1
|
Zhang S, Kaiya H, Kitazawa T. Physiological roles of ghrelin in the regulation of gastrointestinal motility in vertebrates. Gen Comp Endocrinol 2025; 365:114698. [PMID: 40024446 DOI: 10.1016/j.ygcen.2025.114698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Ghrelin is known to be a multifunctional peptide hormone that stimulates not only growth hormone secretion and feeding but also gastrointestinal (GI) functions, including motility, secretion and mucosa proliferation. The aim of this review is to provide a comprehensive overview on the physiological roles of ghrelin in the regulation of GI motility from a comparative perspective. The effects of ghrelin on GI motility differ depending on the species, and ghrelin is a possible regulator of gastric migrating motor complexes (MMCs) in rodents, dogs and house musk shrew (suncus). However, the role of ghrelin has not been clarified in detail in other mammals, including humans and rabbits. Ghrelin is also effective to cause contraction in the GI tract of some non-mammals, but its physiological role is also not clarified at present. Distribution of the growth hormone secretagogue receptor (GHSR, ghrelin receptor) in the GI tract might be connected with the regulatory role of ghrelin in vertebrates. Comparative studies of ghrelin among animals and identification of knowledge gaps must lead us to the functional transition and importance of ghrelin in the GI tract.
Collapse
Affiliation(s)
- Shuangyi Zhang
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan; College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Hiroyuki Kaiya
- Grandsoul Research Institute for Immunology, Inc., Uda, Nara 633-2221, Japan; Faculty of Science, University of Toyama, Toyama 930-8555, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
2
|
Abstract
Gastroparesis is a neurogastrointestinal disorder of motility in which patients experience symptoms of nausea, vomiting, bloating, early satiety, postprandial fullness, upper abdominal discomfort or pain, and delayed gastric emptying of solids based on scintigraphy or stable isotope breath test when mechanical obstruction has been excluded. Symptoms of gastroparesis may result from diverse pathophysiological mechanisms, including antroduodenal hypomotility, pylorospasm, increased gastric accommodation, and visceral hypersensitivity. The most common etiologies of gastroparesis are idiopathic, diabetic, and postsurgical, and less frequent causes are neurodegenerative disorders (Parkinson's disease), myopathies (scleroderma, amyloidosis), medication-induced (glucagon-like peptide-1 agonists and opioid agents), and paraneoplastic syndrome. This review addresses pharmacologic management of gastroparesis including prokinetic and antiemetic agents, pharmacologic agents targeting the pylorus, and effects of neuromodulators. SIGNIFICANCE STATEMENT: Gastroparesis is a neurogastrointestinal motility disorder characterized by delayed gastric emptying without mechanical obstruction with numerous upper gastrointestinal symptoms, including nausea and vomiting. The management of gastroparesis involves nutritional support, medications, and procedures. The only Food and Drug Administration-approved medication for gastroparesis is metoclopramide. This article reviews the pharmacology and efficacy of all classes of antiemetics or prokinetic effects used in gastroparesis. There is still a considerable unmet need for efficacious medications specifically for the treatment of gastroparesis, especially in refractory cases.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | - Kara J Jencks
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
3
|
Goudsward HJ, Ruiz-Velasco V, Stella SL, Herold PB, Holmes GM. Ghrelin Modulates Voltage-Gated Ca 2+ Channels through Voltage-Dependent and Voltage-Independent Pathways in Rat Gastric Vagal Afferent Neurons. Mol Pharmacol 2024; 106:253-263. [PMID: 39187389 PMCID: PMC11493335 DOI: 10.1124/molpharm.124.000957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
The orexigenic gut peptide ghrelin is an endogenous ligand for the growth hormone secretagogue receptor type 1a (GHSR1a). Systemic ghrelin administration has previously been shown to increase gastric motility and emptying. While these effects are known to be mediated by the vagus nerve, the cellular mechanism underlying these effects remains unclear. Therefore, the purpose of the present study was to investigate the signaling mechanism by which GHSR1a inhibits voltage-gated Ca2+ channels in isolated rat gastric vagal afferent neurons using whole-cell patch-clamp electrophysiology. The ghrelin pharmacological profile indicated that Ca2+ currents were inhibited with a log (Ic50) = -2.10 ± 0.44 and a maximal inhibition of 42.8 ± 5.0%. Exposure to the GHSR1a receptor antagonist (D-Lys3)-GHRP-6 reduced ghrelin-mediated Ca2+ channel inhibition (29.4 ± 16.7% vs. 1.9 ± 2.5%, n = 6, P = 0.0064). Interestingly, we observed that activation of GHSR1a inhibited Ca2+ currents through both voltage-dependent and voltage-independent pathways. We also treated the gastric neurons with either pertussis toxin (PTX) or YM-254890 to examine whether the Ca2+ current inhibition was mediated by the Gα i/o or Gα q/11 family of subunits. Treatment with both PTX (Ca2+ current inhibition = 15.7 ± 10.6%, n = 8, P = 0.0327) and YM-254890 (15.2 ± 11.9%, n = 8, P = 0.0269) blocked ghrelin's effects on Ca2+ currents, as compared with control neurons (34.3 ± 18.9%, n = 8). These results indicate GHSR1a can couple to both Gα i/o and Gα q/11 in gastric vagal afferent neurons. Overall, our findings suggest GHSR1a-mediated inhibition of Ca2+ currents occurs through two distinct pathways, offering necessary insights into the cellular mechanisms underlying ghrelin's regulation of gastric vagal afferents. SIGNIFICANCE STATEMENT: This study demonstrated that in gastric vagal afferent neurons, activation of GHSR1a by ghrelin inhibits voltage-gated Ca2+ channels through both voltage-dependent and voltage-independent signaling pathways. These results provide necessary insights into the cellular mechanism underlying ghrelin regulation of gastric vagal afferent activity, which may benefit future studies investigating ghrelin mimetics to treat gastric motility disorders.
Collapse
Affiliation(s)
- Hannah J Goudsward
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V., P.B.H.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Victor Ruiz-Velasco
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V., P.B.H.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Salvatore L Stella
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V., P.B.H.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Paul B Herold
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V., P.B.H.), Penn State University College of Medicine, Hershey, Pennsylvania
| | - Gregory M Holmes
- Departments of Neural and Behavioral Sciences (H.J.G., S.L.S., G.M.H.) and Anesthesiology and Perioperative Medicine (V.R.-V., P.B.H.), Penn State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
4
|
Punger EM, Norris SLW, Stevens SC, Santos KH, Christy AC. Investigating the Effect of Enterally Administered Capromorelin on Body Weight in Mice (Mus musculus). Comp Med 2024; 74:327-335. [PMID: 39025662 PMCID: PMC11524401 DOI: 10.30802/aalas-cm-24-031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 04/24/2024] [Accepted: 06/13/2024] [Indexed: 07/20/2024]
Abstract
Significant weight loss in mice (Mus musculus) is a welfare concern and can alter physiology and behavior in ways that may confound research aims. In this study, factorial design was used to investigate the effect of enterally administered capromorelin on changes in mouse body weight overall and with various research-related interventions, such as administration of analgesics, anesthesia, or surgery. BALB/c mice (n = 61 [27 males/34 females] for analysis) were randomized into 8 intervention-treatment groups with 2 treatment allocations: capromorelin (10 mg/kg) or control, and 4 intervention allocations: no intervention; buprenorphine extended-release (XR) alone; buprenorphine XR, meloxicam, and anesthesia; or surgery under anesthesia with buprenorphine XR, meloxicam, and bupivacaine administered. Mice were habituated to handling, weighing, and voluntary consumption of condensed milk, which was used as the control solution and later a vehicle for capromorelin delivery, for 5 d (days 0 to 4). Then, mice received their interventions followed by 3 days of daily treatment or control administration (days 7 to 9). Body weights were measured daily (days 8 to 11 and day 14) to compare with baseline weights (days 0 to 4 and day 7) and evaluate for treatment and intervention effects on body weight. The interventions resulted in a decrease in group body weights 3 and 4 d after the interventions were conducted. Overall, body weights increased more in mice given capromorelin compared with control, and mice treated with capromorelin returned to, or exceeded, baseline weights faster. The weight loss was mitigated by capromorelin administration in all interventions except for the buprenorphine XR-only group. It is recommended to clinically consider enterally administered capromorelin to mitigate research-induced weight loss in mice.
Collapse
Affiliation(s)
- Elizabeth M Punger
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| | - Sarah L W Norris
- Research Support Branch, Regulated Research Administration Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland
| | - Stephen C Stevens
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| | - Kacee H Santos
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| | - Amanda C Christy
- Veterinary Services Branch, Veterinary Medicine Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland; and
| |
Collapse
|
5
|
Potter K, Gayle EJ, Deb S. Effect of gut microbiome on serotonin metabolism: a personalized treatment approach. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2589-2602. [PMID: 37922012 DOI: 10.1007/s00210-023-02762-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/29/2023] [Indexed: 11/05/2023]
Abstract
Several factors including diet, exercise, and medications influence the makeup of the resilient but adaptable gut microbiome. Bacteria in the gut have a significant role in the homeostasis of the neurotransmitter serotonin, also known as 5-hydroxytryptamine, involved in mood and behavior. The goal of the current work is to review the effect of the gut microbiome on serotonin metabolism, and how it can potentially contribute to the development of a personalized treatment approach for depression and anxiety. Bacterial strains provide innovative therapeutic targets that can be used for disorders, such as depression, that involve dysregulation of serotonin. Advances in bacterial genomic sequencing have increased the accessibility and affordability of microbiome testing, which unlocks a new targeted pathway to modulate serotonin metabolism by targeting the gut-brain axis. Microbiome testing can facilitate the recommendation of strain-specific probiotic supplements based on patient-specific microbial profiles. Several studies have shown that supplementation with probiotics containing specific species of bacteria, such as Bifidobacterium and Lactobacillus, can improve symptoms of depression. Further research is needed to improve the process and interpretation of microbiome testing and how to successfully incorporate testing results into guiding clinical decision-making. This targeted approach centered around the gut-brain axis can provide a novel way to personalize therapy for mental health disorders.
Collapse
Affiliation(s)
- Kristal Potter
- College of Pharmacy, Larkin University, 18301 N. Miami Avenue, Miami, FL, 33169, USA
| | - Erysa J Gayle
- College of Biomedical Sciences, Larkin University, 18301 N. Miami Avenue, Miami, FL, 33169, USA
| | - Subrata Deb
- College of Pharmacy, Larkin University, 18301 N. Miami Avenue, Miami, FL, 33169, USA.
| |
Collapse
|
6
|
Akhigbe RE, Adedamola Aminat BO, Akhigbe TM, Hamed MA. Glutamine Alleviates I/R-Induced Intestinal Injury and Dysmotility Via the Downregulation of Xanthine Oxidase/Uric Acid Signaling and Lactate Generation in Wistar Rats. J Surg Res 2024; 295:431-441. [PMID: 38070257 DOI: 10.1016/j.jss.2023.11.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/14/2023] [Accepted: 11/13/2023] [Indexed: 02/25/2024]
Abstract
INTRODUCTION Disruption of intestinal histoarchitecture and intestinal dysmotility is critical to intestinal ischemia/reperfusion (IR) injury and xanthine oxidase (XO)/uric acid (UA) signaling and increased lactate generation have been reported to play a role. More so, glutamine treatment has been demonstrated to inhibit XO/UA signaling. However, the role of glutamine in intestinal IR injury-induced intestinal dysmotility and the associated mechanisms of action are unclear. Therefore, this study was to investigate the mechanisms underlying the role of glutamine in intestinal IR injury. METHODS Forty male Wistar rats were acclimatized for two weeks and then randomized into four groups. The sham-operated, glutamine-treated, intestinal IR, and IR + glutamine groups. RESULTS Glutamine therapy attenuated the IR-induced increase in intestinal weight, disruption of intestinal histoarchitecture, and intestinal dysmotility. In addition, glutamine ameliorated IR-induced intestinal oxidative stress (increased malondialdehyde, reduced glutathione and superoxide dismutase, catalase, glutathione peroxidase, glutathione-S-transferase, and glucose-6-phosphate dehydrogenase activities), inflammation (increased TNF-α and IL-1β), and apoptosis (increased caspase three activity). These events were accompanied by glutamine alleviation of IR-induced upregulation of intestinal nuclear factor kappa B, XO/UA, and lactate generation. CONCLUSIONS In conclusion, XO/UA signaling and lactate levels are key factors in IR-induced intestinal injury and dysmotility, and glutamine-mediated XO/UA/lactate modulation may attenuate IR-induced intestinal injury and dysmotility.
