effect of CCK; however, a relevant portion of the effects of CCK is due also to an effect of the peptide on brain stem vagal circuits.
Collapse
Affiliation(s)
- Eddy Viard
- Department of Neuroscience, Pennington Biomedical Research Center-Louisiana State University System, 6400 Perkins Rd., Baton Rouge, LA 70808, USA
| | | | | | | |
Collapse
|
39
|
Wang BJ, Cui ZJ. How does cholecystokinin stimulate exocrine pancreatic secretion? From birds, rodents, to humans. Am J Physiol Regul Integr Comp Physiol 2007; 292:R666-R678. [PMID: 17053097 DOI: 10.1152/ajpregu.00131.2006] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The field of cholecystokinin (CCK) stimulation of exocrine pancreatic secretion has experienced major changes in the recent past. This review attempts to summarize the present status of the field. CCK production in the intestinal I cells, the molecular forms of CCK produced and subsequently circulated in the blood, the presence or absence of CCK receptors on the isolated pancreatic acinar cells and the associated signaling for acinar cell secretion, and the actual circuits and sites of action for CCK regulation of exocrine pancreatic secretion in vivo are reviewed in different animal species with an emphasis on birds, rodents, and humans. Clear differences in the relative importance of neural and direct modes of CCK action on pancreatic acinar cells were identified. Rodents seem to be endowed with both modes of action, whereas in humans the neural mode may predominate. In birds, such as duck, the direct mode needs further assistance from pituitary adenylate cyclase-activating peptide/VIP receptors. However, much further work needs to be directed to the neural mode to map out all sites of CCK action and details of the full circuits, and we foresee a major revival for this field of research in the near future.
Collapse
Affiliation(s)
- Bi Jue Wang
- Institute of Cell Biology, Beijing Normal University, Beijing 100875, China
| | | |
Collapse
|
40
|
Baptista V, Browning KN, Travagli RA. Effects of cholecystokinin-8s in the nucleus tractus solitarius of vagally deafferented rats. Am J Physiol Regul Integr Comp Physiol 2006; 292:R1092-100. [PMID: 17122331 PMCID: PMC3062489 DOI: 10.1152/ajpregu.00517.2006] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We have shown recently that cholecystokinin octapeptide (CCK-8s) increases glutamate release from nerve terminals onto neurons of the nucleus tractus solitarius pars centralis (cNTS). The effects of CCK on gastrointestinal-related functions have, however, been attributed almost exclusively to its paracrine action on vagal afferent fibers. Because it has been reported that systemic or perivagal capsaicin pretreatment abolishes the effects of CCK, the aim of the present work was to investigate the response of cNTS neurons to CCK-8s in vagally deafferented rats. In surgically deafferented rats, intraperitoneal administration of 1 or 3 mug/kg CCK-8s increased c-Fos expression in cNTS neurons (139 and 251% of control, respectively), suggesting that CCK-8s' effects are partially independent of vagal afferent fibers. Using whole cell patch-clamp techniques in thin brain stem slices, we observed that CCK-8s increased the frequency of spontaneous and miniature excitatory postsynaptic currents in 43% of the cNTS neurons via a presynaptic mechanism. In slices from deafferented rats, the percentage of cNTS neurons receiving glutamatergic inputs responding to CCK-8s decreased by approximately 50%, further suggesting that central terminals of vagal afferent fibers are not the sole site for the action of CCK-8s in the brain stem. Taken together, our data suggest that the sites of action of CCK-8s include the brain stem, and in cNTS, the actions of CCK-8s are not restricted to vagal central terminals but that nonvagal synapses are also involved.
Collapse
Affiliation(s)
- V Baptista
- Department of Neuroscience, Pennington Biomedical Research Center, Louisiana State University System, 6400 Perkins Road, Baton Rouge, LA 70808, USA
| | | | | |
Collapse
|
41
|
Abstract
Cholecystokinin and gastrin receptors (CCK1R and CCK2R) are G protein-coupled receptors that have been the subject of intensive research in the last 10 years with corresponding advances in the understanding of their functioning and physiology. In this review, we first describe general properties of the receptors, such as the different signaling pathways used to exert short- and long-term effects and the structural data that explain their binding properties, activation, and regulation. We then focus on peripheral cholecystokinin receptors by describing their tissue distribution and physiological actions. Finally, pathophysiological peripheral actions of cholecystokinin receptors and their relevance in clinical disorders are reviewed.
