1
|
KAJI N, IWAOKA K, NAKAMURA S, TSUKAMOTO A. Fuzapladib reduces postsurgical inflammation in the intestinal muscularis externa. J Vet Med Sci 2023; 85:1151-1156. [PMID: 37730381 PMCID: PMC10686772 DOI: 10.1292/jvms.23-0257] [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: 06/16/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023] Open
Abstract
Postoperative ileus (POI) is a surgical complication that induces emesis and anorexia. Fuzapladib (FUZ), an inhibitor of leukocyte-function-associated antigen type 1 (LFA-1) activation, a leukocyte adhesion molecule, exerts anti-inflammatory effects by inhibiting leukocyte migration into the inflammatory site. In this study, we examined the prophylactic impact of FUZ on POI in a mouse model. POI model mice were generated by intestinal manipulation, and the effect of FUZ on intestinal transit and the infiltration of inflammatory cells into the ileal muscularis externa was assessed. The increased number of macrophages was significantly suppressed by FUZ, whereas the infiltration of neutrophils into the ileal muscularis externa was not sufficiently inhibited in the POI model mice. Additionally, FUZ did not ameliorate delayed gastrointestinal transit in POI model mice. In conclusion, our results suggest that FUZ does not improve delayed gastrointestinal transit but partially inhibits inflammation in the ileal muscularis externa in POI model mice. FUZ may be a potential anti-inflammatory agent for the management of post-surgical inflammation.
Collapse
Affiliation(s)
- Noriyuki KAJI
- Laboratory of Pharmacology, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Kosuzu IWAOKA
- Laboratory of Laboratory Animal Science, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Shinichiro NAKAMURA
- Laboratory of Laboratory Animal Science, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| | - Atsushi TSUKAMOTO
- Laboratory of Laboratory Animal Science, School of Veterinary Medicine, Azabu University, Kanagawa, Japan
| |
Collapse
|
2
|
Holzer P, Holzer-Petsche U. Constipation Caused by Anti-calcitonin Gene-Related Peptide Migraine Therapeutics Explained by Antagonism of Calcitonin Gene-Related Peptide's Motor-Stimulating and Prosecretory Function in the Intestine. Front Physiol 2022; 12:820006. [PMID: 35087426 PMCID: PMC8787053 DOI: 10.3389/fphys.2021.820006] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
The development of small-molecule calcitonin gene-related peptide (CGRP) receptor antagonists (gepants) and of monoclonal antibodies targeting the CGRP system has been a major advance in the management of migraine. In the randomized controlled trials before regulatory approval, the safety of these anti-CGRP migraine therapeutics was considered favorable and to stay within the expected profile. Post-approval real-world surveys reveal, however, constipation to be a major adverse event which may affect more than 50% of patients treated with erenumab (an antibody targeting the CGRP receptor), fremanezumab or galcanezumab (antibodies targeting CGRP). In this review article we address the question whether constipation caused by inhibition of CGRP signaling can be mechanistically deduced from the known pharmacological actions and pathophysiological implications of CGRP in the digestive tract. CGRP in the gut is expressed by two distinct neuronal populations: extrinsic primary afferent nerve fibers and distinct neurons of the intrinsic enteric nervous system. In particular, CGRP is a major messenger of enteric sensory neurons which in response to mucosal stimulation activate both ascending excitatory and descending inhibitory neuronal pathways that enable propulsive (peristaltic) motor activity to take place. In addition, CGRP is able to stimulate ion and water secretion into the intestinal lumen. The motor-stimulating and prosecretory actions of CGRP combine in accelerating intestinal transit, an activity profile that has been confirmed by the ability of CGRP to induce diarrhea in mice, dogs and humans. We therefore conclude that the constipation elicited by antibodies targeting CGRP or its receptor results from interference with the physiological function of CGRP in the small and large intestine in which it contributes to the maintenance of peristaltic motor activity, ion and water secretion and intestinal transit.
Collapse
Affiliation(s)
- Peter Holzer
- Division of Pharmacology, Otto Loewi Research Centre, Medical University of Graz, Graz, Austria
| | - Ulrike Holzer-Petsche
- Division of Pharmacology, Otto Loewi Research Centre, Medical University of Graz, Graz, Austria
| |
Collapse
|
3
|
Buscail E, Deraison C. Postoperative Ileus: a Pharmacological Perspective. Br J Pharmacol 2022; 179:3283-3305. [PMID: 35048360 DOI: 10.1111/bph.15800] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/31/2021] [Accepted: 01/05/2022] [Indexed: 11/29/2022] Open
Abstract
Post-operative ileus (POI) is a frequent complication after abdominal surgery. The consequences of POI can be potentially serious such as bronchial inhalation or acute functional renal failure. Numerous advances in peri-operative management, particularly early rehabilitation, have made it possible to decrease POI. Despite this, the rate of prolonged POI ileus remains high and can be as high as 25% of patients in colorectal surgery. From a pathophysiological point of view, POI has two phases, an early neurological phase and a later inflammatory phase, to which we could add a "pharmacological" phase during which analgesic drugs, particularly opiates, play a central role. The aim of this review article is to describe the phases of the pathophysiology of POI, to analyse the pharmacological treatments currently available through published clinical trials and finally to discuss the different research areas for potential pharmacological targets.
Collapse
Affiliation(s)
- Etienne Buscail
- IRSD, INSERM, INRAE, ENVT, University of Toulouse, CHU Purpan (University Hospital Centre), Toulouse, France.,Department of digestive surgery, colorectal surgery unit, Toulouse University Hospital, Toulouse, France
| | - Céline Deraison
- IRSD, INSERM, INRAE, ENVT, University of Toulouse, CHU Purpan (University Hospital Centre), Toulouse, France
| |
Collapse
|
4
|
Sympathetic Denervation Alters the Inflammatory Response of Resident Muscularis Macrophages upon Surgical Trauma and Ameliorates Postoperative Ileus in Mice. Int J Mol Sci 2021; 22:ijms22136872. [PMID: 34206766 PMCID: PMC8268963 DOI: 10.3390/ijms22136872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 01/22/2023] Open
Abstract
Interactions between the peripheral nervous system and resident macrophages (MMs) modulate intestinal homeostatic functions. Activation of β2-adrenergic receptors on MMs has been shown to reduce bacterial challenges. These MMs are also crucial for the development of bowel inflammation in postoperative ileus (POI), an iatrogenic, noninfectious inflammation-based motility disorder. However, the role of the sympathetic nervous system (SNS) in the immune modulation of these MMs during POI or other noninfectious diseases is largely unknown. By employing 6-OHDA-induced denervation, we investigated the changes in the muscularis externa by RNA-seq, quantitative PCR, and flow cytometry. Further, we performed transcriptional phenotyping of sorted CX3CR1+ MMs and ex vivo LPS/M-CSF stimulation on these MMs. By combining denervation with a mouse POI model, we explored distinct changes on CX3CR1+ MMs as well as in the muscularis externa and their functional outcome during POI. Our results identify SNS as an important mediator in noninfectious postoperative inflammation. Upon denervation, MMs anti-inflammatory genes were reduced, and the muscularis externa profile is shaped toward a proinflammatory status. Further, denervation reduced MMs anti-inflammatory genes also in the early phase of POI. Finally, reduced leukocyte infiltration into the muscularis led to a quicker recovery of bowel motility in the late phase of POI.
Collapse
|
5
|
Lee SE, Kim DH, Son SM, Choi SY, You RY, Kim CH, Choi W, Kim HS, Lim YJ, Han JY, Kim HW, Yang IJ, Xu WX, Lee SJ, Kim YC, Yun HY. Physiological function and molecular composition of ATP-sensitive K + channels in human gastric smooth muscle. J Smooth Muscle Res 2020; 56:29-45. [PMID: 32581184 PMCID: PMC7324727 DOI: 10.1540/jsmr.56.29] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Gastric motility is controlled by slow waves. In general, the activation of the
ATP-sensitive K+ (KATP) channels in the smooth muscle
opposes the membrane excitability and produces relaxation. Since metabolic
inhibition and/or diabetes mellitus are accompanied by dysfunctions of gastric
smooth muscle, we examined the possible roles of KATP channels in
human gastric motility. We used human gastric corpus and antrum smooth muscle
preparations and recorded the mechanical activities with a conventional
contractile measuring system. We also identified the subunits of the
KATP channels using Western blot. Pinacidil (10 μM), a
KATP channel opener, suppressed contractions to 30% (basal tone
to −0.2 g) of the control. The inhibitory effect of pinacidil on contraction was
reversed to 59% of the control by glibenclamide (20 μM), a KATP
channel blocker. The relaxation by pinacidil was not affected by a pretreatment
with L-arginine methyl ester, tetraethylammonium, or 4-aminopyridine. Pinacidil
also inhibited the acetylcholine (ACh)-induced tonic and phasic contractions in
a glibenclamide-sensitive manner (42% and 6% of the control, respectively).
Other KATP channel openers such as diazoxide, cromakalim and
nicorandil also inhibited the spontaneous and ACh-induced contractions.
Calcitonin gene-related peptide (CGRP), a gastric neuropeptide, induced muscle
relaxation by the activation of KATP channels in human gastric smooth
muscle. Finally, we have found with Western blot studies, that human gastric
smooth muscle expressed KATP channels which were composed of Kir 6.2
and SUR2B subunits.