Collapse
Affiliation(s)
- Roland Eghoghosoa Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria
| | | | - Tunmise Maryanne Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Breeding and Plant Genetics Unit, Department of Agronomy, Osun State University, Osun State
| | - Moses Agbomhere Hamed
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Osun State, Nigeria; Department of Medical Laboratory Sciences, Afe Babalola University, Ado Ekiti, Ekiti State, Nigeria; Department of Research and Bioinformatics, The Brainwill Laboratory, Osogbo, Osun State, Nigeria.
| |
Collapse
|
7
|
Zhang S, Kaiya H, Kitazawa T. Does ghrelin regulate intestinal motility in rabbits? An in vitro study using isolated duodenal strips. Gen Comp Endocrinol 2023; 344:114384. [PMID: 37722460 DOI: 10.1016/j.ygcen.2023.114384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023]
Abstract
Rabbit duodenum has been used for examining the ability of motilin to cause muscle contraction in vitro. A motilin-related peptide, ghrelin, is known to be involved in the regulation of gastrointestinal (GI) motility in various animals, but its ability to cause rabbit GI contraction have not been well examined. The aim of this study is to clarify the action of rat ghrelin and its interaction with motilin in the rabbit duodenum. The mRNA expression of ghrelin and motilin receptors was also examined using RT-PCR. Rat ghrelin (10-9-10-6 M) did not change the contractile activity of the duodenum measured by the mean muscle tonus and area under the curve of contraction waves. In agreement with this result, the distribution of ghrelin receptor mRNA in the rabbit GI tract varied depending on the GI region from which the samples were taken; the expression level in the duodenum was negligible, but that in the esophagus or stomach was significant. On the other hand, motilin (10-10-10-6 M) caused a concentration-dependent contraction by means of increased mean muscle tonus, and consistently, motilin receptor mRNA was expressed heterogeneously depending on the GI region (esophagus = stomach = colon = rectum < duodenum = jejunum = ileum < cecum). Expression level of motilin receptor was comparable to that of ghrelin receptor in the esophagus and stomach. Pretreatment with ghrelin (10-6 M) prior to motilin did not affect the contractile activity of motilin in the duodenum. In conclusion, ghrelin does not affect muscle contractility or motilin-induced contraction in the rabbit duodenum, which is due to the lack of ghrelin receptors. The present in vitro results suggest that ghrelin might not be a regulator of intestinal motility in rabbits.
Collapse
Affiliation(s)
- Shuangyi Zhang
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China; School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan; Faculty of Science, University of Toyama, Toyama, Toyama 933-8555, Japan; Grandsoul Research Institute for Immunology, Inc., Uda, Nara 633-2221, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
8
|
Sirajudeen S, Shah I, Karam SM, Al Menhali A. Seven-Month Vitamin D Deficiency Inhibits Gastric Epithelial Cell Proliferation, Stimulates Acid Secretion, and Differentially Alters Cell Lineages in the Gastric Glands. Nutrients 2023; 15:4648. [PMID: 37960302 PMCID: PMC10649607 DOI: 10.3390/nu15214648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Vitamin D (VD) deficiency can result from insufficiency of either light exposure or VD intake. We investigated the biological effects of VD deficiency for 7 months on the mouse gastric glands. Varying degrees of VD deficiency were induced in C57BL/6 mice by keeping them on standard diet with constant-dark conditions (SDD) or VD deficient diet with constant-dark conditions (VDD). Samples of serum, glandular stomach, and gastric contents were collected for LCMS/MS, RT-PCR, immunohistochemistry, and acid content measurements. Both SDD and VDD mice had a significant decline in 25OHVD metabolite, gastric epithelial cell proliferation, and mucin 6 gene expression. These effects were enhanced with the severity of VD deficiency from SDD to VDD. Besides and compared to the control group, SDD mice only displayed a significant increase in the number of zymogenic cells (p ≤ 0.0001) and high expression of the adiponectin (p ≤ 0.05), gastrin (p ≤ 0.0001), mucin 5AC (*** p ≤ 0.001) and the Cyclin-dependent kinase inhibitor 1A (**** p ≤ 0.0001). These phenotypes were unique to SDD gastric samples and not seen in the VDD or control groups. This study suggests that the body reacts differently to diverse VD deficiency sources, light or diet.
Collapse
Affiliation(s)
- Shaima Sirajudeen
- Department of Biology, College of Science, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates;
| | - Iltaf Shah
- Zayed bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates; (I.S.); (S.M.K.)
- Department of Chemistry, College of Science, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates
| | - Sherif M. Karam
- Zayed bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates; (I.S.); (S.M.K.)
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates
| | - Asma Al Menhali
- Department of Biology, College of Science, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates;
- Zayed bin Sultan Al Nahyan Center for Health Sciences, United Arab Emirates University (UAEU), Al Ain 15551, United Arab Emirates; (I.S.); (S.M.K.)
| |
Collapse
|
9
|
Craig CF, Finkelstein DI, McQuade RM, Diwakarla S. Understanding the potential causes of gastrointestinal dysfunctions in multiple system atrophy. Neurobiol Dis 2023; 187:106296. [PMID: 37714308 DOI: 10.1016/j.nbd.2023.106296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023] Open
Abstract
Multiple system atrophy (MSA) is a rare, progressive neurodegenerative disorder characterised by autonomic, pyramidal, parkinsonian and/or cerebellar dysfunction. Autonomic symptoms of MSA include deficits associated with the gastrointestinal (GI) system, such as difficulty swallowing, abdominal pain and bloating, nausea, delayed gastric emptying, and constipation. To date, studies assessing GI dysfunctions in MSA have primarily focused on alterations of the gut microbiome, however growing evidence indicates other structural components of the GI tract, such as the enteric nervous system, the intestinal barrier, GI hormones, and the GI-driven immune response may contribute to MSA-related GI symptoms. Here, we provide an in-depth exploration of the physiological, structural, and immunological changes theorised to underpin GI dysfunction in MSA patients and highlight areas for future research in order to identify more suitable pharmaceutical treatments for GI symptoms in patients with MSA.
Collapse
Affiliation(s)
- Colin F Craig
- Gut Barrier and Disease Laboratory, Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - David I Finkelstein
- Parkinson's Disease Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3052, Australia
| | - Rachel M McQuade
- Gut Barrier and Disease Laboratory, Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE), Sunshine Hospital, St Albans, VIC 3021, Australia
| | - Shanti Diwakarla
- Gut Barrier and Disease Laboratory, Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Australian Institute for Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE), Sunshine Hospital, St Albans, VIC 3021, Australia.
| |
Collapse
|
10
|
Clemente-Suárez VJ, Beltrán-Velasco AI, Redondo-Flórez L, Martín-Rodríguez A, Yáñez-Sepúlveda R, Tornero-Aguilera JF. Neuro-Vulnerability in Energy Metabolism Regulation: A Comprehensive Narrative Review. Nutrients 2023; 15:3106. [PMID: 37513524 PMCID: PMC10383861 DOI: 10.3390/nu15143106] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/09/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
This comprehensive narrative review explores the concept of neuro-vulnerability in energy metabolism regulation and its implications for metabolic disorders. The review highlights the complex interactions among the neural, hormonal, and metabolic pathways involved in the regulation of energy metabolism. The key topics discussed include the role of organs, hormones, and neural circuits in maintaining metabolic balance. The review investigates the association between neuro-vulnerability and metabolic disorders, such as obesity, insulin resistance, and eating disorders, considering genetic, epigenetic, and environmental factors that influence neuro-vulnerability and subsequent metabolic dysregulation. Neuroendocrine interactions and the neural regulation of food intake and energy expenditure are examined, with a focus on the impact of neuro-vulnerability on appetite dysregulation and altered energy expenditure. The role of neuroinflammation in metabolic health and neuro-vulnerability is discussed, emphasizing the bidirectional relationship between metabolic dysregulation and neuroinflammatory processes. This review also evaluates the use of neuroimaging techniques in studying neuro-vulnerability and their potential applications in clinical settings. Furthermore, the association between neuro-vulnerability and eating disorders, as well as its contribution to obesity, is examined. Potential therapeutic interventions targeting neuro-vulnerability, including pharmacological treatments and lifestyle modifications, are reviewed. In conclusion, understanding the concept of neuro-vulnerability in energy metabolism regulation is crucial for addressing metabolic disorders. This review provides valuable insights into the underlying neurobiological mechanisms and their implications for metabolic health. Targeting neuro-vulnerability holds promise for developing innovative strategies in the prevention and treatment of metabolic disorders, ultimately improving metabolic health outcomes.
Collapse
Affiliation(s)
- Vicente Javier Clemente-Suárez
- Faculty of Sports Sciences, Universidad Europea de Madrid, Tajo Street, s/n, 28670 Madrid, Spain
- Grupo de Investigación en Cultura, Educación y Sociedad, Universidad de la Costa, Barranquilla 080002, Colombia
| | | | - Laura Redondo-Flórez
- Department of Health Sciences, Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Tajo Street s/n, 28670 Madrid, Spain
| | | | - Rodrigo Yáñez-Sepúlveda
- Faculty of Education and Social Sciences, Universidad Andres Bello, Viña del Mar 2520000, Chile
| | | |
Collapse
|
11
|
Cangemi DJ, Lacy BE. Gastroparesis: Myths, Misconceptions, and Management. Clin Exp Gastroenterol 2023; 16:65-78. [PMID: 37303313 PMCID: PMC10257400 DOI: 10.2147/ceg.s362879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/29/2023] [Indexed: 06/13/2023] Open
Abstract
Gastroparesis (GP), a historically vexing disorder characterized by symptoms of nausea, vomiting, abdominal pain, early satiety, and/or bloating, in the setting of an objective delay in gastric emptying, is often difficult to treat and carries a tremendous burden on the quality of patients' lives, as well as the healthcare system in general. Though the etiology of GP has been fairly well defined, much work has been done recently to better understand the pathophysiology of GP, as well as to identify novel effective and safe treatment options. As our understanding of GP has evolved, many myths and misconceptions still abound in this rapidly changing field. The goal of this review is to identify myths and misconceptions regarding the etiology, pathophysiology, diagnosis, and treatment of GP, in the context of the latest research findings which have shaped our current understanding of GP. Recognition and dispelling of such myths and misconceptions is critical to moving the field forward and ultimately advancing the clinical management of what will hopefully become a better understood and more manageable disorder in the future.
Collapse
Affiliation(s)
- David J Cangemi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| | - Brian E Lacy
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
12
|
Mori H, Verbeure W, Tanemoto R, Sosoranga ER. Physiological functions and potential clinical applications of motilin. Peptides 2023; 160:170905. [PMID: 36436612 DOI: 10.1016/j.peptides.2022.170905] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022]
Abstract
Motilin is a gastrointestinal hormone secreted by the duodenum. This peptide regulates a characteristic gastrointestinal contraction pattern, called the migrating motor complex, during the fasting state. Motilin also affects the pressure of the lower esophageal sphincter, gastric motility and gastric accommodation in the gastrointestinal tract. Furthermore, motilin induces bile discharge into the duodenum by promoting gallbladder contraction, pepsin secretion in the stomach, pancreatic juice and insulin secretion from the pancreas. In recent years, it has been shown that motilin is associated with appetite, and clinical applications are expected for diseases affected by food intake, e.g. obesity, by regulating motilin levels. Gastric acid and bile are the two major physiological regulators for motilin release. Caloric foods have varying effects on motilin levels, depending on their composition. Among non-caloric foods, bitter substances reduce motilin levels and are therefore expected to have an appetite-suppressing effect. Various motilin receptor agonists and antagonists have been developed but have yet to reach clinical use.
Collapse
Affiliation(s)
- Hideki Mori
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Japan
| | - Wout Verbeure
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Rina Tanemoto
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
13
|
Ghusn W, Cifuentes L, Campos A, Sacoto D, De La Rosa A, Feris F, Calderon G, Gonzalez-Izundegui D, Stutzman J, Hurtado MD, Camilleri M, Acosta A. Association Between Food Intake and Gastrointestinal Symptoms in Patients With Obesity. GASTRO HEP ADVANCES 2022; 2:121-128. [PMID: 36741967 PMCID: PMC9894313 DOI: 10.1016/j.gastha.2022.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/25/2022] [Indexed: 06/18/2023]
Abstract
BACKGROUND AND AIMS Hunger, satiation, postprandial satiety, and hedonic eating constitute key food intake parameters. We aim to study whether these symptoms are associated with gastrointestinal symptoms (GIS) in patients with obesity. METHODS This is a cross-sectional study of patients with obesity. Patients completed the following validated biomarkers and questionnaires: hunger was measured via visual analog scale (100 mm) following a standard meal, satiation was measured via ad libitum meal (calories to fullness; kcal), postprandial satiety was measured via gastric emptying scintigraphy (T1/2; mins), and hedonic eating was measured via the Hospital Anxiety and Depression Scale questionnaire. Participants completed the abridged Bowel Disease Questionnaire to evaluate their GIS. We calculated the odds ratios (ORs) adjusted for sex, weight, and age between food intake parameters <25th or >75th percentile observed in a prior cohort of 450 participants with obesity and GIS. RESULTS A total of 274 participants (41 ± 10 [SD] years, 75% females, body mass index 39 ± 8 kg/m2) were included in the analysis. Increased hunger was associated with a lower prevalence of lumpy stools (OR = 0.18, P = .02). Satiation was associated with abdominal pain/discomfort (relieved by defecation [OR = 2.4, P = .02] or associated with change in stool consistency [OR = 2.92, P < .01]), loose/watery stools (OR = 2.09, P = .02), and bloating (OR = 2.49, P < .01). Abnormal postprandial satiety was associated with bloating (OR = 2.26, P < .01) and loose/watery stools (OR = 1.84, P = .04). Hedonic eating was associated with abdominal pain/discomfort with stool frequency change (OR = 2.4, P = .02), >3 bowel movements per day (OR = 1.93, P = .048), bloating (OR = 2.49, P = .01), abdominal pain after meals >1 per month (OR = 4.24, P < .01), and nausea >1 per week (OR = 4.51, P < .01). CONCLUSION Alterations in hunger, satiation, postprandial satiety, and hedonic eating are associated with GIS in patients with obesity.