Collapse
Affiliation(s)
- Marlène Dufresne
- Institut National de la Santé et de la Recherche Médicale U. 531, Institut Louis Bugnard, Centre Hospitalier Universitaire Rangueil, France
| | | | | |
Collapse
|
42
|
Whited KL, Thao D, Lloyd KCK, Kopin AS, Raybould HE. Targeted disruption of the murine CCK1 receptor gene reduces intestinal lipid-induced feedback inhibition of gastric function. Am J Physiol Gastrointest Liver Physiol 2006; 291:G156-62. [PMID: 16574983 DOI: 10.1152/ajpgi.00569.2005] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cholecystokinin (CCK), acting at CCK1 receptors (CCK1Rs) on intestinal vagal afferent terminals, has been implicated in the control of gastrointestinal function and food intake. Using CCK1R(-/-) mice, we tested the hypothesis that lipid-induced activation of the vagal afferent pathway and intestinal feedback of gastric function is CCK1R dependent. In anesthetized CCK1R(+/+) ("wild type") mice, meal-stimulated gastric acid secretion was inhibited by intestinal lipid infusion; this was abolished in CCK1R(-/-) mice. Gastric emptying of whole egg, measured by nuclear scintigraphy in awake mice, was significantly faster in CCK1R(-/-) than CCK1R(+/+) mice. Gastric emptying of chow was significantly slowed in response to administration of CCK-8 (22 pmol) in CCK1R(+/+) but not CCK1R(-/-) mice. Activation of the vagal afferent pathway was measured by immunohistochemical localization of Fos protein in the nucleus of the solitary tract (NTS; a region where vagal afferents terminate). CCK-8 (22 pmol ip) increased neuronal Fos expression in the NTS of fasted CCK1R(+/+) mice; CCK-induced Fos expression was reduced by 97% in CCK1R(-/-) compared with CCK1R(+/+) mice. Intralipid (0.2 ml of 20% Intralipid and 0.04 g lipid), but not saline, gavage increased Fos expression in the NTS of fasted CCK1R(+/+) mice; lipid-induced Fos expression was decreased by 47% in CCK1R(-/-) compared with CCK1R(+/+)mice. We conclude that intestinal lipid activates the vagal afferent pathway, decreases gastric acid secretion, and delays gastric emptying via a CCK1R-dependent mechanism. Thus, despite a relatively normal phenotype, intestinal feedback in response to lipid is severely impaired in these mice.
Collapse
Affiliation(s)
- K L Whited
- Department of Anatomy, Physiology, and Cell Biology, School of Vetinary Medicine, University of California, Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
43
|
Li Y, Wu X, Zhao Y, Chen S, Owyang C. Ghrelin acts on the dorsal vagal complex to stimulate pancreatic protein secretion. Am J Physiol Gastrointest Liver Physiol 2006; 290:G1350-8. [PMID: 16469825 DOI: 10.1152/ajpgi.00493.2005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ghrelin receptors are present in the central nervous system. We hypothesized that ghrelin released from the stomach acts as an endocrine substance and stimulates brain stem vagovagal circuitry to evoke pancreatic secretion. In an in vivo anesthetized rat model, an intravenous infusion of ghrelin at doses of 5, 10, and 25 nmol increased pancreatic protein secretion from a basal level of 125 +/- 6 to 186 +/- 8, 295 +/- 12, and 356 +/- 11 mg/h, respectively. Pretreatment with atropine or hexamethonium or an acute vagotomy, but not a perivagal application of capsaicin, completely abolished pancreatic protein secretion responses to ghrelin. In conscious rats, an intravenous infusion of ghrelin at a dose of 10 nmol resulted in a 2.2-fold increase in pancreatic protein secretion over basal volume. Selective ablation of the area postrema abolished pancreatic protein secretion stimulated by intravenous infusion of ghrelin but did not alter the increase in pancreatic protein secretion evoked by diversion of bile-pancreatic juice. Immunohistochemical staining showed a marked increase in the number of c-Fos-expressing neurons in the area postrema, nucleus of the solitary tract, and dorsal motor nucleus of the vagus after an intravenous infusion of ghrelin in sham-lesioned rats; selective ablation of the area postrema eliminated this increase. In conclusion, ghrelin stimulates pancreatic secretion via a vagal cholinergic efferent pathway. Circulating ghrelin gains access to the brain stem vagovagal circuitry via the area postrema, which represents the primary target on which peripheral ghrelin may act as an endocrine substance to stimulate pancreatic secretion.