Collapse
Affiliation(s)
- Sang Eok Lee
- Department of Surgery, College of Medicine, Konyang University, 158 Gwanjeodong-ro, Seo-gu, Daejeon 35365, Korea
| | - Dae Hoon Kim
- Department of Surgery, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Seung Myeung Son
- Department of Pathology, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Song-Yi Choi
- Department of Pathology, School of Medicine, Chungnam National University, Daejeon, Chungnam 35015, Korea
| | - Ra Young You
- Department of Physiology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Chan Hyung Kim
- Department of Pharmacology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Woong Choi
- Department of Pharmacology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Hun Sik Kim
- Department of Pharmacology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Yung Ji Lim
- College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Ji Young Han
- CheongDam I Plastic surgery, 21, Sinheung-ro 240, Uijeonbu-si, Gyronggido 11651, Korea
| | - Hyun Woo Kim
- College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - In Jun Yang
- Department of Surgery, Seoul Nantional University Bundang Hospital, 166 Gumi-ro, Bundang-gu 13620, Seongnam, Korea
| | - Wen-Xie Xu
- Department of Physiology, College of Medcine, Shanghai Jiaotong University, 800 Dongchun Rd. Shanghai, 200240, P.R. China
| | - Sang Jin Lee
- Department of Physiology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Young Chul Kim
- Department of Physiology, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| | - Hyo-Yung Yun
- Department of Surgery, College of Medicine, Chungbuk National University, Chungdae-ro 1, Seowon-gu, Cheongju, Chungbuk 28644, Korea
| |
Collapse
|
6
|
Kaji N, Nakayama S, Horiguchi K, Iino S, Ozaki H, Hori M. Disruption of the pacemaker activity of interstitial cells of Cajal via nitric oxide contributes to postoperative ileus. Neurogastroenterol Motil 2018; 30. [PMID: 29542843 DOI: 10.1111/nmo.13334] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 02/11/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Interstitial cells of Cajal (ICC) serve as intestinal pacemakers. Postoperative ileus (POI) is a gastrointestinal motility disorder that occurs following abdominal surgery, which is caused by inflammation-induced dysfunction of smooth muscles and enteric neurons. However, the participation of ICC in POI is not well understood. In this study, we investigated the functional changes of ICC in a mouse model of POI. METHODS Intestinal manipulation (IM) was performed to induce POI. At 24 h or 48 h after IM, the field potential of the intestinal tunica muscularis was investigated. Tissues were also examined by immunohistochemistry and electron microscopic analysis. KEY RESULTS Gastrointestinal transit was significantly decreased with intestinal tunica muscularis inflammation at 24 h after IM, which was ameliorated at 48 h after IM. The generation and propagation of pacemaker potentials were disrupted at 24 h after IM and recovered to the control level at 48 h after IM. ICC networks, detected by c-Kit immunoreactivity, were remarkably disrupted at 24 h after IM. Electron microscopic analysis revealed abnormal vacuoles in the ICC cytoplasm. Interestingly, the ICC networks recovered at 48 h after IM. Administration of aminoguanidine, an inducible nitric oxide synthase inhibitor, suppressed the disruption of ICC networks. Ileal smooth muscle tissue cultured in the presence of nitric oxide donor, showed disrupted ICC networks. CONCLUSIONS AND INFERENCES The generation and propagation of pacemaker potentials by ICC are disrupted via nitric oxide after IM, and this disruption may contribute to POI. When inflammation is ameliorated, ICC can recover their pacemaker function.
Collapse
Affiliation(s)
- N Kaji
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - S Nakayama
- Department of Cell Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - K Horiguchi
- Division of Anatomy and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - S Iino
- Division of Anatomy and Neuroscience, Department of Morphological and Physiological Sciences, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - H Ozaki
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - M Hori
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Lisowski ZM, Pirie RS, Blikslager AT, Lefebvre D, Hume DA, Hudson NPH. An update on equine post-operative ileus: Definitions, pathophysiology and management. Equine Vet J 2018; 50:292-303. [DOI: 10.1111/evj.12801] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 11/24/2017] [Indexed: 12/18/2022]
Affiliation(s)
- Z. M. Lisowski
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| | - R. S. Pirie
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| | - A. T. Blikslager
- Department of Clinical Sciences; College of Veterinary Medicine; North Carolina State University; Raleigh North Carolina USA
| | - D. Lefebvre
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| | - D. A. Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
- Mater Research; The University of Queensland; Woolloongabba Queensland Australia
| | - N. P. H. Hudson
- The Roslin Institute and Royal (Dick) School of Veterinary Studies; University of Edinburgh, Easter Bush; Midlothian UK
| |
Collapse
|
8
|
Stakenborg N, Gomez-Pinilla PJ, Boeckxstaens GE. Postoperative Ileus: Pathophysiology, Current Therapeutic Approaches. Handb Exp Pharmacol 2017; 239:39-57. [PMID: 27999957 DOI: 10.1007/164_2016_108] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Postoperative ileus, which develops after each abdominal surgical procedure, is an iatrogenic disorder characterized by a transient inhibition of gastrointestinal motility. Its pathophysiology is complex involving pharmacological (opioids, anesthetics), neural, and immune-mediated mechanisms. The early neural phase, triggered by activation of afferent nerves during the surgical procedure, is short lasting compared to the later inflammatory phase. The latter starts after 3-6 h and lasts several days, making it a more interesting target for treatment. Insight into the triggers and immune cells involved is of great importance for the development of new therapeutic strategies. In this chapter, the pathogenesis and the current therapeutic approaches to treat postoperative ileus are discussed.
Collapse
Affiliation(s)
- N Stakenborg
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - P J Gomez-Pinilla
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium
| | - G E Boeckxstaens
- Division of Gastroenterology and Hepatology, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Herestraat 49, Leuven, 3000, Belgium. .,Division of Gastroenterology and Hepatology, University Hospital Leuven, Herestraat 49, Leuven, 3000, Belgium.
| |
Collapse
|
9
|
Fang JF, Fang JQ, Shao XM, Du JY, Liang Y, Wang W, Liu Z. Electroacupuncture treatment partly promotes the recovery time of postoperative ileus by activating the vagus nerve but not regulating local inflammation. Sci Rep 2017; 7:39801. [PMID: 28051128 PMCID: PMC5209726 DOI: 10.1038/srep39801] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 11/29/2016] [Indexed: 12/11/2022] Open
Abstract
Postoperative ileus (POI) after abdominal surgery significantly lowers the life quality of patients and increase hospital costs. However, few treatment strategies have successfully shortened the duration of POI. Electroacupuncture (EA) is a modern way of administering acupuncture and widely used in various gastrointestinal (GI) diseases in the world. Here, we studied the effect of EA on POI and its underlying mechanisms. Intestinal manipulation resulted in significant delays of GI transit, colonic transit and gastric emptying. Surgery also up-regulated c-fos in nucleus of the solitary tract (NTS) and induced inflammation response in the small intestine. Further, operation and inhale anesthesia inhibited NTS neuron excitation duration for the whole observation time. EA administered at ST36 indeed shortened the recovery time of GI and colonic transit, and significantly increased the gastric emptying. EA also significantly activated the NTS neurons after operation. However, there was no anti-inflammation effect of EA during the whole experiment. Finally, atropine blocked the regulatory effect of EA on GI function, when it was injected after surgery, but not before surgery. Thus, the regulatory effect of EA on POI was mainly mediated by exciting NTS neurons to improve the GI tract transit function but not by activating cholinergic anti-inflammatory pathway.
Collapse
Affiliation(s)
- Jun-Fan Fang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian-Qiao Fang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiao-Mei Shao
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun-Ying Du
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Liang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wen Wang
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhe Liu
- Department of Neurobiology &Acupuncture Research, the Third Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
10
|
Berdún S, Rychter J, Vergara P. Surgical intestinal manipulation increases gene expression of TrkA, CGRP, and PAR-2 IN dorsal root ganglia in the rat. Neurogastroenterol Motil 2016; 28:816-26. [PMID: 26909771 DOI: 10.1111/nmo.12777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 12/23/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Surgical handling of the bowel evokes degranulation of peritoneal mast cells (PMC). Nonetheless, role of PMCs in postoperative ileus (POI) is somewhat controversial. We aimed to investigate if intestinal manipulation elicits changes in afferent mediators related to MC activation and alteration of gastrointestinal (GI) motility. METHODS Postoperative ileus was induced by intestinal manipulation in Sprague-Dawley rats. Additionally, compound 48/80 (C48/80) and ketotifen were used to modulate MC activity. Rat mast cell protease 6 (RMCP-6, ELISA) release was determined in peritoneal lavage 20 min after intestinal manipulation. At 24 h, GI transit was determined. Gene expression of calcitonin gene-related peptide (CGRP), protease-activated receptor-2 (PAR-2), nerve growth factor (NGF), and TrkA receptor was determined (PCR) in dorsal root ganglia (DRG). Ileal wall inflammation was assessed by myeloperoxidase (MPO) activity, interleukin-6 expression (IL-6). KEY RESULTS Intestinal manipulation and exposure to C48/80-induced degranulation of PMCs delayed GI transit and up-regulated IL-6 and MPO activity. Intestinal manipulation, but not C48/80, up-regulated CGRP, PAR-2, and NGF/TrkA in DRGs. Ketotifen only improved gastric emptying and fecal output. Up-regulation of CGRP and TrkA expression in DRG was not prevented by ketotifen. CONCLUSIONS & INFERENCES Postoperative ileus is accompanied by activation of CGRP, NGF-TrkA, and PAR-2 in DRGs. Our results suggest that these mediators could be a target in further POI studies in order to find new therapeutic targets for this medical condition.