Collapse
Affiliation(s)
- Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alejandro Campos
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Daniel Sacoto
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Alan De La Rosa
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Fauzi Feris
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Gerardo Calderon
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Daniel Gonzalez-Izundegui
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jessica Stutzman
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Maria Daniela Hurtado
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiology Research (C.E.N.T.E.R.), Mayo Clinic, Rochester, Minnesota
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
14
|
Role of the Ghrelin System in Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23105380. [PMID: 35628187 PMCID: PMC9141034 DOI: 10.3390/ijms23105380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 02/06/2023] Open
Abstract
The ghrelin system contains several components (e.g., ghrelin with growing number of alternative peptides, growth hormone secretagogue receptors (GHS-Rs), and ghrelin-O-acyl-transferase (GOAT) and participates in regulation of a number of key processes of gastrointestinal (GI) tract cancer progression, including cell proliferation, migration, invasion, apoptosis, inflammation, and angiogenesis. However, its exact role in promoting or inhibiting cancer progression is still unclear. Colorectal cancer (CRC) is one of the most common human malignancies worldwide. Molecular studies suggest an autocrine/paracrine mechanism for the secretion of ghrelin in colorectal carcinogenesis and its contribution to its initial stages. However, the signalling pathways of CRC development involving the ghrelin system are poorly understood. Potential mechanisms of colon carcinogenesis involving components of the ghrelin system were previously described in an animal model and in in vitro studies. However, the diagnostic–prognostic role of serum ghrelin concentrations, tissue expression, or genetic changes of this system in various stages of CRC progression remains an open case. Thus, the aim of this study is to discuss the role of the ghrelin system in colon carcinogenesis, diagnostics and CRC prognostics, as well as the results of studies on the use of ghrelin and its analogues in the therapy of CRC-related syndromes (e.g., cachexia and sarcopenia).
Collapse
|
15
|
Czigle S, Bittner Fialová S, Tóth J, Mučaji P, Nagy M. Treatment of Gastrointestinal Disorders-Plants and Potential Mechanisms of Action of Their Constituents. Molecules 2022; 27:2881. [PMID: 35566230 PMCID: PMC9105531 DOI: 10.3390/molecules27092881] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Revised: 04/19/2022] [Accepted: 04/26/2022] [Indexed: 11/16/2022] Open
Abstract
The worldwide prevalence of gastrointestinal diseases is about 40%, with standard pharmacotherapy being long-lasting and economically challenging. Of the dozens of diseases listed by the Rome IV Foundation criteria, for five of them (heartburn, dyspepsia, nausea and vomiting disorder, constipation, and diarrhoea), treatment with herbals is an official alternative, legislatively supported by the European Medicines Agency (EMA). However, for most plants, the Directive does not require a description of the mechanisms of action, which should be related to the therapeutic effect of the European plant in question. This review article, therefore, summarizes the basic pharmacological knowledge of synthetic drugs used in selected functional gastrointestinal disorders (FGIDs) and correlates them with the constituents of medicinal plants. Therefore, the information presented here is intended as a starting point to support the claim that both empirical folk medicine and current and decades-old treatments with official herbal remedies have a rational basis in modern pharmacology.
Collapse
Affiliation(s)
- Szilvia Czigle
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, SK-832 32 Bratislava, Slovakia; (S.B.F.); (J.T.); (P.M.); (M.N.)
| | | | | | | | | | | |
Collapse
|
16
|
Shinohara Y, Elbadawy M, Yamanaka M, Yamamoto H, Abugomaa A, Usui T, Sasaki K. Effect of the liquid form of traditional Chinese medicine, Hozen-S, on gastric motility in dogs. J Vet Med Sci 2022; 84:841-846. [PMID: 35473800 PMCID: PMC9246680 DOI: 10.1292/jvms.21-0644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Juzen-taiho-to, a traditional Chinese herbal medicine, is used for patients with anorexia
and fatigue in human medicine. In our previous study, granulated Juzen-taiho-to improved
vincristine-induced gastrointestinal adverse effects through increasing gastric motility
in dogs. As the effect of Hozen-S, the sweet liquid form of Juzen-taiho-to, on dog gastric
motility has not been investigated, we examined the effect of administration of Hozen-S on
gastric motility. Furthermore, we assessed dog plasma ghrelin level to further elucidate
the mechanism of the effect of Hozen-S on gastric contraction. Finally, we assessed the
palatability of Hozen-S compared to granulated Juzen-taiho-to and its effect on body
weight in dogs. Administration of Hozen-S significantly increased gastric motility, plasma
ghrelin concentration, and body weight. A palatability evaluation revealed that the dogs
preferred Hozen-S to granulated Juzen-taiho-to. In conclusion, Hozen-S administration to
dogs promoted gastric motility by raising plasma ghrelin levels. Considering these
functional and palatability data, Hozen-S may replace granulated type Juzen-taiho-to and
become a prominent traditional Chinese veterinary medicament.
Collapse
Affiliation(s)
- Yuta Shinohara
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology.,Pet Health & Food Division, Iskara Industry Co., Ltd
| | - Mohamed Elbadawy
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology.,Department of Pharmacology, Faculty of Veterinary Medicine, Benha University
| | - Megumi Yamanaka
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology
| | - Haru Yamamoto
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology
| | - Amira Abugomaa
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology.,Faculty of Veterinary Medicine, Mansoura University
| | - Tatsuya Usui
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology
| | - Kazuaki Sasaki
- Laboratory of Veterinary Pharmacology, Department of Veterinary Medicine, Faculty of Agriculture, Tokyo University of Agriculture and Technology
| |
Collapse
|
17
|
Bukhari SNA. An insight into the multifunctional role of ghrelin and structure activity relationship studies of ghrelin receptor ligands with clinical trials. Eur J Med Chem 2022; 235:114308. [PMID: 35344905 DOI: 10.1016/j.ejmech.2022.114308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 02/06/2022] [Accepted: 03/18/2022] [Indexed: 11/30/2022]
Abstract
Ghrelin is a multifunctional gastrointestinal acylated peptide, primarily synthesized in the stomach and regulates the secretion of growth hormone and energy homeostasis. It plays a central role in modulating the diverse biological, physiological and pathological functions in vertebrates. The synthesis of ghrelin receptor ligands after the finding of growth hormone secretagogue developed from Met-enkephalin led to reveal the endogenous ligand ghrelin and the receptors. Subsequently, many peptides, small molecules and peptidomimetics focusing on the ghrelin receptor, GHS-R1a, were derived. In this review, the key features of ghrelin's structure, forms, its bio-physiological functions, pathological roles and therapeutic potential have been highlighted. A few peptidomimetics and pseudo peptide synthetic perspectives have also been discussed to make ghrelin receptor ligands, clinical trials and their success.
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Aljouf, 2014, Saudi Arabia.
| |
Collapse
|
18
|
Perelló M, Cornejo MP, De Francesco PN, Fernandez G, Gautron L, Valdivia LS. The controversial role of the vagus nerve in mediating ghrelin´s actions: gut feelings and beyond. IBRO Neurosci Rep 2022; 12:228-239. [PMID: 35746965 PMCID: PMC9210457 DOI: 10.1016/j.ibneur.2022.03.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/08/2022] [Accepted: 03/08/2022] [Indexed: 12/26/2022] Open
Abstract
Ghrelin is a stomach-derived peptide hormone that acts via the growth hormone secretagogue receptor (GHSR) and displays a plethora of neuroendocrine, metabolic, autonomic and behavioral actions. It has been proposed that some actions of ghrelin are exerted via the vagus nerve, which provides a bidirectional communication between the central nervous system and peripheral systems. The vagus nerve comprises sensory fibers, which originate from neurons of the nodose and jugular ganglia, and motor fibers, which originate from neurons of the medulla. Many anatomical studies have mapped GHSR expression in vagal sensory or motor neurons. Also, numerous functional studies investigated the role of the vagus nerve mediating specific actions of ghrelin. Here, we critically review the topic and discuss the available evidence supporting, or not, a role for the vagus nerve mediating some specific actions of ghrelin. We conclude that studies using rats have provided the most congruent evidence indicating that the vagus nerve mediates some actions of ghrelin on the digestive and cardiovascular systems, whereas studies in mice resulted in conflicting observations. Even considering exclusively studies performed in rats, the putative role of the vagus nerve in mediating the orexigenic and growth hormone (GH) secretagogue properties of ghrelin remains debated. In humans, studies are still insufficient to draw definitive conclusions regarding the role of the vagus nerve mediating most of the actions of ghrelin. Thus, the extent to which the vagus nerve mediates ghrelin actions, particularly in humans, is still uncertain and likely one of the most intriguing unsolved aspects of the field.
Collapse
|
19
|
Hayakawa S, Tanaka T, Ogawa R, Ito S, Ueno S, Koyama H, Tomotaka O, Sagawa H, Tanaka T, Iwakura H, Takahashi H, Matsuo Y, Mitsui A, Kimura M, Takahashi S, Takiguchi S. Potential Role of TRPV4 in Stretch-Induced Ghrelin Secretion and Obesity. Int J Endocrinol 2022; 2022:7241275. [PMID: 36397882 PMCID: PMC9666045 DOI: 10.1155/2022/7241275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022] Open
Abstract
Obesity is an important health problem, which can be prevented through appetite control. Ghrelin is an appetite-stimulating hormone considered to promote obesity. Thus, we examined whether gastric stretching affects ghrelin secretion. We investigated the role of transient receptor potential vanilloid 4 (TRPV4) in gastric glands in the regulation of ghrelin secretion. TRPV4 immunostaining was performed in tissue samples from 57 patients who underwent gastrectomy. TRPV4 expression was compared between patients with (body mass index (BMI) ≥ 30) and without (BMI <30) obesity. For in vitro experiments, we used MGN3-1 cells, a ghrelin-producing cell line derived from mice. To investigate the bioactivity of TRPV4, MGN3-1 cells were treated with TRPV4 agonists and antagonists, and changes in intracellular Ca2+ concentration were confirmed. The concentration of ghrelin in the cell supernatant was measured using the ELISA with and without 120% stretch stimulation. TRPV4 expression was significantly higher in patients with obesity than in those without at all sites, except the fornix. Immunostaining confirmed the expression of TRPV4 in MGN3-1 cells. TRPV4 agonist administration increased intracellular Ca2+ concentration and ghrelin secretion in MGN3-1 cells, whereas the administration of the agonist combined with the antagonist decreased intracellular Ca2+ concentration and ghrelin secretion. Ghrelin secretion significantly increased in response to a 120% stretch in MGN3-1 cells. However, secretion was not increased by stretch when cells were treated with a TRPV4 antagonist. TRPV4 regulates ghrelin secretion in response to stretch in the stomach, which may affect body weight.
Collapse
Affiliation(s)
- Shunsuke Hayakawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Tatsuya Tanaka
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Sunao Ito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shuhei Ueno
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Hiroyuki Koyama
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Okubo Tomotaka
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Hiroyuki Sagawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Tomohiro Tanaka
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroshi Iwakura
- Department of Pharmacotherapeutics, Wakayama Medical University, Kimiidera, Wakayama, Wakayama, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Akira Mitsui
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Masahiro Kimura
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| |
Collapse
|
20
|
Deschaine SL, Leggio L. From "Hunger Hormone" to "It's Complicated": Ghrelin Beyond Feeding Control. Physiology (Bethesda) 2022; 37:5-15. [PMID: 34964687 PMCID: PMC8742734 DOI: 10.1152/physiol.00024.2021] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Discovered as a peptide involved in releasing growth hormone, ghrelin was initially characterized as the "hunger hormone." However, emerging research indicates that ghrelin appears to play an important part in relaying information regarding nutrient availability and value and adjusting physiological and motivational processes accordingly. These functions make ghrelin an interesting therapeutic candidate for metabolic and neuropsychiatric diseases involving disrupted nutrition that can further potentiate the rewarding effect of maladaptive behaviors.