Collapse
Affiliation(s)
- Ying Li
- Gastroenterology Research Unit, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, 48109-0682, USA.
| | | | | | | | | |
Collapse
|
44
|
Sartor DM, Shulkes A, Verberne AJM. An enteric signal regulates putative gastrointestinal presympathetic vasomotor neurons in rats. Am J Physiol Regul Integr Comp Physiol 2005; 290:R625-33. [PMID: 16239368 DOI: 10.1152/ajpregu.00639.2005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ingestion of a meal results in gastrointestinal (GI) hyperemia and is associated with systemic and paracrine release of a number of peptide hormones, including cholecystokinin (CCK) and 5-hydroxytryptamine (5-HT). Systemic administration of CCK octapeptide inhibits a subset of presympathetic neurons of the rostroventrolateral medulla (RVLM) that may be responsible for driving the sympathetic vasomotor tone to the GI viscera. The aim of this study was to determine whether endogenous release of CCK and/or 5-HT also inhibits CCK-sensitive RVLM neurons. The effects of intraduodenal administration of the secretagogues sodium oleate (SO) and soybean trypsin inhibitor (SBTI) on circulating levels of CCK and 5-HT were examined. In separate experiments, the discharge rates of barosensitive, medullospinal, CCK-sensitive RVLM presympathetic vasomotor neurons were recorded after rapid intraduodenal infusion of SO-SBTI or water. Alternatively, animals were pretreated with the CCK1 receptor antagonists devazepide and lorglumide or the 5-HT3 antagonist MDL-72222 before SO-SBTI administration. Secretagogue infusion significantly increased the level of circulating CCK, but not 5-HT. SO-SBTI significantly decreased (58%) the neuronal firing rate of CCK-sensitive RVLM neurons compared with water (5%). CCK1 receptor antagonists did not reverse SO-SBTI-induced neuronal inhibition (58%), whereas the 5-HT3 antagonist significantly attenuated the effect (22%). This study demonstrates a functional relation between a subset of RVLM presympathetic vasomotor neurons and meal-related signals arising from the GI tract. It is likely that endogenously released 5-HT acts in a paracrine fashion on GI 5-HT3 receptors to initiate reflex inhibition of these neurons, resulting in GI vasodilatation by withdrawal of sympathetic tone.
Collapse
Affiliation(s)
- Daniela M Sartor
- Clinical Pharmacology and Therapeutics Unit, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, Victoria 3084, Australia.
| | | | | |
Collapse
|
45
|
|
46
|
Darcel NP, Liou AP, Tomé D, Raybould HE. Activation of vagal afferents in the rat duodenum by protein digests requires PepT1. J Nutr 2005; 135:1491-5. [PMID: 15930458 DOI: 10.1093/jn/135.6.1491] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Intestinal infusion of protein digests activates a vago-vagal reflex inhibition of gastric motility. Protein digests release cholecystokinin (CCK) from enteroendocrine cells; however, the precise cellular mechanisms leading to vagal afferent activation is unclear. The hypothesis that the oligopeptide transporter PepT1 plays a major role in the initiation of this vago-vagal reflex was tested by recording activation of duodenal vagal afferent activity and inhibition of gastric motility in response to protein hydrolysates in the presence of 4-aminomethylbenzoic acid (4-AMBA), a competitive inhibitor of PepT1, or 4-aminophenylacetic acid (4-APAA), an inactive 4-AMBA analog. Duodenal infusion of the protein hydrolysate increased vagal afferent discharge and inhibited gastric motility; these responses were abolished by concomitant infusion of 4-AMBA, but not 4-APAA. Duodenal infusion with Cefaclor, a substrate of PepT1, increased duodenal vagal afferent activity; Cefaclor and protein hydrolysates selectively activated CCK-responsive vagal afferents. This study demonstrates that products of protein digestion increase spontaneous activity of CCK-sensitive duodenal vagal afferents via a mechanism involving the oligopeptide transporter PepT1.