Collapse
Affiliation(s)
- S Berdún
- Department of Cell Biology, Physiology and Immunology, Veterinary School, Universitat Autònoma de Barcelona, Barcelona, Spain
- Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - J Rychter
- Department of Cell Biology, Physiology and Immunology, Veterinary School, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain
| | - P Vergara
- Department of Cell Biology, Physiology and Immunology, Veterinary School, Universitat Autònoma de Barcelona, Barcelona, Spain
- Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain
| |
Collapse
|
11
|
Ge W, Chen G, Ding YT. Effect of chewing gum on the postoperative recovery of gastrointestinal function. Int J Clin Exp Med 2015; 8:11936-11942. [PMID: 26550107 PMCID: PMC4612792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/06/2015] [Indexed: 06/05/2023]
Abstract
Postoperative gastrointestinal dysfunction remains a source of morbidity and the major determinant of length of stay after abdominal operation. There are many different reasons for postoperative gastrointestinal dysfunction such as stress response, perioperative interventions, bowel manipulation and so on. The mechanism of enhanced recovery from postoperative gastrointestinal dysfunction with the help of chewing gum is believed to be the cephalic-vagal stimulation of digestion which increases the promotability of neural and humoral factors that act on different parts of the gastrointestinal tract. Recently, there were a series of randomized controlled trials to confirm the role of chewing gum in the recovery of gastrointestinal function. The results suggested that chewing gum enhanced early recovery of bowel function following abdominal surgery expect the gastrointestinal surgery. However, the effect of chewing gum in gastrointestinal surgery was controversial.
Collapse
Affiliation(s)
- Wei Ge
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008, Jiangsu Province, P. R. China
| | - Gang Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008, Jiangsu Province, P. R. China
| | - Yi-Tao Ding
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008, Jiangsu Province, P. R. China
| |
Collapse
|
12
|
Rychter J, Ortega O, Berdun S, Arenas C, Lopez I, Espin F, Vergara P, Clavé P. Mast cell degranulation inhibits motor patterns of human ileum and sigmoid colon in vitro: relevance for postoperative ileus. Neurogastroenterol Motil 2015; 27:1098-109. [PMID: 25974622 DOI: 10.1111/nmo.12589] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 04/16/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Local release of mast cell proteases during gastrointestinal surgery is associated with the inhibition of motility and postoperative ileus (POI). We determined whether activation of intramuscular mast cell affects the motor patterns of the human ileum and colon and whether proteases are involved. METHODS Motor response of ileal and colonic circular muscle strips was measured in organ bath. Mast cell degranulation was induced by compound 48/80 (c48/80; 25-675 μg/mL). Motor response was quantified as tone, rhythmic phasic contractions (RPCs) and contractions to electric field stimulation (EFS; 40 Hz), and bethanechol-evoked contractions. Ketotifen (10(-6) mol/L) and a protease inhibitor cocktail (P8340) were used to evaluate the role of mast cell mediators. KEY RESULTS (a) c48/80 impaired the spontaneous and the electrically evoked motor response in small bowel and colonic strips (sigmoid colon EC50 : 460.0 μg/mL for RPCs and 8.9 μg/mL for electrically evoked contraction amplitudes) and bethanechol-evoked contractions. (b) Preincubation with ketotifen (10(-6) mol/L, 1 h) prevented the impairment of RPCs and EFS-evoked contractions in the sigmoid colon and ileum but not in the right colon. (c) Preincubation with P8340 also prevented the impairment of contractions in the sigmoid colon but not in the ileum or the right colon. CONCLUSIONS & INFERENCES Mast cell degranulation by c48/80 inhibits the spontaneous and the nerve-mediated motor response in the human ileum and colon. The effect is partially mediated by mast cell proteases and could be relevant in the pathophysiology of POI.
Collapse
Affiliation(s)
- J Rychter
- CIBERehd, Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Instituto de Salud Carlos III, Barcelona, Catalonia, Spain.,Department of Cell Biology, Physiology and Immunology, Veterinary School, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - O Ortega
- Department of Surgery, Hospital de Mataró, Universitat Autónoma de Barcelona, Mataró, Catalonia, Spain
| | - S Berdun
- Department of Cell Biology, Physiology and Immunology, Veterinary School, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - C Arenas
- CIBERehd, Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Instituto de Salud Carlos III, Barcelona, Catalonia, Spain
| | - I Lopez
- Department of Surgery, Hospital de Mataró, Universitat Autónoma de Barcelona, Mataró, Catalonia, Spain
| | - F Espin
- Department of Surgery, Hospital de Mataró, Universitat Autónoma de Barcelona, Mataró, Catalonia, Spain
| | - P Vergara
- CIBERehd, Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Instituto de Salud Carlos III, Barcelona, Catalonia, Spain.,Department of Cell Biology, Physiology and Immunology, Veterinary School, Universitat Autònoma de Barcelona, Barcelona, Catalonia, Spain
| | - P Clavé
- CIBERehd, Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas, Instituto de Salud Carlos III, Barcelona, Catalonia, Spain.,Department of Surgery, Hospital de Mataró, Universitat Autónoma de Barcelona, Mataró, Catalonia, Spain
| |
Collapse
|
13
|
Glowka TR, Steinebach A, Stein K, Schwandt T, Lysson M, Holzmann B, Tsujikawa K, de Jonge WJ, Kalff JC, Wehner S. The novel CGRP receptor antagonist BIBN4096BS alleviates a postoperative intestinal inflammation and prevents postoperative ileus. Neurogastroenterol Motil 2015; 27:1038-49. [PMID: 25929169 DOI: 10.1111/nmo.12584] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 04/13/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Abdominal surgery results in neuronal mediator release and subsequent acute intestinal hypomotility. This phase is followed by a longer lasting inflammatory phase resulting in postoperative ileus (POI). Calcitonin gene-related peptide (CGRP) has been shown to induce motility disturbances and in addition may be a candidate mediator to elicit neurogenic inflammation. We hypothesized that CGRP contributes to intestinal inflammation and POI. METHODS The effect of CGRP in POI was tested in mice treated with the highly specific CGRP receptor antagonist BIBN4096BS and in CGRP receptor-deficient (RAMP-1(-/-) ) mice. POI severity was analyzed by cytokine expression, muscular inflammation and gastrointestinal (GI) transit. Peritoneal and muscularis macrophages and mast cells were analyzed for CGRP receptor expression and functional response to CGRP stimulation. KEY RESULTS Intestinal manipulation (IM) resulted in CGRP release from myenteric nerves, and a concurrent increased interleukin (IL)-6 and IL-1β transcription and leukocyte infiltration in the muscularis externa and increased GI transit time. CGRP potentiates IM-induced cytokine transcription within the muscularis externa and peritoneal macrophages. BIBN4096BS reduced cytokine levels and leukocyte infiltration and normalized GI transit. RAMP1(-/-) mice showed a significantly reduced leukocyte influx. CGRP receptor was expressed in muscularis and peritoneal macrophages but not mast cells. CGRP mediated macrophage activation but failed to induce mast cell degranulation and cytokine expression. CONCLUSIONS & INFERENCES CGRP is immediately released during abdominal surgery and induces a neurogenic inflammation via activation of abdominal macrophages. BIBN4096BS prevented IM-induced inflammation and restored GI motility. These findings suggest that CGRP receptor antagonism could be instrumental in the prevention of POI.
Collapse
Affiliation(s)
- T R Glowka
- Department of Surgery, University of Bonn, Bonn, Germany
| | - A Steinebach
- Department of Surgery, University of Bonn, Bonn, Germany
| | - K Stein
- Department of Surgery, University of Bonn, Bonn, Germany
| | - T Schwandt
- Department of Surgery, University of Bonn, Bonn, Germany
| | - M Lysson
- Department of Surgery, University of Bonn, Bonn, Germany
| | - B Holzmann
- Department of Surgery, Technical University Munich, Munich, Germany
| | - K Tsujikawa
- Department of Immunology, Graduate School of Pharmaceutical Sciences, Osaka, Japan
| | - W J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| | - J C Kalff
- Department of Surgery, University of Bonn, Bonn, Germany
| | - S Wehner
- Department of Surgery, University of Bonn, Bonn, Germany.,Tytgat Institute for Liver and Intestinal Research, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
14
|
Sham feeding with chewing gum after elective colorectal resectional surgery: a randomized clinical trial. Ann Surg 2013; 257:1016-24. [PMID: 23470575 DOI: 10.1097/sla.0b013e318286504a] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To determine whether sham feeding with chewing gum improved gastrointestinal recovery after colorectal resection surgery, in the presence of routine postoperative feeding. BACKGROUND Sham feeding with chewing gum has been shown to accelerate the return of gut function after colorectal surgery. This study sought to determine whether sham feeding with gum, after colorectal resection, accelerates return of gastrointestinal function in patients on a rapid feeding enhanced recovery program. METHODS A randomized "two armed" controlled clinical trial was performed. Equal groups of open and laparoscopic colorectal resection surgical patients were recruited. Patients in the intervention arm received chewing gum 4 times a day postoperatively. All patients in the trial were placed on an established, standardized Enhanced Recovery After Surgery program. The primary outcome was time to return of gut function, assessed by time to flatus and first bowel motion. Secondary outcomes were time to tolerate diet, symptoms of ileus in the form of nausea, vomiting and distension, pain as assessed by analgesic consumption and visual analogue scales, complications, and length of hospital stay. RESULTS A total of 161 patients were recruited. Postoperative morbidity was equivalent between groups, with no complications related to gum chewing. There was no difference between groups with respect to the primary outcomes of time to flatus and bowel motion. There was less perception of pain in the intervention group on days 2 to 5, and no difference with respect to all other secondary outcomes. CONCLUSIONS Sham feeding with gum, after open and laparoscopic colorectal resectional surgery is safe, but does not hasten the return of gastrointestinal function in patients who receive accelerated postoperative feeding. (ACTRN12607000538448).
Collapse
|
15
|
Costes LMM, Boeckxstaens GE, de Jonge WJ, Cailotto C. Neural networks in intestinal immunoregulation. Organogenesis 2013; 9:216-23. [PMID: 23867810 PMCID: PMC3896593 DOI: 10.4161/org.25646] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Key physiological functions of the intestine are governed by nerves and neurotransmitters. This complex control relies on two neuronal systems: an extrinsic innervation supplied by the two branches of the autonomic nervous system and an intrinsic innervation provided by the enteric nervous system. As a result of constant exposure to commensal and pathogenic microflora, the intestine developed a tightly regulated immune system. In this review, we cover the current knowledge on the interactions between the gut innervation and the intestinal immune system. The relations between extrinsic and intrinsic neuronal inputs are highlighted with regards to the intestinal immune response. Moreover, we discuss the latest findings on mechanisms underlying inflammatory neural reflexes and examine their relevance in the context of the intestinal inflammation. Finally, we discuss some of the recent data on the identification of the gut microbiota as an emerging player influencing the brain function.