Collapse
Affiliation(s)
- Sara L. Deschaine
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland
| | - Lorenzo Leggio
- 1Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Baltimore and Bethesda, Maryland,2Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, Maryland,3Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, School of Public Health, Brown University, Providence, Rhode Island,4Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, Maryland,5Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia
| |
Collapse
|
21
|
Kärkkäinen O, Farokhnia M, Klåvus A, Auriola S, Lehtonen M, Deschaine SL, Piacentino D, Abshire KM, Jackson SN, Leggio L. Effect of intravenous ghrelin administration, combined with alcohol, on circulating metabolome in heavy drinking individuals with alcohol use disorder. Alcohol Clin Exp Res 2021; 45:2207-2216. [PMID: 34590334 PMCID: PMC8642277 DOI: 10.1111/acer.14719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 08/30/2021] [Accepted: 09/14/2021] [Indexed: 12/11/2022]
Abstract
BACKGROUND Ghrelin may influence several alcohol-related behaviors in animals and humans by modulating central and/or peripheral biological pathways. The aim of this exploratory analysis was to investigate associations between ghrelin administration and the human circulating metabolome during alcohol exposure in nontreatment seeking, heavy drinking individuals with alcohol use disorder (AUD). METHODS We used serum samples from a randomized, crossover, double-blind, placebo-controlled human laboratory study with intravenous (IV) ghrelin or placebo infusion in two experiments. During each session, participants received a loading dose (3 µg/kg) followed by continuous infusion (16.9 ng/kg/min) of acyl ghrelin or placebo. The first experiment included an IV alcohol self-administration (IV-ASA) session and the second experiment included an IV alcohol clamp (IV-AC) session, both with the counterbalanced infusion of ghrelin or placebo. Serum metabolite profiles were analyzed from repeated blood samples collected during each session. RESULTS In both experiments, ghrelin infusion was associated with an altered serum metabolite profile, including significantly increased levels of cortisol (IV-ASA q-value = 0.0003 and IV-AC q < 0.0001), corticosterone (IV-ASA q = 0.0202 and IV-AC q < 0.0001), and glycochenodeoxycholic acid (IV-ASA q = 0.0375 and IV-AC q = 0.0013). In the IV-ASA experiment, ghrelin infusion increased levels of cortisone (q = 0.0352) and fatty acids 18:1 (q = 0.0406) and 18:3 (q = 0.0320). Moreover, in the IV-AC experiment, ghrelin infusion significantly increased levels of glycocholic acid (q < 0.0001) and phenylalanine (q = 0.0458). CONCLUSION IV ghrelin infusion, combined with IV alcohol administration, was associated with increases in the circulating metabolite levels of corticosteroids and glycine-conjugated bile acids, among other changes. Further research is needed to understand the role that metabolomic changes play in the complex interaction between ghrelin and alcohol.
Collapse
Affiliation(s)
- Olli Kärkkäinen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Mehdi Farokhnia
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, Baltimore and Bethesda, Maryland, USA
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Anton Klåvus
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Seppo Auriola
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Marko Lehtonen
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| | - Sara L. Deschaine
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, Baltimore and Bethesda, Maryland, USA
| | - Daria Piacentino
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, Baltimore and Bethesda, Maryland, USA
- Center on Compulsive Behaviors, National Institutes of Health, Bethesda, MD, USA
| | - Kelly M. Abshire
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, Baltimore and Bethesda, Maryland, USA
| | - Shelley N. Jackson
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland, USA
| | - Lorenzo Leggio
- Clinical Psychoneuroendocrinology and Neuropsychopharmacology Section, Translational Addiction Medicine Branch, National Institute on Drug Abuse Intramural Research Program and National Institute on Alcohol Abuse and Alcoholism Division of Intramural Clinical and Biological Research, Baltimore and Bethesda, Maryland, USA
- Center on Compulsive Behaviors, National Institutes of Health, Bethesda, MD, USA
- Translational Analytical Core, National Institute on Drug Abuse Intramural Research Program, Baltimore, Maryland, USA
- Medication Development Program, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
- Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, Providence, RI, USA
- Division of Addiction Medicine, Department of Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
22
|
Camilleri M, Atieh J. New Developments in Prokinetic Therapy for Gastric Motility Disorders. Front Pharmacol 2021; 12:711500. [PMID: 34504426 PMCID: PMC8421525 DOI: 10.3389/fphar.2021.711500] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022] Open
Abstract
Prokinetic agents amplify and coordinate the gastrointestinal muscular contractions to facilitate the transit of intra-luminal content. Following the institution of dietary recommendations, prokinetics are the first medications whose goal is to improve gastric emptying and relieve symptoms of gastroparesis. The recommended use of metoclopramide, the only currently approved medication for gastroparesis in the United States, is for a duration of less than 3 months, due to the risk of reversible or irreversible extrapyramidal tremors. Domperidone, a dopamine D2 receptor antagonist, is available for prescription through the FDA's program for Expanded Access to Investigational Drugs. Macrolides are used off label and are associated with tachyphylaxis and variable duration of efficacy. Aprepitant relieves some symptoms of gastroparesis. There are newer agents in the pipeline targeting diverse gastric (fundic, antral and pyloric) motor functions, including novel serotonergic 5-HT4 agonists, dopaminergic D2/3 antagonists, neurokinin NK1 antagonists, and ghrelin agonist. Novel targets with potential to improve gastric motor functions include the pylorus, macrophage/inflammatory function, oxidative stress, and neurogenesis. In the current review, we discuss the use of pharmacological approaches with potential to enhance motor functions in the management of gastroparesis.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research (CENTER), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | | |
Collapse
|
23
|
Kitazawa T, Kaiya H. Motilin Comparative Study: Structure, Distribution, Receptors, and Gastrointestinal Motility. Front Endocrinol (Lausanne) 2021; 12:700884. [PMID: 34497583 PMCID: PMC8419268 DOI: 10.3389/fendo.2021.700884] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/16/2021] [Indexed: 12/26/2022] Open
Abstract
Motilin, produced in endocrine cells in the mucosa of the upper intestine, is an important regulator of gastrointestinal (GI) motility and mediates the phase III of interdigestive migrating motor complex (MMC) in the stomach of humans, dogs and house musk shrews through the specific motilin receptor (MLN-R). Motilin-induced MMC contributes to the maintenance of normal GI functions and transmits a hunger signal from the stomach to the brain. Motilin has been identified in various mammals, but the physiological roles of motilin in regulating GI motility in these mammals are well not understood due to inconsistencies between studies conducted on different species using a range of experimental conditions. Motilin orthologs have been identified in non-mammalian vertebrates, and the sequence of avian motilin is relatively close to that of mammals, but reptile, amphibian and fish motilins show distinctive different sequences. The MLN-R has also been identified in mammals and non-mammalian vertebrates, and can be divided into two main groups: mammal/bird/reptile/amphibian clade and fish clade. Almost 50 years have passed since discovery of motilin, here we reviewed the structure, distribution, receptor and the GI motility regulatory function of motilin in vertebrates from fish to mammals.
Collapse
Affiliation(s)
- Takio Kitazawa
- Comparative Animal Pharmacology, Department of Veterinary Science, Rakuno Gakuen University, Ebetsu, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Japan
| |
Collapse
|
24
|
Carbone F, Vanuytsel T, Tack J. The effect of prucalopride on gastric sensorimotor function and satiation in healthy volunteers. Neurogastroenterol Motil 2021; 33:e14083. [PMID: 33615630 DOI: 10.1111/nmo.14083] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Gastric motor function alterations have been implicated in the pathogenesis of functional dyspepsia with postprandial distress syndrome (PDS). Prucalopride, a 5-TH4 agonist, is known to stimulate gastrointestinal motility. We aimed to evaluate the effect of prucalopride on gastric sensorimotor function in healthy subjects (HV). METHODS Barostat and intragastric pressure (IGP) measurements were performed in 17 HV (59% females, age 29.4 ± 2.7 y) after treatment with placebo or prucalopride (2 mg) (single-blind cross-over). Isobaric stepwise distensions and gastric sensations were assessed to determine gastric compliance and sensitivity. Gastric accommodation (GA) with the barostat was quantified before and after ingestion of 200 ml of a nutrient drink (ND). GA measured by IGP was quantified as the drop of IGP from baseline during the intragastric infusion of ND until maximal satiation (60 ml/min). KEY RESULTS Prucalopride did not affect barostat assessed gastric compliance or sensitivity. No differences were observed in GA after prucalopride. During the barostat study, 10 min after the meal, 7 HVs reported significantly higher ratings for nausea after prucalopride (p < 0.001), and vomiting was induced in 4 of the HVs. A positive correlation was observed between the delta mean perception of nausea with the delta mean increase of intra-balloon volume before and after meal ingestion (r = 0.37, p = 0.03). During IGP measurements, no effect on nutrient tolerance was observed and increased cramp severity scores were observed which were associated with a significant increase of distal IGP (r = 0.78, p < 0.0001). CONCLUSIONS & INFERENCES Prucalopride does not enhances gastric accommodation but it might increase sensitivity to gastric distention. Furthermore, the increase in sensitivity seems to be related to an increase in nausea with distension. Clinicaltrials.gov: NCT04429802.
Collapse
Affiliation(s)
- Florencia Carbone
- Translational Research Center for Gastrointestinal Disorders (TARGID, KULeuven, Leuven, Belgium
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders (TARGID, KULeuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID, KULeuven, Leuven, Belgium
| |
Collapse
|
25
|
Verbeure W, Deloose E, Tóth J, Rehfeld JF, Van Oudenhove L, Depoortere I, Tack J. The endocrine effects of bitter tastant administration in the gastrointestinal system: intragastric versus intraduodenal administration. Am J Physiol Endocrinol Metab 2021; 321:E1-E10. [PMID: 34029163 DOI: 10.1152/ajpendo.00636.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Bitter tastants are recently introduced as potential hunger-suppressive compounds, the so-called "Bitter pill." However, the literature about bitter administration lacks consistency in methods and findings. We want to test whether hunger ratings and hormone plasma levels are affected by: 1) the site of administration: intragastrically (IG) or intraduodenally (ID), 2) the bitter tastant itself, quinine hydrochloride (QHCl) or denatonium benzoate (DB), and 3) the timing of infusion. Therefore, 14 healthy, female volunteers participated in a randomized, placebo-controlled six-visit crossover study. After an overnight fast, DB (1 µmol/kg), QHCl (10 µmol/kg), or placebo were given IG or ID via a nasogastric feeding tube. Blood samples were taken 10 min before administration and every 10 min after administration for a period of 2 h. Hunger was rated at the same time points on a visual analogue scale. ID bitter administration did not affect hunger sensations, motilin, or acyl-ghrelin release compared with its placebo infusion. IG QHCl infusion tended to suppress hunger increase, especially between 50 and 70 min after infusion, simultaneously with reduced motilin values. Here, acyl-ghrelin was not affected. IG DB did not affect hunger or motilin, however acyl-ghrelin levels were reduced 50-70 minutes after infusion. Plasma values of glucagon-like peptide 1 and cholecystokinin were too low to be properly detected or to have any physiological relevance. In conclusion, bitter tastants should be infused into the stomach to reduce hunger sensations and orexigenic gut peptides. QHCl has the best potential to reduce hunger sensations, and it should be infused 60 min before food intake.NEW & NOTEWORTHY Bitter tastants are a potential new weight-loss treatment. This is a noninvasive, easy approach, which should be received with considerable enthusiasm by the public. However, literature about bitter administration lacks consistency in methods and findings. We summarize how the compound should be given based on: the site of administration, the best bitter compound to use, and at what timing in respect to the meal. This paper is therefore a fundamental step to continue research toward the further development of the "bitter pill."
Collapse
Affiliation(s)
- Wout Verbeure
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Eveline Deloose
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Joran Tóth
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lukas Van Oudenhove
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Inge Depoortere
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, KU Leuven, Leuven, Belgium
| |
Collapse
|
26
|
Tack J, Verbeure W, Mori H, Schol J, Van den Houte K, Huang IH, Balsiger L, Broeders B, Colomier E, Scarpellini E, Carbone F. The gastrointestinal tract in hunger and satiety signalling. United European Gastroenterol J 2021; 9:727-734. [PMID: 34153172 PMCID: PMC8280794 DOI: 10.1002/ueg2.12097] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/18/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Different peripheral pathways are implicated in the regulation of the food ingestion-digestion cycle. METHODS Narrative review on gastrointestinal mechanisms involved in satiety and hunger signalling. RESULTS Combined mechano- and chemoreceptors, peripherally released peptide hormones and neural pathways provide feedback to the brain to determine sensations of hunger (increase energy intake) or satiation (cessation of energy intake) and regulate the human metabolism. The gastric accommodation reflex, which consists of a transient relaxation of the proximal stomach during food intake, has been identified as a major determinant of meal volume, through activation of tension-sensitive gastric mechanoreceptors. Motilin, whose release is the trigger of gastric Phase 3, has been identified as the major determinant of return of hunger after a meal. In addition, the release of several peptide hormones such as glucagon-like peptide 1 (GLP-1), cholecystokinin as well as motilin and ghrelin contributes to gut-brain signalling with relevance to control of hunger and satiety. A number of nutrients, such as bitter tastants, as well as pharmacological agents, such as endocannabinoid receptor antagonists and GLP-1 analogues act on these pathways to influence hunger, satiation and food intake. CONCLUSION Gastrointestinal mechanisms such as gastric accommodation and motilin release are key determinants of satiety and hunger.