Collapse
Affiliation(s)
- N P Darcel
- Department of Anatomy, Physiology and Cell Biology, UC Davis School of Veterinary Medicine, Davis, CA 95616, USA
| | | | | | | |
Collapse
|
47
|
Liao Z, Li ZS, Lu Y, Wang WZ. Glutamate receptors within the nucleus of solitary tract contribute to pancreatic secretion stimulated by intraduodenal hypertonic saline. Auton Neurosci 2005; 120:62-7. [PMID: 15897013 DOI: 10.1016/j.autneu.2005.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2005] [Revised: 04/13/2005] [Accepted: 04/14/2005] [Indexed: 02/05/2023]
Abstract
It is well known that central transmission of vago-vagal reflex within the nucleus of solitary tract (NST) plays an important role in the regulation of gastrointestinal functions. The present study was designed to assess the role of NST glutamate receptor mechanism in pancreatic secretion evoked by intraduodenal hypertonic saline (HS) in anesthetized rats. Intraduodenal infusion of HS significantly (P<0.01) stimulated pancreatic protein output (from 2.60+/-0.09 to 4.18+/-0.24 mg/15 min). Bilaterally microinjected L-glutamate (5 nmol) into the medial nucleus of solitary tract (mNST) produced a significant increase of pancreatic protein secretion (from 2.65+/-0.12 to 4.80+/-0.34 mg/15 min, P<0.01). Bilateral injection of glutamate receptor antagonist kynurenic acid (KYN, 5 nmol) into the mNST completely abolished the increase of pancreatic protein output stimulated by intraduodenal HS (from 4.28+/-0.21 to 2.83+/-0.19 mg/15 min). Either NMDA receptor antagonist dl-2-amino-5-phosphonopentanoic acid (AP5, 1.5 nmol) or AMPA/Kainate receptor antagonist 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX, 1.5 nmol) injected into the mNST markedly attenuated (P<0.05) the pancreatic protein secretion stimulated by intraduodenal HS. In conclusion, these findings showed that blockade of the NST glutamate receptors, including NMDA and AMPA/Kainate receptors antagonized pancreatic secretion evoked by intraduodenal osmolality factor, and suggested that glutamate receptor mechanism within the NST contributed to the central regulation of pancreatic secretion.
Collapse
Affiliation(s)
- Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, 174 Chang-Hai Road, Shanghai 200433, PR China.
| | | | | | | |
Collapse
|
48
|
Deng X, Wood PG, Eagon PK, Whitcomb DC. Rapid adaptation of pancreatic exocrine function to short-term alcohol feeding in rats. Pancreatology 2005; 5:183-95. [PMID: 15855815 DOI: 10.1159/000085270] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2003] [Accepted: 07/15/2004] [Indexed: 12/11/2022]
Abstract
BACKGROUND Chronic alcohol consumption increases the risk of pancreatitis in humans. Functional hyperstimulation/hypersecretion of the pancreas during chronic alcohol consumption appears to precede the onset of pancreatitis, and may contribute to the increased susceptibility to pancreatitis in alcoholics. However, the origin, nature and timing of hyperstimulation/hypersecretion are unknown. METHODS Male Wistar rats were pair-fed ethanol liquid diet for 15-18 days (including one 9-day dose ramp-up phase) or regular liquid diets before placement of pancreatic, biliary, duodenal and venous catheters. Basal and stimulated pancreatic secretions were measured with or without acute alcohol infusion. Pancreatic secretion was stimulated with intravenous bethanechol, 2-deoxy-D-glucose (2-DG), cholecystokinin (CCK), octapeptide (CCK-8), intraduodenal meal, or vehicle. RESULTS Acute alcohol potentiated 2-DG stimulated pancreatic secretion (184%, p < 0.05), whereas the response to CCK was unchanged, and the response to bethanechol was decreased (78%, p < 0.05). Short-term alcohol exposure lessened the exaggerated protein secretory response to 2-DG seen in acute alcohol exposure rats and increased the protein response to bethanechol (141%, p < 0.05), CCK (187%, p < 0.05) and meal (217%, p < 0.05). CONCLUSION The pancreas is sensitive to acute alcohol ingestion with inhibition of acinar cell function. Rapid adaptation occurs with short-term alcohol feeding, resulting in an exaggerated response to cholinergic input at the acinar cells, plus disinhibition of CCK and meal-stimulated pancreatic secretion. The central response to 2-DG and CCK are similar to area postrema lesions. Adaptation appears to be in response to alcohol-associated inhibition of the neurohormonal stimulatory pathway and compensatory upregulation at the acinar cell level.