Collapse
Affiliation(s)
- Léa M M Costes
- Department of Neurogastroenterology; Tytgat Institute for Liver and Intestinal Research; Academic Medical Center (AMC); Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
16
|
Huang CC, Hsu HH, Huang JY, Lao HC, Cheng JK, Chen CC, Lin CS. Treating a patient with intractable paralytic ileus using thoracic epidural analgesia. ACTA ANAESTHESIOLOGICA TAIWANICA : OFFICIAL JOURNAL OF THE TAIWAN SOCIETY OF ANESTHESIOLOGISTS 2012; 50:78-80. [PMID: 22769863 DOI: 10.1016/j.aat.2012.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 01/04/2012] [Accepted: 01/09/2012] [Indexed: 11/25/2022]
Abstract
Postoperative ileus is considered an undesirable response to major abdominal surgery that leads to discomfort, complications, morbidity, and the prolongation of hospital stays. Although thoracic epidural analgesia has been introduced to prevent and/or reduce postoperative ileus, it is rarely used as a way to treat postoperative ileus. A 65-year-old man developed paralytic ileus after undergoing a colectomy. Despite conservative and surgical management, postoperative morbidity persisted. A continuous infusion of 0.2% levobupivacaine at a rate of 4 mL/hour was administered for 4 days via a thoracic epidural catheter that had been percutaneously tunneled into the T11-T12 epidural space. With this treatment, daily drainage from a nasogastric tube was gradually decreased and flatus was noted. A week later, the patient could start receiving a liquid diet. Therefore, thoracic epidural analgesia can be used to treat or alleviate paralytic ileus.
Collapse
Affiliation(s)
- Chien-Chung Huang
- Department of Anesthesiology, Mackay Memorial Hospital, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
17
|
Wehner S, Vilz TO, Stoffels B, Kalff JC. Immune mediators of postoperative ileus. Langenbecks Arch Surg 2012; 397:591-601. [PMID: 22382699 DOI: 10.1007/s00423-012-0915-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Accepted: 01/24/2012] [Indexed: 02/08/2023]
Abstract
UNLABELLED CLINICAL BACKGROUND: In all patients undergoing abdominal surgery, a transient phase of interruption of bowel motility, named postoperative ileus (POI) occurs. POI is often accepted as an unavoidable "physiological" response and a self-limiting complication after surgery although it has a significant impact on patient morbidity with prolonged hospitalization and increased costs. Annual economic burden has been estimated as much as US $1.47 billion in the USA (Iyer et al. in J Manag Care Pharm 15(6):485-494, 2009). PATHOPHYSIOLOGY The pathophysiology has been elucidated within the last decades, demonstrating that both, neurogenic and inflammatory mechanisms are involved in response to the surgical trauma. It is now generally accepted that POI pathogenesis processes in two phases: a first neurogenic phase is accountable for the immediate postoperative impairment of bowel motility. This is followed by a second immunological phase that can last for days and mainly affects strength and length of POI. More recent findings demonstrate a bidirectional interaction between the nervous and the immune system, and this interaction significantly contributed to our present understanding of POI pathophysiology. Although nerval mechanisms have a significant impact in the early phase of POI, the contribution of the immune system and subsequently its manipulation has risen as the most promising strategy in prevention or treatment of the clinically relevant prolonged form of POI. AIMS The present manuscript will give an update on the inflammatory responses, the involved cell types, and participating immune mediators in POI.
Collapse
Affiliation(s)
- Sven Wehner
- Department of Surgery, University of Bonn, Sigmund-Freud-Str. 25, 53105 Bonn, Germany
| | | | | | | |
Collapse
|
18
|
Stengel A, Wang L, Taché Y. Stress-related alterations of acyl and desacyl ghrelin circulating levels: mechanisms and functional implications. Peptides 2011; 32:2208-17. [PMID: 21782868 PMCID: PMC3220774 DOI: 10.1016/j.peptides.2011.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2011] [Revised: 07/04/2011] [Accepted: 07/05/2011] [Indexed: 12/11/2022]
Abstract
Ghrelin is the only known peripherally produced and centrally acting peptide hormone that stimulates food intake and digestive functions. Ghrelin circulates as acylated and desacylated forms and recently the acylating enzyme, ghrelin-O-acyltransferase (GOAT) and the de-acylating enzyme, thioesterase 1/lysophospholipase 1 have been identified adding new layers of complexity to the regulation of ghrelin. Stress is known to alter gastrointestinal motility and food intake and was recently shown to modify circulating ghrelin and GOAT levels with differential responses related to the type of stressors including a reduction induced by physical stressors (abdominal surgery and immunological/endotoxin injection, exercise) and elevation by metabolic (cold exposure, acute fasting and caloric restriction) and psychological stressors. However, the pathways underlying the alterations of ghrelin under these various stress conditions are still largely to be defined and may relate to stress-associated autonomic changes. There is evidence that alterations of circulating ghrelin may contribute to the neuroendocrine and behavioral responses along with sustaining the energetic requirement needed upon repeated exposure to stressors. A better understanding of these mechanisms will allow targeting components of ghrelin signaling that may improve food intake and gastric motility alterations induced by stress.
Collapse
Affiliation(s)
- Andreas Stengel
- CURE: Digestive Diseases Research Center, David Geffen School of Medicine, University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, United States.
| | | | | |
Collapse
|
19
|
Stengel A, Goebel-Stengel M, Wang L, Shaikh A, Lambrecht NWG, Rivier J, Taché Y. Abdominal surgery inhibits circulating acyl ghrelin and ghrelin-O-acyltransferase levels in rats: role of the somatostatin receptor subtype 2. Am J Physiol Gastrointest Liver Physiol 2011; 301:G239-48. [PMID: 21636529 PMCID: PMC3154605 DOI: 10.1152/ajpgi.00018.2011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Clinical studies are evaluating the efficacy of synthetic ghrelin agonists in postoperative ileus management. However, the control of ghrelin secretion under conditions of postoperative gastric ileus is largely unknown. Peripheral somatostatin inhibits ghrelin secretion in animals and humans. We investigated the time course of ghrelin changes postsurgery in fasted rats and whether somatostatin receptor subtype 2 (sst(2)) signaling is involved. Abdominal surgery (laparotomy and 1-min cecal palpation) induced a rapid and long-lasting decrease in plasma acyl ghrelin levels as shown by the 64, 67, and 59% reduction at 0.5, 2, and 5 h postsurgery, respectively, compared with sham (anesthesia alone for 10 min, P < 0.05). Levels were partly recovered at 7 h and fully restored at 24 h. The percentage of acyl ghrelin reduction was significantly higher than that of desacyl ghrelin at 2 h postsurgery and not at any other time point. This was associated with a 48 and 23% decrease in gastric and plasma ghrelin-O-acyltransferase protein concentrations, respectively (P < 0.001). Ghrelin-positive cells in the oxyntic mucosa expressed sst(2a) receptor and the sst(2) agonist S-346-011 inhibited fasting acyl ghrelin levels by 64 and 77% at 0.5 and 2 h, respectively. The sst(2) antagonist S-406-028 prevented the abdominal surgery-induced decreased circulating acyl ghrelin but not the delayed gastric emptying assessed 0.5 h postinjection. These data show that activation of sst(2) receptor located on gastric X/A-like cells plays a key role in the rapid inhibition of circulating acyl ghrelin induced by abdominal surgery while not being primarily involved in the early phase of postoperative gastric ileus.
Collapse
Affiliation(s)
- Andreas Stengel
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Miriam Goebel-Stengel
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Lixin Wang
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Almaas Shaikh
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| | - Nils W. G. Lambrecht
- 2Gastrointestinal Endocrinology, Veterans Affairs Long Beach Healthcare System, Long Beach; and
| | - Jean Rivier
- 3Peptide Biology Laboratories, Salk Institute, La Jolla, California
| | - Yvette Taché
- 1Department of Medicine, CURE Digestive Diseases Research Center, Center for Neurobiology of Stress, Digestive Diseases Division at University of California Los Angeles and Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles;
| |
Collapse
|
20
|
Mueller MH, Karpitschka M, Gao Z, Mittler S, Kasparek MS, Renz B, Sibaev A, Glatzle J, Li Y, Kreis ME. Vagal innervation and early postoperative ileus in mice. J Gastrointest Surg 2011; 15:891-900; discussion 900-1. [PMID: 21437764 DOI: 10.1007/s11605-011-1481-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 02/27/2011] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Postoperative ileus is characterized by infiltrates of leukocytes in the gut wall 24 h after surgery, which is subject to vagal modulation. We hypothesized that vagal modulation is irrelevant during earlier hours of postoperative ileus and aimed to determine whether afferent neuronal feedback to the central nervous system is altered by vagal innervation during this early period. METHODS C57BL6 mice were laparotomized and received standardized small bowel manipulation to induce postoperative ileus. Subgroups were vagotomized 3-4 days prior to experiments while control animals were sham-operated. Three or 9 h later a 2-cm jejunal segment was harvested for multi-unit mesenteric afferent nerve recordings in vitro. Intestinal motility was monitored continuously and intestinal muscularis was stained for myeloperoxidase to determine infiltration of leukocytes. RESULTS Peak amplitudes of intestinal motility and afferent nerve discharge at baseline were not different in all subgroups. Afferent discharge to 5-HT (500 μM) was virtually absent following vagotomy at 3 and 9 h of postoperative ileus (POI) compared to controls (p < 0.05). Maximum afferent nerve discharge to bradykinin and peak firing during maximum distension at 60 mmHg was not different in all subgroups while luminal distension from 10 to 30 mmHg was lower at 3 h of POI following vagotomy compared to controls (p < 0.05). The number of myeloperoxidase positive cells was similar at 3 h of POI in both subgroups; however, at 9 h of POI, ileus counts were increased to 713 ± 99 cells following vagotomy compared to 47 ± 6 cells per square millimeter in control animals. CONCLUSIONS Vagal afferents mediate sensitivity to low-threshold distension and 5-HT during postoperative ileus but not to high-threshold distension and bradykinin. Vagal inhibition of the intestinal immune response is present at 9 h but not detectable earlier, i.e., at 3 h of postoperative ileus when spinal reflex inhibition may prevail.