Collapse
Affiliation(s)
- Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Wout Verbeure
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Hideki Mori
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Jolien Schol
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Karen Van den Houte
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - I-Hsuan Huang
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Lukas Balsiger
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Bert Broeders
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Esther Colomier
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Emidio Scarpellini
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Florencia Carbone
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| |
Collapse
|
27
|
Guo X, Lv J, Xi R. The specification and function of enteroendocrine cells in Drosophila and mammals: a comparative review. FEBS J 2021; 289:4773-4796. [PMID: 34115929 DOI: 10.1111/febs.16067] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/26/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
Enteroendocrine cells (EECs) in both invertebrates and vertebrates derive from intestinal stem cells (ISCs) and are scattered along the digestive tract, where they function in sensing various environmental stimuli and subsequently secrete neurotransmitters or neuropeptides to regulate diverse biological and physiological processes. To fulfill these functions, EECs are specified into multiple subtypes that occupy specific gut regions. With advances in single-cell technology, organoid culture experimental systems, and CRISPR/Cas9-mediated genomic editing, rapid progress has been made toward characterization of EEC subtypes in mammals. Additionally, studies of genetic model organisms-especially Drosophila melanogaster-have also provided insights about the molecular processes underlying EEC specification from ISCs and about the establishment of diverse EEC subtypes. In this review, we compare the regulation of EEC specification and function in mammals and Drosophila, with a focus on EEC subtype characterization, on how internal and external regulators mediate EEC subtype specification, and on how EEC-mediated intra- and interorgan communications affect gastrointestinal physiology and pathology.
Collapse
Affiliation(s)
- Xingting Guo
- National Institute of Biological Sciences, Beijing, China
| | - Jiaying Lv
- National Institute of Biological Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Rongwen Xi
- National Institute of Biological Sciences, Beijing, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| |
Collapse
|
28
|
Kim SK, Joung JY, Ahn YC, Jung IC, Son CG. Beneficial Potential of Banha-Sasim-Tang for Stress-Sensitive Functional Dyspepsia via Modulation of Ghrelin: A Randomized Controlled Trial. Front Pharmacol 2021; 12:636752. [PMID: 33959008 PMCID: PMC8093827 DOI: 10.3389/fphar.2021.636752] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Functional dyspepsia (FD) is a highly complex pathophysiologic disease, which shows low recovery and high relapse rates. Therefore, a growing number of clinicians and patients are looking for alternative herbal medicine. Banha-Sasim-Tang (BST) is an herbal prescription for treating a wide range of dyspepsia in traditional Korean medicine (TKM). Aim of the study: This study aimed to evaluate the efficacy of BST on FD in patients with psychological stress-related symptoms and to investigate the involvement of ghrelin. This study is registered at https://cris.nih.go.kr/with the identification number KCT 0002811. Materials and methods: A total of 31 participants with FD who met the Rome IV criteria and the psychological stress-related pattern were enrolled in a double-blind, randomized, and controlled study. Participants were randomly assigned to the BST group (10 g twice daily for 4 weeks) or placebo group. The primary endpoint was a change in the Nepean Dyspepsia Index-Korean (NDI-K) score. The secondary endpoints were changes in the Visual Analog Scale (VAS) scores and plasma ghrelin level. Results: All participants completed the study (n = 15 for BST, n = 16 for placebo). BST decreased NDI-K scores compared with placebo, but the difference was not statistically significant (37.40 ± 27.40 vs 22.50 ± 23.85, p = 0.12). VAS scores and plasma total ghrelin levels were significantly improved in patients who were treated with BST (3.19 ± 1.60 vs 1.38 ± 2.85, p = 0.03 for VAS and 105.69 ± 287.89 vs -142.31 ± 314.32, p = 0.03 for total ghrelin). No BST-related adverse effects were observed during the trial. Conclusion: Our results indicate the clinical potential of BST for FD patients and are the first study to show the modulation of plasma ghrelin as one of its corresponding mechanisms. Clinical Trial Registration: https://cris.nih.go.kr/, identifier KCT 0002811.
Collapse
Affiliation(s)
- Sul-Ki Kim
- Liver and Immunology Research Center, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Jin-Yong Joung
- Liver and Immunology Research Center, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Yo-Chan Ahn
- Department of Health Service Management, Daejeon University, Daejeon, Republic of Korea
| | - In-Chul Jung
- Departments of Neuropsychiatry, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| | - Chang-Gue Son
- Liver and Immunology Research Center, College of Korean Medicine, Daejeon University, Daejeon, Republic of Korea
| |
Collapse
|
29
|
Nunez‐Salces M, Li H, Feinle‐Bisset C, Young RL, Page AJ. The regulation of gastric ghrelin secretion. Acta Physiol (Oxf) 2021; 231:e13588. [PMID: 33249751 DOI: 10.1111/apha.13588] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/19/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022]
Abstract
Ghrelin is a gastric hormone with multiple physiological functions, including the stimulation of food intake and adiposity. It is well established that circulating ghrelin levels are closely associated with feeding patterns, rising strongly before a meal and lowering upon food intake. However, the mechanisms underlying the modulation of ghrelin secretion are not fully understood. The purpose of this review is to discuss current knowledge on the circadian oscillation of circulating ghrelin levels, the neural mechanisms stimulating fasting ghrelin levels and peripheral mechanisms modulating postprandial ghrelin levels. Furthermore, the therapeutic potential of targeting the ghrelin pathway is discussed in the context of the treatment of various metabolic disorders, including obesity, type 2 diabetes, diabetic gastroparesis and Prader-Willi syndrome. Moreover, eating disorders including anorexia nervosa, bulimia nervosa and binge-eating disorder are also discussed.
Collapse
Affiliation(s)
- Maria Nunez‐Salces
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| | - Hui Li
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| | - Christine Feinle‐Bisset
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Richard L. Young
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
- Intestinal Nutrient Sensing Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Amanda J. Page
- Vagal Afferent Research Group Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Centre of Research Excellence in Translating Nutritional Science to Good Health Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute (SAHMRI) Adelaide SA Australia
| |
Collapse
|
30
|
Seidel M, Markmann Jensen S, Healy D, Dureja A, Watson HJ, Holst B, Bulik CM, Sjögren JM. A Systematic Review and Meta-Analysis Finds Increased Blood Levels of All Forms of Ghrelin in Both Restricting and Binge-Eating/Purging Subtypes of Anorexia Nervosa. Nutrients 2021; 13:nu13020709. [PMID: 33672297 PMCID: PMC7926807 DOI: 10.3390/nu13020709] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/16/2022] Open
Abstract
Anorexia nervosa (AN) is a severe psychiatric condition associated with high mortality and chronicity. The hunt for state, trait, subtyping, and prognostic biomarkers is ongoing and the orexigenic hormone ghrelin and its different forms, acyl ghrelin and desacyl ghrelin, have been proposed to be increased in AN, especially in the restrictive subtype. A systematic literature search was performed using established databases up to 30 November 2020. Forty-nine studies met inclusion criteria for cross-sectional and longitudinal meta-analyses on total ghrelin, acyl ghrelin, and desacyl ghrelin. All forms of ghrelin were increased in the acute stage of anorexia nervosa during fasting compared to healthy controls. Previous notions on differences in ghrelin levels between AN subtypes were not supported by current data. In addition, a significant decrease in total ghrelin was observed pre-treatment to follow-up. However, total ghrelin levels at follow-up were still marginally elevated compared to healthy controls, whereas for acyl ghrelin, no overall effect of treatment was observed. Due to heterogeneity in follow-up designs and only few data on long-term recovered patients, longitudinal results should be interpreted with caution. While the first steps towards a biomarker in acute AN have been completed, the value of ghrelin as a potential indicator of treatment success or recovery status or its use in subtype differentiation are yet to be established.
Collapse
Affiliation(s)
- Maria Seidel
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 171 65 Solna, Sweden; (M.S.); (C.M.B.)
- Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, Technische Universität Dresden, 1099 Dresden, Germany
| | - Signe Markmann Jensen
- Research Unit Eating Disorders, Psychiatric Center Ballerup, Maglevænget 32, 2750 Ballerup, Denmark; (S.M.J.); (D.H.); (A.D.)
| | - Darren Healy
- Research Unit Eating Disorders, Psychiatric Center Ballerup, Maglevænget 32, 2750 Ballerup, Denmark; (S.M.J.); (D.H.); (A.D.)
| | - Aakriti Dureja
- Research Unit Eating Disorders, Psychiatric Center Ballerup, Maglevænget 32, 2750 Ballerup, Denmark; (S.M.J.); (D.H.); (A.D.)
| | - Hunna J. Watson
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- School of Psychology, Curtin University, Perth U1987, Australia
- Division of Paediatrics, University of Western Australia, Perth 6907, Australia
| | - Birgitte Holst
- Department of Biomedical Sciences, University of Copenhagen, 1353 Copenhagen, Denmark;
| | - Cynthia M. Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, 171 65 Solna, Sweden; (M.S.); (C.M.B.)
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jan Magnus Sjögren
- Research Unit Eating Disorders, Psychiatric Center Ballerup, Maglevænget 32, 2750 Ballerup, Denmark; (S.M.J.); (D.H.); (A.D.)
- Department of Clinical Medicine, University of Copenhagen, 2200 N Copenhagen, Denmark
- Correspondence:
| |
Collapse
|
31
|
Petri M, Singh I, Baker C, Underkofler C, Rasouli N. Diabetic gastroparesis: An overview of pathogenesis, clinical presentation and novel therapies, with a focus on ghrelin receptor agonists. J Diabetes Complications 2021; 35:107733. [PMID: 32948398 DOI: 10.1016/j.jdiacomp.2020.107733] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 08/29/2020] [Accepted: 08/29/2020] [Indexed: 12/19/2022]
Abstract
Diabetic gastroparesis is defined as delayed gastric emptying without mechanical obstruction in the setting of diabetes. Symptoms range from mild bloating to severe vomiting episodes and can result in frequent hospitalizations and poor quality of life. It is suspected that diabetic gastroparesis is underdiagnosed due to its similar presentation to other conditions such as gastroesophageal reflux disease. The pathogenesis of diabetic gastroparesis remains unclear, but proposed mechanisms include vagal dysfunction, hyperglycemia, interstitial cells of Cajal network disturbances, loss of neural nitric oxide synthase expression in the myenteric plexus, and oxidative stress. Current management for diabetic gastroparesis focuses on dietary and lifestyle changes as well as improved glycemic control. Limited options for medical therapies are available that include prokinetic and antiemetic medications. Metoclopramide is the only FDA-approved medication for the treatment of gastroparesis. Metoclopramide improves symptoms of gastroparesis although extended treatment presents challenges such as decreased efficacy over time and increased risks for adverse events. We summarize the current knowledge of the pathophysiology of diabetic gastroparesis and review current and investigational treatments for diabetes gastroparesis.
Collapse
Affiliation(s)
- Madison Petri
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12401 East 17th Avenue, Aurora, CO, USA
| | - Inderpreet Singh
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12401 East 17th Avenue, Aurora, CO, USA
| | - Chelsea Baker
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12401 East 17th Avenue, Aurora, CO, USA
| | - Chantal Underkofler
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12401 East 17th Avenue, Aurora, CO, USA
| | - Neda Rasouli
- Department of Medicine, University of Colorado Anschutz Medical Campus, 12401 East 17th Avenue, Aurora, CO, USA.
| |
Collapse
|
32
|
Zhang S, Teraoka H, Kaiya H, Kitazawa T. Motilin- and ghrelin-induced contractions in isolated gastrointestinal strips from three species of frogs. Gen Comp Endocrinol 2021; 300:113649. [PMID: 33153968 DOI: 10.1016/j.ygcen.2020.113649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/07/2020] [Accepted: 10/17/2020] [Indexed: 12/26/2022]
Abstract
Ghrelin (GHRL) and motilin (MLN), gut peptides isolated from the mucosa of the stomach and duodenum, respectively, stimulate gastrointestinal (GI) motility in mammals and birds. However, the functions of MLN and GHRL in amphibian GI tracts have not been examined in detail. To clarify the regulation of GI motility by the two peptides, the effects of human MLN and rat GHRL on contractility of isolated GI strips from three species of frogs, the black-spotted pond frog (pond frog; Pelophylax nigromaculata), bullfrog (Lithobates catesbeiana) and Western clawed frog (Xenopus; Xenopus tropicalis), were examined in in vitro experiments. The GI tract of each frog was divided into the stomach, upper intestine, middle intestine and lower intestine. Human MLN caused contractions of the stomach in the pond frog and upper intestine in the bullfrog and Xenopus, but other GI regions were insensitive to human MLN. Erythromycin did not cause contraction of the upper intestine of the bullfrog and Xenopus. Rat GHRL did not cause contraction of the stomach and small intestines in the pond frog and bullfrog, but it caused a concentration-dependent contraction in the stomach and upper intestine of Xenopus, while des-acyl rat GHRL did not cause any contraction of them. In conclusion, human MLN caused the contraction of the stomach or upper intestine in the three species of frogs, but GHRL was effective only in the stomach and upper intestine of Xenopus. On the basis of these data, MLN but not GHRL causes the GI region-dependent contractions in the frogs.