Collapse
Affiliation(s)
- Xiaoying Deng
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
49
|
Abstract
The existence of high- and low-affinity cholecystokinin (CCK)-A receptors on rodent pancreatic acini is well established. Until recently, CCK was believed to act directly on pancreatic acini to stimulate pancreatic secretion in both rodents and humans. However, conclusive evidence that human pancreatic acini lack functional CCK-A receptors has been presented. Despite substantial differences in rodent and human pancreatic physiology, CCK appears to act via vagal cholinergic pathways to mediate pancreatic secretion in both species. Structural and functional evidence suggests that CCK acts on vagal afferent fibers, which may explain how CCK doses that produce physiologic plasma CCK levels act via vagal cholinergic pathways to stimulate pancreatic secretion. Although most knowledge of vagal CCK-A receptors comes from research on rodents, physiologic studies suggest that this information is applicable to humans. In contrast to its effect on satiety, which is mediated by low-affinity vagal CCK-A receptors, CCK acts through high-affinity CCK-A receptors to evoke pancreatic secretion, suggesting that different affinity states of the vagal CCK receptors mediate different digestive functions. Vagal afferent pathways also transmit sensory information about the mechanical and physiochemical state of the digestive tract, mediated in part by serotonin, which, in turn, influences pancreatic secretion. A synergistic interaction between CCK and serotonin at the level of the nodose ganglia may explain the robust postprandial pancreatic secretion despite a modest postprandial increase in plasma CCK. Important physiologically, these findings not only explain discrepancies in previous in vivo vs. in vitro studies, but they revolutionize our current concept of the mechanism of CCK on pancreatic exocrine secretion.
Collapse
Affiliation(s)
- Chung Owyang
- Department of Internal Medicine, 3912 Taubman Center, University of Michigan Health System, Ann Arbor, Michigan 48109-0362, USA.
| | | |
Collapse
|
50
|
Li Y, Wu X, Zhu J, Yan J, Owyang C. Hypothalamic regulation of pancreatic secretion is mediated by central cholinergic pathways in the rat. J Physiol 2004; 552:571-87. [PMID: 14561838 PMCID: PMC2343380 DOI: 10.1113/jphysiol.2003.049122] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The vago-vagal reflex plays an important role in mediating pancreatic secretion evoked by cholecystokinin and non-cholecystokinin-dependent luminal factors. We hypothesize that the vago-vagal reflex mediating pancreatic secretion in the rat is under central control and regulated by cholinergic pathways in the hypothalamus. To test this hypothesis, we demonstrated that chronic decerebration decreased basal pancreatic enzyme secretion from 318 +/- 12 to 233 +/- 9 mg h-1 and reduced the net increase in pancreatic secretion stimulated by intraduodenal infusion of 5 % peptone and hypertonic NaCl by 54 % and 45 %, respectively. Intracerebroventricular administration of methscopolamine (MSCP, 50 nmol (5 mul)-1), a blood-brain barrier-impermeant cholinergic muscarinic receptor antagonist, evoked results similar to those achieved by chronic decerebration. To localize the sites of action, we demonstrated that microinjection of MSCP (20 nmol) into the lateral hypothalamic nucleus or the paraventricular nucleus resulted in inhibition of both basal pancreatic protein secretion and luminally stimulated pancreatic secretion by 48 % and 52 %, respectively. Intracerebroventricular injection of hemicholinium-3 at doses known to deplete the endogenous ACh store produced similar inhibitory results. In addition, microinjection of ACh (5 pmol) or the muscarinic M1 receptor agonist McN-A-343 (30 ng) into the lateral hypothalamic nucleus increased pancreatic secretion over basal levels by 46 % and 40 %, respectively. Selective lesions of lateral septal cholinergic neurons decreased basal pancreatic secretion and inhibited peptone-induced pancreatic secretion by 30 %. Destruction of the lateral parabrachial nucleus produced a 44 % inhibition of peptone-induced pancreatic section. Finally, microinjection of glutamate into the lateral septum or the lateral parabrachial nucleus stimulated vagal pancreatic efferent nerve firings from a basal level of 0 +/- 0.5 impulses (30 s)-1 to 4.5 +/- 0.5 and 14 +/- 2 impulses (30 s)-1, respectively, and pancreatic protein output increased 50 % and 84 % over basal levels. Administration of MSCP to the paraventricular nucleus eliminated these effects. These observations suggest that cholinergic neurons of the lateral septum and lateral parabrachial nucleus regulate pancreatic secretion. Further, cholinergic input from the lateral parabrachial nucleus to the hypothalamus plays a major role in the modulation of vagal pancreatic efferent nerve activity and pancreatic secretion evoked by the vago-vagal reflex.
Collapse
Affiliation(s)
- Ying Li
- Gastroenterology Research Unit, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|