Collapse
Affiliation(s)
- Mario H Mueller
- Walter-Brendel Institute for Surgical Research, Ludwig-Maximilian's University, Munich, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Role of the vagus nerve on the development of postoperative ileus. Langenbecks Arch Surg 2010; 395:407-11. [PMID: 20333399 DOI: 10.1007/s00423-010-0594-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2009] [Accepted: 01/07/2010] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Postoperative ileus involves reflex inhibition of intestinal motility within hours after surgery and a subsequent intestinal inflammatory response that is characterized by efferent vagal modulation via acetylcholine receptors on intestinal macrophages. We aimed to characterize the role of vagal modulation of intestinal motility during the early hours after surgery. METHODS C57BL6 mice underwent laparotomy and standardized small bowel manipulation to induce postoperative ileus. Subgroups were vagotomized 3-4 days prior to experiments or received pharmacological inhibition of the acetylcholine alpha7 subunit with the inhibitor alpha-bungarotoxin, while control animals were sham operated and remained otherwise untreated. Three hours later, a 2-cm jejunal segment was harvested with the mesentery attached. Mesenteric afferent nerve recordings were established in an organ bath generating a multiunit signal with subsequent computerized analysis. Intraluminal pressure was continuously recorded to assess intestinal motility. Afferent nerve responses were quantified at baseline and to chemical stimulation with bradykinin (0.5 microM) or serotonin (5-HT; 500 microM) and following mechanical stimulation by continuous ramp distension to 60 mmHg. RESULTS Peak amplitudes of intestinal motility and afferent nerve discharge at baseline were not different following chronic vagotomy, alpha-bungarotoxin or sham operation. Maximum afferent discharge to 5-HT following alpha-bungarotoxin was comparable to sham controls, while the response was reduced in chronically vagotomized animals (p < 0.05). Maximum afferent nerve discharge to bradykinin and peak firing during maximum distension at 60 mmHg was similar in the different subgroups. At luminal distension from 10 to 30 mmHg, afferent discharge was lower in vagotomized animals compared to sham controls (p < 0.05) but unchanged after alpha-bungarotoxin. CONCLUSIONS Sensitivity to low-threshold distension and 5-HT is mediated via vagal afferents during postoperative ileus, while sensitivity to high-threshold distension and bradykinin is independent of vagal afferent innervation. Early inhibition of intestinal motility at 3 h after onset of postoperative ileus does not appear to depend on vagal innervation.
Collapse
|
22
|
Differential Sensitization of Afferent Neuronal Pathways During Postoperative Ileus in the Mouse Jejunum. Ann Surg 2008; 247:791-802. [DOI: 10.1097/sla.0b013e31816a9d97] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
23
|
Balsama L, Weese J. Alvimopan: a peripheral acting µ-opioid-receptor antagonist used for the treatment of postoperative ileus. ACTA ACUST UNITED AC 2007. [DOI: 10.2217/14750708.4.5.653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
24
|
Fukuda H, Tsuchida D, Koda K, Miyazaki M, Pappas TN, Takahashi T. Inhibition of sympathetic pathways restores postoperative ileus in the upper and lower gastrointestinal tract. J Gastroenterol Hepatol 2007; 22:1293-9. [PMID: 17688668 DOI: 10.1111/j.1440-1746.2007.04915.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Postoperative ileus (POI) is a transient bowel dysmotility following abdominal surgery. The effects of adrenergic blocking agents and celiac ganglionectomy were studied in rats to investigate the possible involvement of the adrenergic pathway in whole gastrointestinal (GI) transit in the early and late phases of POI. METHODS After laparotomy, the terminal ileum was manipulated for 10 min. (51)Cr was administered into the stomach or colon immediately after surgery. In another group, (51)Cr was administered 24 h after surgery. Three hours after (51)Cr was administered, the rats were killed, and GI and colonic transit was calculated as a geometric center (GC). RESULTS Gastrointestinal transit was significantly delayed 3 h after intestinal manipulation, compared with GI transit in rats that had anesthesia only. Three hours after intestinal manipulation, guanethidine (5 mg/kg) and yohimbine (3 mg/kg) significantly improved delayed GI transit. Celiac ganglionectomy also significantly improved delayed GI transit. Twenty-seven hours after intestinal manipulation, guanethidine, yohimbine and celiac ganglionectomy improved delayed GI transit induced by intestinal manipulation. Colonic transit was delayed 3 h after intestinal manipulation, and delayed colonic transit was partially restored within 27 h of intestinal manipulation. Guanethidine, yohimbine and celiac ganglionectomy improved delayed colonic transit 3 h and 27 h after intestinal manipulation. CONCLUSIONS Adrenoceptors activated by intestinal manipulation impair the motility of the entire GI tract in both the early and the late phase of POI.
Collapse
Affiliation(s)
- Hiroyuki Fukuda
- Department of Surgery, Duke University Medical Center and VA Medical Center, Durham, North Carolina, USA
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
Nausea and vomiting following surgery may either occur as postoperative nausea and vomiting, which is a condition that is mainly related to anesthesia, or as secondary to postoperative ileus, which denotes inhibition of gastrointestinal motility following surgery. Postoperative ileus is a multifactorial event with several contributing mechanisms. Although postoperative nausea and vomiting pathophysiology has been quite well-studied little is known about it. There are multiple targets for treatment, prevention, and its successful empirical management e.g. by 5-HT(3) receptor antagonists. This review describes different aspects of the pathophysiology of postoperative ileus and postoperative nausea and vomiting, their relevance to postoperative care, and the standardized approach to manage postoperative nausea and vomiting that was established by Apfel and coworkers. Despite the recent advances in the understanding and treatment of conditions that trigger nausea and vomiting in the postoperative period, these symptoms remain a significant problem that affects patients' recovery, comfort, and treatment cost.
Collapse
Affiliation(s)
- M E Kreis
- Ludwig-Maximilians Universität, Chirurgische Klinik Grosshadern, Marchioninistrasse, 15 81377, München, Germany.
| |
Collapse
|
26
|
Martinez V, Wang L, Taché Y. Peripheral adrenomedullin inhibits gastric emptying through CGRP8-37 -sensitive receptors and prostaglandins pathways in rats. Peptides 2006; 27:1376-82. [PMID: 16337713 DOI: 10.1016/j.peptides.2005.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2005] [Revised: 11/04/2005] [Accepted: 11/04/2005] [Indexed: 11/24/2022]
Abstract
The effects of intravenous (iv) adrenomedullin (AM) on gastric emptying were investigated in conscious rats. AM induced a maximal 50% inhibition of gastric emptying at a dose of 1.2 nmol/kg. AM was about two-fold less potent than alpha-calcitonin gene-related peptide (alpha-CGRP), which induced a similar 50% maximal inhibition of gastric emptying at 0.6 nmol/kg. Delayed gastric emptying induced by i.v. AM and alpha-CGRP was prevented by peripheral injection of the selective CGRP1 antagonist, CGRP8-37, and by pretreatment with indomethacin, while not altered by blockade of the sympathetic nervous system with propranolol. These data indicate that peripheral AM inhibits gastric emptying through the interaction with CGRP8-37 -sensitive receptors, likely CGRP1 receptors, and the recruitment of prostaglandin-dependent mechanisms.
Collapse
Affiliation(s)
- V Martinez
- CURE: Digestive Diseases Research Center, Department of Medicine, David Geffen School of Medicine at UCLA, University of California Los Angeles and VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA.
| | | | | |
Collapse
|
27
|
Mueller MH, Kampitoglou D, Glatzle J, Hahn J, Kreis ME. Systemic capsaicin inhibits neuronal activation in the brainstem during postoperative ileus in the mouse. Langenbecks Arch Surg 2006; 391:88-95. [PMID: 16572327 DOI: 10.1007/s00423-006-0042-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2005] [Accepted: 03/03/2006] [Indexed: 10/24/2022]
Abstract
INTRODUCTION Neuronal inhibitory reflex mechanisms contribute to postoperative ileus after abdominal surgery. During this condition, sensory neurons in the brainstem are activated. We aimed to determine the contribution of capsaicin-sensitive afferents to central vagal sensitivity in mice during postoperative ileus. MATERIALS AND METHODS Under enflurane anesthesia, C57BL/6 mice were laparotomized and the small bowel was manipulated to induce ileus or was left untouched as a sham-treatment group. A subgroup of ileus animals was pre-treated with Capsaicin (1 microm/kg, i.p.) 48 h before small bowel manipulation. The animals were killed 24 h later and the brainstem was removed for Fos immunohistochemistry, which was quantified in the nucleus of the solitary tract (nTS). Spontaneous jejunal motility was recorded in vitro. Leukocyte infiltration in the intestinal muscularis was studied by myeloperoxidase staining as an index of postoperative inflammation. RESULTS There were 30+/-9 Fos-positive neurons counted in the nTS after ileus and 6+/-2 in sham controls (Bregma -7.70 mm, P=0.01). A reduction to 8+/-3 was observed after Capsaicin pre-treatment in ileus animals (P<0.05). Peak amplitudes of spontaneous jejunal motility were 2+/-0.3 cmH2O during postoperative ileus, 3+/-0.6 cmH2O after ileus with capsaicin pre-treatment, and 10+/-2 cmH2O in control animals (N=6, both P<0.05). The number of leukocytes infiltrating the muscularis was 39+/-9/mm2 during ileus and 1.8+/-1/mm2 in controls (mean+/-SEM, P<0.01, N=6). After capsaicin, this number increased to 72+/-28/mm2 in ileus animals (P<0.05 vs control animals, N=7). CONCLUSION The inhibition of capsaicin-sensitive vagal afferent pathways appears to boost rather than to attenuate the inflammatory response during postoperative ileus, while intestinal motility remained unchanged. This suggests a protective role of the capsaicin-sensitive afferent innervation for the inflammatory phase of postoperative ileus.