Collapse
Affiliation(s)
- Shuangyi Zhang
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroki Teraoka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 564-8565, Japan
| | - Takio Kitazawa
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
33
|
Tsuji N, Umehara Y, Takenaka M, Minami Y, Watanabe T, Nishida N, Kudo M. Verrucous antral gastritis in relation to Helicobacter pylori infection, nutrition, and gastric atrophy. Gastroenterol Rep (Oxf) 2020; 8:293-298. [PMID: 32843976 PMCID: PMC7434579 DOI: 10.1093/gastro/goz057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/28/2019] [Accepted: 09/29/2019] [Indexed: 11/14/2022] Open
Abstract
Background There have been few studies in the English literature regarding verrucous gastritis (VG). The present study investigated the clinical and endoscopic features of verrucous antral gastritis, especially focusing on Helicobacter pylori infection, nutrition, and gastric atrophy. Methods We performed a retrospective study of patients who underwent routine endoscopy with indigo carmine chromoendoscopy and a comparative study was conducted between VG-positive and VG-negative groups. VG was subdivided into classical and numerous types based on the number and distribution of verrucous lesions. Demographic, clinical, and endoscopic data including body mass index (BMI), serum albumin and cholesterol, gastric atrophy, reflux oesophagitis, Barrett's oesophagus, and H. pylori status were collected. Univariate and multivariable analyses were performed to identify factors associated with VG. Results We analysed the data of 621 patients undergoing routine endoscopy and found that VG (n = 352) was significantly associated with increased BMI (1.12 [1.05-1.18], P < 0.01), reflux esophagitis (1.96 [1.10-3.28], P < 0.01), and H. pylori negativity with or without a history of eradication (9.94 [6.00-16.47] and 6.12 [3.51-10.68], P < 0.001, respectively). Numerous-type (n = 163) VG was associated with both closed- and open-type gastric atrophy (9.9 [4.04-21.37] and 8.10 [3.41-19.24], P < 0.001, respectively). There were no statistical differences between groups regarding age, sex, total cholesterol, albumin, and bile-colored gastric juice. Conclusions Verrucous antral gastritis was related to increased BMI, reflux esophagitis, and H. pylori negativity. Numerous-type verrucous lesions were associated with gastric atrophy. These indicate that VG may be a physiological phenomenon due to high gastric acidity, mechanical overload, and vulnerability of background mucosa.
Collapse
Affiliation(s)
- Naoko Tsuji
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasuko Umehara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Mamoru Takenaka
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasunori Minami
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Tomohiro Watanabe
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka-Sayama, Japan
| |
Collapse
|
34
|
Pietraszko W, Furgala A, Gorecka-Mazur A, Kwinta B, Kaszuba-Zwoinska J, Polak J, Fiszer U, Gil K, Krygowska-Wajs A. Assessments of plasma acyl-ghrelin levels in Parkinson's disease patients treated with deep brain stimulation. Peptides 2020; 128:170299. [PMID: 32305796 DOI: 10.1016/j.peptides.2020.170299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/26/2020] [Accepted: 03/12/2020] [Indexed: 12/26/2022]
Abstract
Gastrointestinal dysfunction is the most common non-motor symptom in Parkinson's disease (PD) with rates rising as the disease progresses. Deep brain stimulation of subthalamic nucleus (STN DBS) improves motor functions in advanced PD. However, the effect of STN DBS on ghrelin concentration and consequently on motility disturbances as well as body weight is unclear. The objective of this study was to assess acyl-ghrelin levels in comparison to weight in advanced PD patients treated with STN DBS. Plasma concentrations of acyl-ghrelin was measured in 29 PD patients in the fasting state and at 30, 60, 120, and 180 min after a standard meal preoperatively and 3 months after surgery. The level of acyl-ghrelin in PD patients were compared with 30 age and sex-matched healthy controls. We reported that mean plasma acyl-ghrelin levels were decreased in PD patients before STN DBS in fasting (p = 0.0003) and in 30 min postprandial phase (p = 0.04) compared with healthy controls. The plasma acyl-ghrelin levels after STN DBS increased in pre-prandial and postprandial phase in PD patients at the investigated time points. Body weight gained on average 2.33 kg during the first 3 months after surgery. There was no correlation between the acyl-ghrelin plasma levels and BMI. After STN DBS in fasting and postprandial phase plasma acyl-ghrelin levels were increased. The results showed that STN DBS therapy elicited a modification of ghrelin levels, increasing its concentration in pre- and postprandial state. In addition, body weight was increased during 3 months after surgery.
Collapse
Affiliation(s)
- Wojciech Pietraszko
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Agata Furgala
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Agnieszka Gorecka-Mazur
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Borys Kwinta
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Jolanta Kaszuba-Zwoinska
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Jaroslaw Polak
- Department of Neurosurgery, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland
| | - Urszula Fiszer
- Department of Neurology and Epileptology, Centre of Postgraduate Medical Education, Warsaw, Czerniakowska 231, Poland
| | - Krzysztof Gil
- Department of Pathophysiology, Jagiellonian University, Medical College, Krakow, Czysta 18, Poland
| | - Anna Krygowska-Wajs
- Department of Neurology, Jagiellonian University, Medical College, Krakow, Botaniczna 3, Poland.
| |
Collapse
|
35
|
Abstract
Gastroparesis is characterized by delayed gastric emptying, with symptoms such as nausea, vomiting and abdominal pain, in the absence of mechanical obstruction. In most cases, it is idiopathic although diabetes mellitus is another leading cause. The physiology of gastric emptying is a complex process which is influenced by various inputs including the central nervous system, enteric nervous system and gut hormones. Developments in our understanding of gastroparesis have now demonstrated dysfunction in these systems, thus disrupting normal gastric emptying. Once mechanical obstruction is excluded, gastric scintigraphy remains the gold standard for diagnosis although wireless motility capsule and breath testing are alternative methods for diagnosis. Treatment for gastroparesis is challenging, and widely available therapies are often limited either by their poor evidence for efficacy or concerns over their long-term safety profile. Novel prokinetic agents have shown initial promise in clinical trials, and new endoscopic techniques such as gastric per-oral endoscopic myotomy are emerging. These new treatment modalities may provide an option in refractory gastroparesis with the adage of reduced morbidity compared to surgical treatments.
Collapse
Affiliation(s)
- A Sullivan
- Homerton University Hospital, London, UK
| | | | - A Ruban
- Department of Surgery and Cancer, Imperial College, London, UK.
| |
Collapse
|
36
|
Pfeiffer RF, Isaacson SH, Pahwa R. Clinical implications of gastric complications on levodopa treatment in Parkinson's disease. Parkinsonism Relat Disord 2020; 76:63-71. [PMID: 32461054 DOI: 10.1016/j.parkreldis.2020.05.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 12/16/2022]
Abstract
Disorders of the gastrointestinal (GI) tract are common and distressing nonmotor symptoms of Parkinson's disease (PD) that can adversely affect levodopa absorption and lead to OFF periods, also known as motor fluctuations. Gastroparesis, which is primarily defined as delayed gastric emptying (DGE), and Helicobacter pylori infection, which is present with increased frequency in PD, are among the most common and important GI disorders reported in PD that may impair oral levodopa absorption and increase OFF time. Symptoms of gastroparesis include nausea, vomiting, postprandial bloating, fullness, early satiety, abdominal pain, and weight loss. DGE has been reported in a substantial fraction of individuals with PD. Symptoms of H. pylori infection include gastritis and peptic ulcers. Studies have found that DGE and H. pylori infection are correlated with delayed peak levodopa plasma levels and increased incidence of motor fluctuations. Therapeutic strategies devised to minimize the potential that gastric complications will impair oral levodopa absorption and efficacy in PD patients include treatments that circumvent the GI tract, such as apomorphine injection, levodopa intestinal gel delivery, levodopa inhalation powder, and deep brain stimulation. Other strategies aim at improving gastric emptying in PD patients, primarily including prokinetic agents.
Collapse
Affiliation(s)
- Ronald F Pfeiffer
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA.
| | - Stuart H Isaacson
- Parkinson's Disease and Movement Disorders Center of Boca Raton, Boca Raton, FL, USA
| | - Rajesh Pahwa
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
37
|
Sallam HS, Urvil P, Savidge TC, Chen JDZ. Ghrelin abates bacterial translocation following burn injury by improving gastric emptying. Neurogastroenterol Motil 2020; 32:e13742. [PMID: 31603615 DOI: 10.1111/nmo.13742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/14/2019] [Accepted: 09/18/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND In severe burns, increased intestinal permeability facilitates bacterial translocation, resulting in systemic endotoxemia and multi- organ failure. We investigated the role of burn-induced gastrointestinal dysmotility (BIGD) in promoting bacterial translocation following burn injury, and the protective effect of ghrelin in this process. METHODS We assessed gastric emptying (GE%) and intestinal transit (IT by geometric center "GC") in a 60% total body surface area scald burn rat model and measured bacterial counts in mesenteric lymph nodes (MLN) and distal small intestine by colony-forming unit per gram of tissue (CFU/g). A group of animals was treated with ghrelin or saline after burn. KEY RESULTS Scald burn was associated with a significant delay in GE (62% ± 4% vs 74% ± 4%; P = .02) and a trend of delay in intestinal transit (GC: 5.5 ± 0.1 vs 5.8 ± 0.2; P = .09). Concurrently, there was a marginal increase in small intestinal bacterial overgrowth (6 × 105 vs 2 × 105 CFU/g; P = .05) and significant translocation to MLN (2 × 102 vs 4 × 101 ; P = .03). We observed a negative correlation between GE and intestinal bacterial overgrowth (rs = -0.61; P = .002) and between IT and translocation (rs = -0.63; P = .004). Ghrelin administration significantly accelerated GE following burn injury (91% ± 3% vs 62% ± 4; P = .03), reduced small intestinal bacterial overgrowth, and completely inhibited translocation to MLN (0.0 vs 5 × 102 ; P = .01). CONCLUSIONS & INFERENCES Burn-induced gastrointestinal dysmotility is correlated with the systemic translocation of gram-negative gut bacteria that are implicated in multiple organ failure in burn patients. Therapeutic interventions to restore BIGD are warranted (Neurogastroenterol Motil, 2012, 24, 78).
Collapse
Affiliation(s)
- Hanaa S Sallam
- Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA.,Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Petri Urvil
- Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Tor C Savidge
- Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Jiande D Z Chen
- Division of Gastroenterology, Department of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
38
|
Akalu Y, Molla MD, Dessie G, Ayelign B. Physiological Effect of Ghrelin on Body Systems. Int J Endocrinol 2020; 2020:1385138. [PMID: 32565790 PMCID: PMC7267865 DOI: 10.1155/2020/1385138] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/08/2020] [Accepted: 05/13/2020] [Indexed: 02/06/2023] Open
Abstract
Ghrelin is a relatively novel multifaceted hormone that has been found to exert a plethora of physiological effects. In this review, we found/confirmed that ghrelin has effect on all body systems. It induces appetite; promotes the use of carbohydrates as a source of fuel while sparing fat; inhibits lipid oxidation and promotes lipogenesis; stimulates the gastric acid secretion and motility; improves cardiac performance; decreases blood pressure; and protects the kidneys, heart, and brain. Ghrelin is important for learning, memory, cognition, reward, sleep, taste sensation, olfaction, and sniffing. It has sympatholytic, analgesic, antimicrobial, antifibrotic, and osteogenic effects. Moreover, ghrelin makes the skeletal muscle more excitable and stimulates its regeneration following injury; delays puberty; promotes fetal lung development; decreases thyroid hormone and testosterone; stimulates release of growth hormone, prolactin, glucagon, adrenocorticotropic hormone, cortisol, vasopressin, and oxytocin; inhibits insulin release; and promotes wound healing. Ghrelin protects the body by different mechanisms including inhibition of unwanted inflammation and induction of autophagy. Having a clear understanding of the ghrelin effect in each system has therapeutic implications. Future studies are necessary to elucidate the molecular mechanisms of ghrelin actions as well as its application as a GHSR agonist to treat most common diseases in each system without any paradoxical outcomes on the other systems.