Collapse
Affiliation(s)
- Mario H Mueller
- Department of Surgery, Ludwig-Maximilian University, Munich, Germany
| | | | | | | | | |
Collapse
|
28
|
Affiliation(s)
- Benjamin Person
- Department of Colorectal Surgery, Cleveland Clinic Florida, Weston, FL, USA
| | | |
Collapse
|
29
|
Fukuda H, Tsuchida D, Koda K, Miyazaki M, Pappas TN, Takahashi T. Impaired gastric motor activity after abdominal surgery in rats. Neurogastroenterol Motil 2005; 17:245-50. [PMID: 15787944 DOI: 10.1111/j.1365-2982.2004.00602.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Postoperative ileus (POI) is a transient bowel dysmotility that occurs following abdominal surgery. Several mechanisms have been proposed such as neural reflex and inflammatory changes. We focused on gastric motility after abdominal surgery in rats. To investigate the time course of gastric motility after surgery, gastric motility was continuously recorded before, during and after surgery. After laparotomy, terminal ileum was manipulated for 10 min. Gastric motility was recorded by a strain gauge transducer implanted on the serosal surface of the stomach. To investigate whether peripheral sympathetic nerve is involved in the pathogenesis of POI, effects of guanethidine and celiac ganglionectomy were tested on the postoperative gastric motility. Although isoflurane anaesthesia reduced the gastric motility to 40%, the motility recovered immediately when isoflurane was withdrawn. Intestinal manipulation reduced the postoperative gastric motility for 3-24 h after surgery, compared with preoperative levels. Guanethidine administration and celiac ganglionectomy restored the impaired gastric motility. Feeding increased the gastric motility in each group. It is suggested that the pathogenesis of postoperative gastric ileus induced by intestinal manipulation involves viscero-sympathetic pathways. Intestinal manipulation causes impaired gastric motility via inhibitory sympathetic efferent pathway. Feeding may improve the postoperative gastric motility.
Collapse
Affiliation(s)
- H Fukuda
- Department of Surgery, Duke University Medical Center and VA Medical Center, Durham, NC 27705, USA
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Postoperative ileus is an iatrogenic condition that follows abdominal surgery. Three main mechanisms are involved in its causation, namely neurogenic, inflammatory and pharmacological mechanisms. In the acute postoperative phase, mainly spinal and supraspinal adrenergic and non-adrenergic pathways are activated. Recent studies, however, show that the prolonged phase of postoperative ileus is caused by an enteric molecular inflammatory response and the subsequent recruitment of leucocytes into the muscularis of the intestinal segments manipulated during surgery. This inflammation impairs local neuromuscular function and activates neurogenic inhibitory pathways, inhibiting motility of the entire gastrointestinal tract. The mechanisms underlying the recruitment of the inflammatory cells, and their interaction with the intestinal afferent innervation, are discussed. Finally, opioids administered for postoperative pain control also contribute to a large extent to the reduction in propulsive gastrointestinal motility observed after abdominal surgery.
Collapse
Affiliation(s)
- A J Bauer
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| | | |
Collapse
|
31
|
de Jonge WJ, The FO, van der Coelen D, Bennink RJ, Reitsma PH, van Deventer SJ, van den Wijngaard RM, Boeckxstaens GE. Mast cell degranulation during abdominal surgery initiates postoperative ileus in mice. Gastroenterology 2004; 127:535-45. [PMID: 15300586 DOI: 10.1053/j.gastro.2004.04.017] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Inflammation of the intestinal muscularis following manipulation during surgery plays a crucial role in the pathogenesis of postoperative ileus. Here, we evaluate the role of mast cell activation in the recruitment of infiltrates in a murine model. METHODS Twenty-four hours after control laparotomy or intestinal manipulation, gastric emptying was determined. Mast cell degranulation was determined by measurement of mast cell protease-I in peritoneal fluid. Intestinal inflammation was assessed by determination of tissue myeloperoxidase activity and histochemical staining. RESULTS Intestinal manipulation elicited a significant increase in mast cell protease-I levels in peritoneal fluid and resulted in recruitment of inflammatory infiltrates to the intestinal muscularis. This infiltrate was associated with a delay in gastric emptying 24 hours after surgery. Pretreatment with mast cell stabilizers ketotifen (1 mg/kg, p.o.) or doxantrazole (5 mg/kg, i.p.) prevented both manipulation-induced inflammation and gastroparesis. Reciprocally, in vivo exposure of an ileal loop to the mast cell secretagogue compound 48/80 (0.2 mg/mL for 1 minute) induced muscular inflammation and delayed gastric emptying. The manipulation-induced inflammation was dependent on the presence of mast cells because intestinal manipulation in mast cell-deficient Kit/Kitv mice did not elicit significant leukocyte recruitment. Reconstitution of Kit/Kitv mice with cultured bone marrow-derived mast cells from congenic wild types restored the manipulation-induced inflammation. CONCLUSIONS Our results show that degranulation of connective tissue mast cells is a key event for the establishment of the intestinal infiltrate that mediates postoperative ileus following abdominal surgery.
Collapse
Affiliation(s)
- Wouter J de Jonge
- Department of Gastroenterology and Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
PURPOSE Postoperatively, some patients experience a prolonged inhibition of coordinated bowel activity, which causes accumulation of secretions and gas, resulting in nausea, vomiting, abdominal distension, and pain. This prolonged inhibition can take days or weeks to resolve and often is referred to as postoperative paralytic ileus lasting more than three days after surgery. This article reviews the etiology, pathophysiology, and treatment options of postoperative ileus. METHODS The relevant literature from 1965 to 2003 was identified and reviewed using MEDLINE database of the U.S. Medical Library of Medicine. Both retrospective and prospective studies were included in this review. RESULTS The pathophysiology of postoperative ileus is multifactorial. The duration of postoperative ileus correlates with the degree of surgical trauma and is most extensive after colonic surgery. However, postoperative ileus can develop after all types of surgery including extraperitoneal surgery. A variety of treatment options have been used to decrease the duration of postoperative ileus. However, it is difficult to compare these studies because of small sample sizes and differences in operations performed, anesthesia protocols provided both intraoperatively and postoperatively, patient comorbidities, and in the measured end points, such as the time to the presence of bowel sounds, flatus, or bowel movements, tolerance of solid food, or discharge from the hospital. However, despite these drawbacks, some conclusions can be made. CONCLUSIONS Paralytic postoperative ileus continues to be a significant problem after abdominal and other types of surgery. The etiology is multifactorial and is best treated with a combination of different approaches. Currently, the important factors that could effect the duration and recovery from postoperative ileus include limitation of narcotic use by substituting alternative medications such as nonsteroidals and placing a thoracic epidural with local anesthetic when possible. The selective use of nasogastric decompression and correction of electrolyte imbalances also are important factors to consider.
Collapse
Affiliation(s)
- Mirza K Baig
- Department of Colorectal Surgery, Cleveland Clinic Florida, Weston, Florida 33331, USA
| | | |
Collapse
|
33
|
Hang CH, Shi JX, Li JS, Wu W, Li WQ, Yin HX. Levels of vasoactive intestinal peptide, cholecystokinin and calcitonin gene-related peptide in plasma and jejunum of rats following traumatic brain injury and underlying significance in gastrointestinal dysfunction. World J Gastroenterol 2004; 10:875-80. [PMID: 15040036 PMCID: PMC4727008 DOI: 10.3748/wjg.v10.i6.875] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To study the alterations of brain-gut peptides following traumatic brain injury (TBI) and to explore the underlying significance of these peptides in the complicated gastrointestinal dysfunction.
METHODS: Rat models of focal traumatic brain injury were established by impact insult method, and divided into 6 groups (6 rats each group) including control group with sham operation and TBI groups at postinjury 3, 12, 24, 72 h, and d 7. Blood and proximal jejunum samples were taken at time point of each group and gross observations of gastrointestinal pathology were recorded simultaneously. The levels of vasoactive intestinal peptide (VIP) in plasma, calcitonin gene-related peptide (CGRP) and cholecystokinin (CCK) in both plasma and jejunum were measured by enzyme immunoassay (EIA). Radioimmunoassay (RIA) was used to determine the levels of VIP in jejunum.
RESULTS: Gastric distension, delayed gastric emptying and intestinal dilatation with a large amount of yellowish effusion and thin edematous wall were found in TBI rats through 12 h and 72 h, which peaked at postinjury 72 h. As compared with that of control group (247.8 ± 29.5 ng/L), plasma VIP levels were significantly decreased at postinjury 3, 12 and 24 h (106.7 ± 34.1 ng/L, 148.7 ± 22.8 ng/L, 132.8 ± 21.6 ng/L, respectively), but significantly increased at 72 h (405.0 ± 29.8 ng/L) and markedly declined on d 7 (130.7 ± 19.3 ng/L). However, Plasma levels CCK and CGRP were significantly increased through 3 h and 7 d following TBI (126-691% increases), with the peak at 72 h. Compared with control (VIP, 13.6 ± 1.4 ng /g; CGRP, 70.6 ± 17.7 ng/g); VIP and CGRP levels in jejunum were significantly increased at 3 h after TBI (VIP, 35.4 ± 5.0 ng/g; CGRP, 103.8 ± 22.1 ng/g), and declined gradually at 12 h and 24 h (VIP, 16.5 ± 1.8 ng/g, 5.5 ± 1.4 ng/g; CGRP, 34.9 ± 9.7 ng/g, 18.5 ± 7.7 ng/g), but were significantly increased again at 72 h (VIP, 48.7 ± 9.5 ng/g; CGRP, 142.1 ± 24.3 ng/g), then declined in various degrees on d 7 (VIP, 3.8 ± 1.1 ng/g; CGRP, 102.5 ± 18.1 ng/g). The CCK levels in jejunum were found to change in a similar trend as that in plasma with the concentrations of CCK significantly increased following TBI (99-517% increases) and peaked at 72 h.
CONCLUSION: Traumatic brain injury can lead to significant changes of brain-gut peptides in both plasma and small intestine, which may be involved in the pathogenesis of complicated gastrointestinal dysfunction.
Collapse
Affiliation(s)
- Chun-Hua Hang
- Department of Neurosurgery, Jinling Hospital, 305 East Zhongshan Road, Nanjing 210002, Jiangsu Province, China.
| | | | | | | | | | | |
Collapse
|
34
|
Kreiss C, Birder LA, Kiss S, VanBibber MM, Bauer AJ. COX-2 dependent inflammation increases spinal Fos expression during rodent postoperative ileus. Gut 2003; 52:527-34. [PMID: 12631664 PMCID: PMC1773580 DOI: 10.1136/gut.52.4.527] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/15/2002] [Indexed: 12/08/2022]
Abstract
BACKGROUND AND AIMS Cyclooxygenase 2 (COX-2) and prostaglandins (PGs) participate in the pathogenesis of inflammatory postoperative ileus. We sought to determine whether the emerging neuronal modulator COX-2 plays a significant role in primary afferent activation during postoperative ileus using spinal Fos expression as a marker. METHODS Rats, and COX-2(+/+) and COX-2(-/-) mice underwent simple intestinal manipulation. The effect of intestinal manipulation on Fos immunoreactivity (IR) in the L(5)-S(1) spinal cord, in situ circumference, and postoperative leucocytic infiltrate of the intestinal muscularis was measured. Postoperative PGE(2) production was measured in peritoneal lavage fluid. The dependence of these parameters on COX-2 was studied in pharmacological (DFU, Merck- Frosst, selective COX-2 inhibitor) and genetic (COX-2(-/-) mice) models. RESULTS Postoperative Fos IR increased 3.7-fold in rats and 2.2-fold in mice. Both muscularis leucocytic infiltrate and the circumference of the muscularis increased significantly in rats and COX-2(+/+) mice postoperatively, indicating dilating ileus. Surgical manipulation markedly increased PGE(2) levels in the peritoneal cavity. DFU pretreatment and the genetic absence of COX-2(-/-) prevented dilating ileus, and leucocytic infiltrate was diminished by 40% with DFU and by 54% in COX-2(-/-) mice. DFU reversed postsurgical intra- abdominal PGE(2) levels to normal. Fos IR after intestinal manipulation was attenuated by approximately 50% in DFU treated rats and in COX-2(-/-) mice. CONCLUSIONS Postoperatively, small bowel manipulation causes a significant and prolonged increase in spinal Fos expression, suggesting prolonged primary afferent activation. COX-2 plays a key role in this response. This activation of primary afferents may subsequently initiate inhibitory motor reflexes to the gut, contributing to postoperative ileus.
Collapse
Affiliation(s)
- C Kreiss
- Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | |
Collapse
|
35
|
Zittel TT, Meile T, Huge A, Kreis ME, Becker HD, Jehle EC. Preoperative intraluminal application of capsaicin increases postoperative gastric and colonic motility in rats. J Gastrointest Surg 2001; 5:503-13. [PMID: 11986001 DOI: 10.1016/s1091-255x(01)80088-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
In a model to investigate postoperative gastrointestinal motility with strain gauge transducers in awake rats, we tested the effects of intraluminal capsaicin infusion into the cecum 2 days or 14 days prior to abdominal surgery. Acute infusion of capsaicin into the cecum for 30 minutes increased the gastric, small intestinal, and colonic motility index by up to 115%, 34%, and 59%, respectively, compared to vehicle infusion. Intraluminal capsaicin infusion 2 days prior to abdominal surgery significantly increased the intraoperative gastric and colonic motility index by 166% and 100%, respectively, compared to vehicle, but had no effect on small intestinal motility. The postoperative decrease in gastric or colonic motility was completely prevented by capsaicin pretreatment, representing a 73% and a 72% increase in the motility index during the first postoperative hour and a 40% and a 29% increase in the motility index during the second postoperative hour compared to vehicle, whereas the postoperative decrease in small intestinal motility was not altered by capsaicin pretreatment. In contrast, intraluminal capsaicin infusion 14 days prior to abdominal surgery had no effect on postoperative inhibition of gastrointestinal motility. Our results suggest that capsaicin-sensitive visceral afferent C-fibers, presumably of the submucosa, play an important role in mediating postoperative ileus. Intraluminal capsaicin does probably ablate these nerve fibers temporarily, with no systemic side effects observed with the use of the tail flick test as a measure of skin nociception.
Collapse
Affiliation(s)
- T T Zittel
- Department of General and Transplantation Surgery, University Hospital, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany.
| | | | | | | | | | | |
Collapse
|
36
|
Gschossmann JM, Coutinho SV, Miller JC, Huebel K, Naliboff B, Wong HC, Walsh JH, Mayer EA. Involvement of spinal calcitonin gene-related peptide in the development of acute visceral hyperalgesia in the rat. Neurogastroenterol Motil 2001; 13:229-36. [PMID: 11437985 DOI: 10.1046/j.1365-2982.2001.00262.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This study aimed to characterize the role of the neuropeptide calcitonin gene-related peptide (CGRP) in the development of mechanically induced visceral hyperalgesia. Tonic colorectal distension (CRD) was performed in fasted, conscious male Sprague-Dawley rats. The visceromotor reflex associated with noxious CRD was determined as the number of contractions during each of two consecutive tonic distensions (10 min at 60 mmHg), which were separated by a series of phasic distensions (repeated 15-s distensions to 80 mmHg at 30-s intervals). The effect of the CGRP receptor antagonist h-CGRP8-37 given intrathecally (i.t.) (0.03-3 nmol rat-1) or intravenously (i.v.) (20 microg kg-1 bodyweight [bw]) on the visceromotor response was evaluated. The dose for i.v. administration was chosen based on previous results from similar studies. In addition, the effect of a CGRP monoclonal antibody (6 mg kg-1 bw) given intravenously was evaluated. Compared to the baseline response, a significant increase in the number of abdominal contractions was observed during the second tonic distension. The i.t. application of h-CGRP8-37 dose-dependently reduced the numbers of abdominal contractions both during the first and the second tonic distension period, with a maximum effect observed at a peptide concentration of 3 nmol. Intravenous administration of h-CGRP8-37 or of the CGRP antiserum produced a small reduction of the visceromotor response induced by the second tonic distension and had no effect on colonic compliance. The development of mechanically induced colorectal hyperalgesia by repeated tonic distension involves the spinal release of CGRP, while peripheral release of CGRP plays only a minor role.
Collapse
Affiliation(s)
- J M Gschossmann
- CURE Digestive Diseases Research Center/Neuroenteric Disease Program and Animal Models Core, Departments of Medicine and Physiology and Brain Research Institute, UCLA School of Medicine, and VA Wadsworth Medical Center, Los Angeles, CA 90073, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
BACKGROUND Postoperative ileus has traditionally been accepted as a normal response to tissue injury. No data support any beneficial effect of ileus and indeed it may contribute to delayed recovery and prolonged hospital stay. Efforts should, therefore, be made to reduce such ileus. METHODS Material was identified from a Medline search of the literature, previous review articles and references cited in original papers. This paper updates knowledge on the pathophysiology and treatment of postoperative ileus. RESULTS AND CONCLUSION Pathogenesis mainly involves inhibitory neural reflexes and inflammatory mediators released from the site of injury. The most effective method of reducing ileus is thoracic epidural blockade with local anaesthetic. Opioid-sparing analgesic techniques and non-steroidal anti-inflammatory agents also reduce ileus, as does laparoscopic surgery. Of the prokinetic agents only cisapride is proven beneficial; the effect of early enteral feeding remains unclear. However, postoperative ileus may be greatly reduced when all of the above are combined in a multimodal rehabilitation strategy.
Collapse
Affiliation(s)
- K Holte
- Department of Surgical Gastroenterology, Hvidovre University Hospital, Denmark
| | | |
Collapse
|
38
|
Abstract
Peptides involved in the endocrine and enteric nervous systems as well as in the central nervous system exert concerted action on gastrointestinal motility. Mechanical and chemical stimuli which induce peptide release from the epithelial endocrine cells are the earliest step in the initiation of peristaltic activities. Gut peptides exert hormonal effects, but peptide-containing stimulatory (Ach/substance P/tachykinin) and inhibitory (VIP/PACAP/NO) neurons are also involved in the induction of ascending contraction and descending relaxation, respectively. The dorsal vagal complex (DVC), located in the medulla of the brainstem, constitutes the basic neural circuitry of vago-vagal reflex control of gastrointestinal motility. Several gut peptides act on the DVC to modify vagal cholinergic reflexes directly (PYY and PP) or indirectly via afferent fibers in the periphery (CCK and GLP-1). The DVC is also a primary site of action of many neuropeptides (such as TRH and NPY) in mediating gastrointestinal motor activities. The identification over the last few years of a number of neuropeptide systems has greatly changed the field of feeding and body weight regulation. By exploring the brain and gut systems that employ recently identified peptidergic molecules, it will be possible to elaborate on the central and peripheral pathways involved in the regulation of gastrointestinal motility.