Collapse
Affiliation(s)
- Yonas Akalu
- Department of Physiology, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Meseret Derbew Molla
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Gashaw Dessie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| |
Collapse
|
39
|
Jepsen SL, Vestergaard ET, Larraufie P, Gribble FM, Reimann F, Jørgensen JOL, Holst JJ, Kuhre RE. Ghrelin Does Not Directly Stimulate Secretion of Glucagon-like Peptide-1. J Clin Endocrinol Metab 2020; 105:dgz046. [PMID: 31608930 PMCID: PMC6941855 DOI: 10.1210/clinem/dgz046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/20/2019] [Indexed: 12/25/2022]
Abstract
CONTEXT The gastrointestinal hormone ghrelin stimulates growth hormone secretion and appetite, but recent studies indicate that ghrelin also stimulates the secretion of the appetite-inhibiting and insulinotropic hormone glucagon-like peptide-1 (GLP-1). OBJECTIVE To investigate the putative effect of ghrelin on GLP-1 secretion in vivo and in vitro. SUBJECTS AND METHODS A randomized placebo-controlled crossover study was performed in eight hypopituitary subjects. Ghrelin or saline was infused intravenously (1 pmol/min × kg) after collection of baseline sample (0 min), and blood was subsequently collected at time 30, 60, 90, and 120 minutes. Mouse small intestine was perfused (n = 6) and GLP-1 output from perfused mouse small intestine was investigated in response to vascular ghrelin administration in the presence and absence of a simultaneous luminal glucose stimulus. Ghrelin receptor expression was quantified in human (n = 11) and mouse L-cells (n = 3) by RNA sequencing and RT-qPCR, respectively. RESULTS Ghrelin did not affect GLP-1 secretion in humans (area under the curve [AUC; 0-120 min]: ghrelin infusion = 1.37 ± 0.05 min × nmol vs. saline infusion = 1.40 ± 0.06 min × nmol [P = 0.63]), but induced peripheral insulin resistance. Likewise, ghrelin did not stimulate GLP-1 secretion from the perfused mouse small intestine model (mean outputs during baseline/ghrelin infusion = 19.3 ± 1.6/25.5 ± 2.0 fmol/min, n = 6, P = 0.16), whereas glucose-dependent insulinotropic polypeptide administration, used as a positive control, doubled GLP-1 secretion (P < 0.001). Intraluminal glucose increased GLP-1 secretion by 4-fold (P < 0.001), which was not potentiated by ghrelin. Finally, gene expression of the ghrelin receptor was undetectable in mouse L-cells and marginal in human L-cells. CONCLUSIONS Ghrelin does not interact directly with the L-cell and does not directly affect GLP-1 secretion.
Collapse
Affiliation(s)
- Sara Lind Jepsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Esben Thyssen Vestergaard
- Medical Research Laboratories Aarhus University, Aarhus N, Denmark
- Department of Pediatrics, Randers Regional Hospital, Randers, Denmark
| | - Pierre Larraufie
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, UK
| | - Fiona Mary Gribble
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, UK
| | - Frank Reimann
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, UK
| | | | - Jens Juul Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rune Ehrenreich Kuhre
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Zhang S, Okuhara Y, Iijima M, Takemi S, Sakata I, Kaiya H, Teraoka H, Kitazawa T. Identification of pheasant ghrelin and motilin and their actions on contractility of the isolated gastrointestinal tract. Gen Comp Endocrinol 2020; 285:113294. [PMID: 31585115 DOI: 10.1016/j.ygcen.2019.113294] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/02/2019] [Accepted: 09/29/2019] [Indexed: 12/11/2022]
Abstract
Motilin and ghrelin were identified in the pheasant by molecular cloning, and the actions of both peptides on the contractility of gastrointestinal (GI) strips were examined in vitro. Molecular cloning indicated that the deduced amino acid sequences of the pheasant motilin and ghrelin were a 22-amino acid peptide, FVPFFTQSDIQKMQEKERIKGQ, and a 26-amino acid peptide, GSSFLSPAYKNIQQQKDTRKPTGRLH, respectively. In in vitro studies using pheasant GI strips, chicken motilin caused contraction of the proventriculus and small intestine, whereas the crop and colon were insensitive. Human motilin, but not erythromycin, caused contraction of small intestine. Chicken motilin-induced contractions in the proventriculus and ileum were not inhibited by a mammalian motilin receptor antagonist, GM109. Neither atropine (a cholinergic receptor antagonist) nor tetrodotoxin (a neuron blocker) inhibited the responses of chicken motilin in the ileum but both drugs decreased the responses to motilin in the proventriculus, suggesting that the contractile mechanisms of motilin in the proventriculus was neurogenic, different from that of the small intestine (myogenic). On the other hand, chicken and quail ghrelin did not cause contraction in any regions of pheasant GI tract. Since interaction of ghrelin and motilin has been reported in the house musk shrew, interaction of two peptides was examined. The chicken motilin-induced contractions were not modified by ghrelin, and ghrelin also did not cause any contraction under the presence of motilin, suggesting the absence of interaction in both peptides. In conclusion, both the motilin system and ghrelin system are present in the pheasant. Regulation of GI motility by motilin might be common in avian species. However, absence of ghrelin actions in any GI regions suggests the avian species-related difference in regulation of GI contractility by ghrelin.
Collapse
Affiliation(s)
- Shuangyi Zhang
- Department of Veterinary Science, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan; School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Yuji Okuhara
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Mio Iijima
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-okubo, Sakura-ku, Saitama 338-8570, Japan
| | - Shota Takemi
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-okubo, Sakura-ku, Saitama 338-8570, Japan
| | - Ichiro Sakata
- Area of Regulatory Biology, Division of Life Science, Graduate School of Science and Engineering, Saitama University, 255 Shimo-okubo, Sakura-ku, Saitama 338-8570, Japan
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka 565-8565, Japan
| | - Hiroki Teraoka
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan
| | - Takio Kitazawa
- Department of Veterinary Science, Rakuno Gakuen University, Ebetsu, Hokkaido 069-8501, Japan.
| |
Collapse
|
41
|
Abstract
This review covers the epidemiology, pathophysiology, clinical features, diagnosis, and management of diabetic gastroparesis, and more broadly diabetic gastroenteropathy, which encompasses all the gastrointestinal manifestations of diabetes mellitus. Up to 50% of patients with type 1 and type 2 DM and suboptimal glycemic control have delayed gastric emptying (GE), which can be documented with scintigraphy, 13C breath tests, or a wireless motility capsule; the remainder have normal or rapid GE. Many patients with delayed GE are asymptomatic; others have dyspepsia (i.e., mild to moderate indigestion, with or without a mild delay in GE) or gastroparesis, which is a syndrome characterized by moderate to severe upper gastrointestinal symptoms and delayed GE that suggest, but are not accompanied by, gastric outlet obstruction. Gastroparesis can markedly impair quality of life, and up to 50% of patients have significant anxiety and/or depression. Often the distinction between dyspepsia and gastroparesis is based on clinical judgement rather than established criteria. Hyperglycemia, autonomic neuropathy, and enteric neuromuscular inflammation and injury are implicated in the pathogenesis of delayed GE. Alternatively, there are limited data to suggest that delayed GE may affect glycemic control. The management of diabetic gastroparesis is guided by the severity of symptoms, the magnitude of delayed GE, and the nutritional status. Initial options include dietary modifications, supplemental oral nutrition, and antiemetic and prokinetic medications. Patients with more severe symptoms may require a venting gastrostomy or jejunostomy and/or gastric electrical stimulation. Promising newer therapeutic approaches include ghrelin receptor agonists and selective 5-hydroxytryptamine receptor agonists.
Collapse
Affiliation(s)
- Adil E Bharucha
- Clinical Enteric Neuroscience Translational and Epidemiological Research Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Yogish C Kudva
- Division of Endocrinology. Mayo Clinic, Rochester, Minnesota
| | - David O Prichard
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
42
|
Fazio Coles TE, Fothergill LJ, Hunne B, Nikfarjam M, Testro A, Callaghan B, McQuade RM, Furness JB. Quantitation and chemical coding of enteroendocrine cell populations in the human jejunum. Cell Tissue Res 2019; 379:109-120. [PMID: 31478137 DOI: 10.1007/s00441-019-03099-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
Abstract
Recent studies reveal substantial species and regional differences in enteroendocrine cell (EEC) populations, including differences in patterns of hormone coexpression, which limit extrapolation between animal models and human. In this study, jejunal samples, with no histologically identifiable pathology, from patients undergoing Whipple's procedure were investigated for the presence of gastrointestinal hormones using double- and triple-labelling immunohistochemistry and high-resolution confocal microscopy. Ten hormones (5-HT, CCK, secretin, proglucagon-derived peptides, PYY, GIP, somatostatin, neurotensin, ghrelin and motilin) were localised in EEC of the human jejunum. If only single staining is considered, the most numerous EEC were those containing 5-HT, CCK, ghrelin, GIP, motilin, secretin and proglucagon-derived peptides. All hormones had some degree of colocalisation with other hormones. This included a population of EEC in which GIP, CCK and proglucagon-derived peptides are costored, and four 5-HT cell populations, 5-HT/GIP, 5-HT/ghrelin, 5-HT/PYY, and 5-HT/secretin cell groups, and a high degree of overlap between motilin and ghrelin. The presence of 5-HT in many secretin cells is consistent across species, whereas lack of 5-HT and CCK colocalisation distinguishes human from mouse. It seems likely that the different subclasses of 5-HT cells subserve different roles. At a subcellular level, we examined the vesicular localisation of secretin and 5-HT, and found these to be separately stored. We conclude that hormone-containing cells in the human jejunum do not comply with a one-cell, one-hormone classification and that colocalisations of hormones are likely to define subtypes of EEC that have different roles.
Collapse
Affiliation(s)
- Therese E Fazio Coles
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Linda J Fothergill
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3010, Australia
| | - Billie Hunne
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mehrdad Nikfarjam
- Department of Surgery, University of Melbourne, Austin Health, Melbourne, Victoria, 3084, Australia
| | - Adam Testro
- Liver and Intestinal Transplant Unit, Austin Health, Heidelberg, Victoria, 3084, Australia
| | - Brid Callaghan
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Rachel M McQuade
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3010, Australia
| | - John B Furness
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia. .,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, 3010, Australia.
| |
Collapse
|
43
|
Sarnelli G, Annunziata G, Magno S, Oriolo C, Savastano S, Colao A. Taste and the Gastrointestinal tract: from physiology to potential therapeutic target for obesity. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2019; 9:1-9. [PMID: 31391920 DOI: 10.1038/s41367-019-0012-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Flavor is the combination of gustatory, olfactory and trigeminal sensations, representing the three main sensory pathways that allow detecting environmental chemical substances. Taste, in particular, is a complex chemosensory path that allows identification of substances present in ingested foods and beverages. In this manuscript, we propose a conceptual roadmap from aspects related to the evolution and the physiological role of taste, up to the current knowledge about its implication in the modulation of a healthy state, or obesity. More specifically, we focused on the role of stimulation of taste receptors in releasing gut hormones (also known as enterohormones), and their effects on the regulation of food intake, by inducing satiety, either by locally acting (in the gastrointestinal tract), or centrally (in the brain). Recent evidence demonstrated that some enterohormones are able to modulate gastrointestinal motility, thus affecting an orexigenic responses in the central nervous system. In keeping with this, we discuss the ability of the gustatory system to be a final checkpoint control for food intake regulation, and we speculate about taste perception manipulation in the management of obesity.
Collapse
Affiliation(s)
- Giovanni Sarnelli
- 1Department of Clinical Medicine and Surgery, Division of Gastroenterology, University of Naples Federico II, Naples, Italy
| | | | - Silvia Magno
- Obesity Center at the Endocrinology Unit, Department of Clinical and Experimental Medicine, Pisa, Italy
| | - Claudia Oriolo
- 4Endocrinology Unit, Medical Department of Care Continuity and Disability, University of Bologna, Bologna, Italy
| | - Silvia Savastano
- 5Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Napoli, Italy
| | - Annamaria Colao
- 5Dipartimento di Medicina Clinica e Chirurgia, Unit of Endocrinology, Federico II University Medical School of Naples, Napoli, Italy
| | | |
Collapse
|
44
|
Mashaqi S, Badr MS. The Impact of Obstructive Sleep Apnea and Positive Airway Pressure Therapy on Metabolic Peptides Regulating Appetite, Food Intake, Energy Homeostasis, and Systemic Inflammation: A Literature Review. J Clin Sleep Med 2019; 15:1037-1050. [PMID: 31383242 DOI: 10.5664/jcsm.7890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 04/04/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Sleep-related breathing disorders are very common and highly associated with many comorbid diseases. They have many metabolic consequences that impact appetite, energy expenditure, and systemic inflammation. These consequences are mediated through peptides (eg, ghrelin, leptin, adiponectin, resistin, apelin, obestatin, and neuropeptide Y). METHODS We searched the literature (PubMed) for sleep-disordered breathing (SDB) and metabolic peptides and included 15, 22, 14, 4 and 2 articles for ghrelin, leptin, adiponectin, resistin, and apelin respectively. RESULTS Our review of the published literature suggests that leptin levels seem to correlate with body mass index and adiposity rather than obstructive sleep apnea. Conversely, levels of adiponectin and ghrelin are influenced by obstructive sleep apnea alone. Finally, resistin and apelin seem to be not correlated with obstructive sleep apnea. Regarding positive airway pressure (PAP) impact, it seems that PAP therapy affected the levels of these peptides (mainly ghrelin). CONCLUSIONS There is significant controversy in the literature regarding the impact of SDB and PAP therapy on these metabolic peptides. This could be due to the lack of randomized clinical trials and the variability of the methodology used in these studies. Further research is needed to assess the impact of SDB and PAP therapy on the levels of these peptides and whether this impact is also related to body mass index and body fat composition.
Collapse
Affiliation(s)
- Saif Mashaqi
- Division of Sleep Medicine, University of North Dakota School of Medicine - Sanford Health, Fargo, North Dakota
| | - M Safwan Badr
- Department of Internal Medicine, Wayne State University, Detroit, Michigan
| |
Collapse
|
45
|
Kim JN, Kim BJ. The Mechanism of Action of Ghrelin and Motilin in the Pacemaker Potentials of Interstitial Cells of Cajal from the Murine Small Intestine. Mol Cells 2019; 42:470-479. [PMID: 31250620 PMCID: PMC6602145 DOI: 10.14348/molcells.2019.0028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/26/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
Interstitial cells of Cajal (ICCs) are pacemaker cells that exhibit periodic spontaneous depolarization in the gastrointestinal (GI) tract and generate pacemaker potentials. In this study, we investigated the effects of ghrelin and motilin on the pacemaker potentials of ICCs isolated from the mouse small intestine. Using the whole-cell patch-clamp configuration, we demonstrated that ghrelin depolarized pacemaker potentials of cultured ICCs in a dose-dependent manner. The ghrelin receptor antagonist [D-Lys] GHRP-6 completely inhibited this ghrelin-induced depolarization. Intracellular guanosine 5'-diphosphate-β-S and pre-treatment with Ca2+free solution or thapsigargin also blocked the ghrelin-induced depolarization. To investigate the involvement of inositol triphosphate (IP3), Rho kinase, and protein kinase C (PKC) in ghrelin-mediated pacemaker potential depolarization of ICCs, we used the IP3 receptor inhibitors 2-aminoethoxydiphenyl borate and xestospongin C, the Rho kinase inhibitor Y-27632, and the PKC inhibitors staurosporine, Go6976, and rottlerin. All inhibitors except rottlerin blocked the ghrelin-induced pacemaker potential depolarization of ICCs. In addition, motilin depolarized the pacemaker potentials of ICCs in a similar dose-dependent manner as ghrelin, and this was also completely inhibited by [D-Lys] GHRP-6. These results suggest that ghrelin induced the pacemaker potential depolarization through the ghrelin receptor in a G protein-, IP3-, Rho kinase-, and PKC-dependent manner via intracellular and extracellular Ca2+ regulation. In addition, motilin was able to depolarize the pacemaker potentials of ICCs through the ghrelin receptor. Therefore, ghrelin and its receptor may modulate GI motility by acting on ICCs in the murine small intestine.
Collapse
Affiliation(s)
- Jeong Nam Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612,
Korea
- Healthy Aging Korean Medical Research Center, Pusan National University School of Korean Medicine, Yangsan 50612,
Korea
| | - Byung Joo Kim
- Division of Longevity and Biofunctional Medicine, Pusan National University School of Korean Medicine, Yangsan 50612,
Korea
- Healthy Aging Korean Medical Research Center, Pusan National University School of Korean Medicine, Yangsan 50612,
Korea
| |
Collapse
|
46
|
Mani BK, Shankar K, Zigman JM. Ghrelin's Relationship to Blood Glucose. Endocrinology 2019; 160:1247-1261. [PMID: 30874792 PMCID: PMC6482034 DOI: 10.1210/en.2019-00074] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/09/2019] [Indexed: 12/16/2022]
Abstract
Much effort has been directed at studying the orexigenic actions of administered ghrelin and the potential effects of the endogenous ghrelin system on food intake, food reward, body weight, adiposity, and energy expenditure. Although endogenous ghrelin's actions on some of these processes remain ambiguous, its glucoregulatory actions have emerged as well-recognized features during extreme metabolic conditions. The blood glucose-raising actions of ghrelin are beneficial during starvation-like conditions, defending against life-threatening falls in blood glucose, but they are seemingly detrimental in obese states and in certain monogenic forms of diabetes, contributing to hyperglycemia. Also of interest, blood glucose negatively regulates ghrelin secretion. This article reviews the literature suggesting the existence of a blood glucose-ghrelin axis and highlights the factors that mediate the glucoregulatory actions of ghrelin, especially during metabolic extremes such as starvation and diabetes.
Collapse
Affiliation(s)
- Bharath K Mani
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Kripa Shankar
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jeffrey M Zigman
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, Texas
- Correspondence: Jeffrey M. Zigman, MD, PhD, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390. E-mail:
| |
Collapse
|
47
|
Saeidi A, Jabbour G, Ahmadian M, Abbassi-Daloii A, Malekian F, Hackney AC, Saedmocheshi S, Basati G, Ben Abderrahman A, Zouhal H. Independent and Combined Effects of Antioxidant Supplementation and Circuit Resistance Training on Selected Adipokines in Postmenopausal Women. Front Physiol 2019; 10:484. [PMID: 31105587 PMCID: PMC6499001 DOI: 10.3389/fphys.2019.00484] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 04/08/2019] [Indexed: 12/18/2022] Open
Abstract
We examined the effects of the independent and combined effects of Zataria Multiflora supplementation and circuit resistance training (CRT) on selected adipokines among postmenopausal women. Forty-eight postmenopausal women were divided into four groups: Exercise (EG, n = 12), Zataria Multiflora (ZMG, n = 12), exercise and Zataria Multiflora (ZMEG, n = 12), and control (CG, n = 12). Participants in experimental groups either performed CRT (3 sessions per week with intensity at 55% of one-repetition maximum) or supplemented with Zataria Multiflora (500 mg every day after breakfast with 100 ml of water), or their combination, for 8 weeks. Blood samples were collected at pre- and post-intervention for measuring selected adipokines, including visfatin, omentin-1, vaspin, FGF-21, adiponectin, leptin, and ghrelin. Our findings demonstrated that visfatin, vaspin, and leptin levels significantly decreased over the intervention period (all p < 0.05), with these values were lower in EG and ZMEG in comparison to CG at post-intervention (all p < 0.05). Visfatin and vaspin levels were also lower in ZMEG in comparison to EG at post-intervention (both p < 0.05). In contrast, omentin-1, ghrelin, adiponectin, and FGF21 significantly increased in EG and EMG (all p < 0.05) after CRT. These findings suggest that Zataria Multiflora supplementation by itself has little effect on measured adipokines; however, its combination with CRT produced noticeable effects on circulating levels of these adipokines, even more than CRT alone. Consequently, a combination of CRT and Zataria Multiflora supplementation may represent a potentially beneficial non-pharmacologic intervention on some selected adipokines in postmenopausal women.
Collapse
Affiliation(s)
- Ayoub Saeidi
- Exercise Biochemistry Division, Faculty of Physical Education and Sports Sciences, University of Mazandaran, Babolsar, Iran
| | - Georges Jabbour
- Sport Science Program, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Mehdi Ahmadian
- Department of Physical Education and Sports Sciences, Islamic Azad University, Aliabad-e Katul, Iran
| | | | - Fatemeh Malekian
- Southern University Agricultural Land Grant Campus, Baton Rouge, LA, United States
| | - Anthony C. Hackney
- Department of Exercise & Sports Science, The University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Saber Saedmocheshi
- Exercise Physiology Division, Faculty of Sport Science, Birjand University, Birjand, Iran
| | - Gholam Basati
- Department of Clinical Biochemistry, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Hassane Zouhal
- Movement Sports Science Laboratory, University of Rennes, Rennes, France
| |
Collapse
|
48
|
The potential probiotic Lactobacillus rhamnosus CNCM I-3690 strain protects the intestinal barrier by stimulating both mucus production and cytoprotective response. Sci Rep 2019; 9:5398. [PMID: 30931953 PMCID: PMC6443702 DOI: 10.1038/s41598-019-41738-5] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/11/2019] [Indexed: 12/14/2022] Open
Abstract
The gut barrier plays an important role in human health. When barrier function is impaired, altered permeability and barrier dysfunction can occur, leading to inflammatory bowel diseases, irritable bowel syndrome or obesity. Several bacteria, including pathogens and commensals, have been found to directly or indirectly modulate intestinal barrier function. The use of probiotic strains could be an important landmark in the management of gut dysfunction with a clear impact on the general population. Previously, we found that Lactobacillus rhamnosus CNCM I-3690 can protect intestinal barrier functions in mice inflammation model. Here, we investigated its mechanism of action. Our results show that CNCM I-3690 can (i) physically maintain modulated goblet cells and the mucus layer and (ii) counteract changes in local and systemic lymphocytes. Furthermore, mice colonic transcriptome analysis revealed that CNCM I-3690 enhances the expression of genes related to healthy gut permeability: motility and absorption, cell proliferation; and protective functions by inhibiting endogenous proteases. Finally, SpaFED pili are clearly important effectors since an L. rhamnosus ΔspaF mutant failed to provide the same benefits as the wild type strain. Taken together, our data suggest that CNCM I-3690 restores impaired intestinal barrier functions via anti-inflammatory and cytoprotective responses.
Collapse
|
49
|
Deloose E, Verbeure W, Depoortere I, Tack J. Motilin: from gastric motility stimulation to hunger signalling. Nat Rev Endocrinol 2019; 15:238-250. [PMID: 30675023 DOI: 10.1038/s41574-019-0155-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
After the discovery of motilin in 1972, motilin and the motilin receptor were studied intensely for their role in the control of gastrointestinal motility and as targets for treating hypomotility disorders. The genetic revolution - with the use of knockout models - sparked novel insights into the role of multiple peptides but contributed to a decline in interest in motilin, as this peptide and its receptor exist only as pseudogenes in rodents. The past 5 years have seen a major surge in interest in motilin, as a series of studies have shown its relevance in the control of hunger and regulation of food intake in humans in both health and disease. Luminal stimuli, such as bitter tastants, have been identified as modulators of motilin release, with effects on hunger and food intake. The current state of knowledge and potential implications for therapy are summarized in this Review.
Collapse
Affiliation(s)
- Eveline Deloose
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Wout Verbeure
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Inge Depoortere
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Jan Tack
- Translational Research in Gastrointestinal Disorders (TARGID), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium.
| |
Collapse
|
50
|
Koutouratsas T, Kalli T, Karamanolis G, Gazouli M. Contribution of ghrelin to functional gastrointestinal disorders’ pathogenesis. World J Gastroenterol 2019; 25:539-551. [PMID: 30774270 PMCID: PMC6371003 DOI: 10.3748/wjg.v25.i5.539] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 12/20/2018] [Accepted: 01/15/2019] [Indexed: 02/06/2023] Open
Abstract
Functional gastrointestinal disorders (FGID) are heterogeneous disorders with a variety of clinical manifestations, primarily defined by signs and symptoms rather than a definite underlying cause. Their pathophysiology remains obscure and, although it is expected to differ according to the specific FGID, disruptions in the brain-gut axis are now thought to be a common denominator in their pathogenesis. The hormone ghrelin is an important component of this axis, exerting a wide repertoire of physiological actions, including regulation of gastrointestinal motility and protection of mucosal tissue. Ghrelin’s gene shows genetic polymorphism, while its protein product undergoes complex regulation and metabolism in the human body. Numerous studies have studied ghrelin’s relation to the emergence of FGIDs, its potential value as an index of disease severity and as a predictive marker for symptom relief during attempted treatment. Despite the mixed results currently available in scientific literature, the plethora of statistically significant findings shows that disruptions in ghrelin genetics and expression are plausibly related to FGID pathogenesis. The aim of this paper is to review current literature studying these associations, in an effort to uncover certain patterns of alterations in both genetics and expression, which could delineate its true contribution to FGID emergence, either as a causative agent or as a pathogenetic intermediate.
Collapse
Affiliation(s)
- Tilemachos Koutouratsas
- Department of Basic Medical Science, Laboratory of Biology, School of Medicine, University of Athens, Athens 11527, Greece
| | - Theodora Kalli
- Gastroenterology Department, Larnaca General Hospital, Larnaca 6301, Cyprus
| | - Georgios Karamanolis
- Gastroenterology Unit, 2nd Department of Surgery, “Aretaieio” University Hospital, School of Medicine, University of Athens, Athens 11527, Greece
| | - Maria Gazouli
- Department of Basic Medical Science, Laboratory of Biology, School of Medicine, University of Athens, Athens 11527, Greece
| |
Collapse
|