Collapse
Affiliation(s)
- M Fujimiya
- Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | |
Collapse
|
39
|
Boeckxstaens GE, Hollmann M, Heisterkamp SH, Robberecht P, de Jonge WJ, van Den Wijngaard RM, Tytgat GN, Blommaart PJ. Evidence for VIP(1)/PACAP receptors in the afferent pathway mediating surgery-induced fundic relaxation in the rat. Br J Pharmacol 2000; 131:705-10. [PMID: 11030719 PMCID: PMC1572383 DOI: 10.1038/sj.bjp.0703625] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
We previously reported activation of an inhibitory adrenergic and a non-adrenergic non-cholinergic (NANC) pathway during abdominal surgery relaxing the rat gastric fundus. In the present study, we investigated the possible role of nitric oxide (NO) and vasoactive intestinal polypeptide (VIP) in the NANC part of the surgery-induced fundic relaxation. The effect of the NO biosynthesis inhibitor N(G)-nitro-L-arginine (L-NOARG), the non-selective VIP receptor antagonist [D-p-Cl-Phe(6),Leu(17)]-VIP and the selective VIP(1) receptor antagonist [Acetyl-His(1),D-Phe(2),Lys(15),Arg(16), Leu(17)]-VIP was investigated on the non-adrenergic fundic relaxation induced by manipulation of the small intestine followed by resection of the caecum. Guanethidine partly reduced the manipulation-induced fundic relaxation. Addition of L-NOARG reduced this non-adrenergic component, whereas the non-selective VIP receptor antagonist had no significant effect. Combination of L-NOARG and the non-selective VIP antagonist however further reduced the relaxation to manipulation. The selective VIP(1) receptor antagonist reduced the mean and maximal relaxation induced by abdominal surgery in the presence of guanethidine. When combined with L-NOARG, the relaxation of the gastric fundus was almost completely abolished. The VIP(1) receptor antagonist alone had no significant effect on the mean and maximal relaxation, but enhanced recovery of fundic tone. In conclusion, as VIP(1) receptors are not present in the rat gastric fundus, these results suggest that the NANC inhibitory pathway activated during abdominal surgery involves VIP(1) receptors, most likely in the afferent limb. The inhibitory neurotransmitters released at the level of the gastric fundus smooth muscle are NO and a substance different from VIP.
Collapse
Affiliation(s)
- G E Boeckxstaens
- Division of Gastroenterology & Hepatology, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Boeckxstaens GE, Hirsch DP, Kodde A, Moojen TM, Blackshaw A, Tytgat GN, Blommaart PJ. Activation of an adrenergic and vagally-mediated NANC pathway in surgery-induced fundic relaxation in the rat. Neurogastroenterol Motil 1999; 11:467-74. [PMID: 10583854 DOI: 10.1046/j.1365-2982.1999.00172.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The aim of this study was to pharmacologically characterize and investigate the possible contribution of adrenergic and nonadrenergic noncholinergic (NANC) pathways involved in the relaxation of the rat gastric fundus following abdominal surgery. Using an intragastric balloon, the effect of skin incision (SI), laparotomy (LT) and manipulation of the small intestine followed by caecal resection (M + R) on fundic pressure was evaluated. SI resulted in a brief relaxation of the gastric fundus abolished by guanethidine and blocked by hexamethonium and the combination of phentolamine, propranolol and atropine (PPA). LT induced a longer lasting relaxation which was abolished by guanethidine and hexamethonium. It was blocked by PPA and the combination of ganglionectomy and vagotomy, but unaffected by atropine, vagotomy or ganglionectomy. M + R induced a long-lasting relaxation which was only partly blocked by guanethidine or PPA, illustrating an inhibitory NANC component. Vagotomy combined with guanethidine completely abolished the relaxation following M + R, whereas it was significantly blocked by hexamethonium and the combination of ganglionectomy with vagotomy. These results indicate that SI, LT and M + R induce inhibition of fundic motility via an adrenergic mechanism. During M + R, an additional vagally mediated inhibitory NANC pathway is activated. Finally, we suggest that LT and M + R inhibit the gastric fundus via both a splanchnic and a vagal reflex pathway.
Collapse
Affiliation(s)
- G E Boeckxstaens
- Division of Gastroenterology & Hepatology, Academic Medical Centre, Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
41
|
Freeman ME, Cheng G, Hocking MP. Role of alpha- and beta-calcitonin gene-related peptide in postoperative small bowel ileus. J Gastrointest Surg 1999; 3:39-43. [PMID: 10457322 DOI: 10.1016/s1091-255x(99)80006-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Ablation of a-calcitonin gene-related polypeptide (CGRP) containing neurons with the afferent neurotoxin capsaicin improves postoperative foregut transit in a rodent model. Similarly, administration of a selective alpha-CGRP antibody or hCGRP((8-37)), a CGRP receptor antagonist, improves postoperative gastric emptying. Unlike the stomach, which contains only alpha-CGRP, the small bowel additionally contains beta-CGRP. The role of the latter in postoperative small bowel transit is unknown. The purpose of this study was to evaluate the effect of an alpha-CGRP antibody and hCGRP((8-37)) on postoperative small bowel transit. Male Sprague-Dawley rats underwent placement of duodenal catheters and were randomly assigned to 1 of 11 groups. Four groups were pretreated with 1% capsaicin. One week later, all animals underwent standardized laparotomy following administration of a control antibody or the alpha-CGRP mono-clonal antibody, or during infusion of hCGRP((8-37)) at varying doses. Small bowel transit was measured 25 minutes postoperatively. The alpha-CGRP antibody sped postoperative transit when given alone or in combination with capsaicin. In contrast, animals treated with hCGRP((8-37)) showed no significant improvement in postoperative transit, and the beneficial effect of capsaicin was blocked. Unlike their similar effects on postoperative gastric emptying, we found that hCGRP((8-37)) and the alpha-CGRP antibody had differing effects on postoperative small bowel transit. The reason for this is unknown but may be related to their differing specificities for alpha- and beta-CGRP.
Collapse
Affiliation(s)
- M E Freeman
- Department of Surgery, VA Medical Center and the University of Florida, Gainesville, Florida 32610, USA
| | | | | |
Collapse
|
42
|
Zittel TT, Lloyd KC, Rothenhöfer I, Wong H, Walsh JH, Raybould HE. Calcitonin gene-related peptide and spinal afferents partly mediate postoperative colonic ileus in the rat. Surgery 1998; 123:518-27. [PMID: 9591004 DOI: 10.1067/msy.1998.88090] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Calcitonin gene-related peptide (CGRP) is a widely distributed neuropeptide contained in intrinsic and extrinsic neurons of the gastrointestinal wall that has been shown to be released by noxious stimulation, to be involved in nociception, to inhibit gastrointestinal motility, and to partly mediate postoperative gastric ileus. We hypothesized that abdominal surgery-induced release of CGRP might inhibit postoperative colonic motility and food intake. METHODS Colonic transit, stool pellet number, stool pellet weight, and food intake were measured for 48 hours after induction of postoperative ileus in rats. CGRP was immunoneutralized by preoperative injection of CGRP monoclonal antibody, or visceral afferent nerve fibers containing CGRP were functionally ablated by topical capsaicin treatment of the vagus nerves or of the celiac/superior mesenteric ganglia before abdominal surgery. RESULTS Abdominal surgery increased colonic transit time and decreased 24-hour cumulative stool pellet number, stool pellet weight, and food intake. CGRP immunoneutralization reversed postoperative inhibition of colonic transit, 24-hour cumulative stool pellet number, stool pellet weight, and food intake by 77%, 82%, 80%, and 52%, respectively. Whereas ablation of vagal afferent nerve fibers had no effect, spinal afferent nerve fiber ablation reversed postoperative inhibition of 24-hour cumulative stool pellet number, stool pellet weight, and food intake by 41%, 38%, and 19%, respectively. CONCLUSIONS CGRP and spinal afferent nerve fibers partly mediate postoperative colonic ileus and inhibition of food intake in the rat. By the magnitude of reversal of postoperative ileus, CGRP seems to be an important mediator of postoperative colonic ileus. Our results for the first time show involvement of a neuropeptide and spinal afferents in the mediation of postoperative colonic ileus and postoperative inhibition of food intake in rats.
Collapse
Affiliation(s)
- T T Zittel
- CURE/Gastroenteric Biology Center, West Los Angeles VA Medical Center, Calif., USA
| | | | | | | | | | | |
Collapse
|
43
|
Abstract
The maintenance of gastric mucosal integrity depends on the rapid alarm of protective mechanisms in the face of pending injury. Afferent neurons of extrinsic origin constitute an emergency system that is called into operation when the gastric mucosa is endangered by acid and other noxious chemicals. The function of these chemoceptive afferents can be manipulated selectively and explored with the excitotoxin capsaicin. Most of the homeostatic actions of capsaicin-sensitive afferents are brought about by peptides released from their peripheral endings in the gastric wall. When stimulated, chemoceptive afferents enhance gastric blood flow and activate hyperemia-dependent and hyperemia-independent mechanisms of protection and repair. In the rodent stomach, these local regulatory roles of sensory neurons are mediated by calcitonin gene-related peptide acting via calcitonin gene-related peptide 1 receptors and neurokinin A acting via neurokinin 2 receptors, with both peptides using nitric oxide as their common messenger. In addition, capsaicin-sensitive neurons form the afferent arc of autonomic reflexes that control secretory and motor functions of the stomach. The pathophysiological potential of the neural emergency system is best portrayed by the gastric hyperemic response to acid backdiffusion, which is signaled by afferent nerve fibers. This mechanism limits damage to the surface of the mucosa and creates favorable conditions for rapid restitution and healing of the wounded mucosa.
Collapse
Affiliation(s)
- P Holzer
- Department of Experimental and Clinical Pharmacology, University of Graz, Graz, Austria
| |
Collapse
|
44
|
Huge A, Kreis ME, Jehle EC, Ehrlein HJ, Starlinger M, Becker HD, Zittel TT. A model to investigate postoperative ileus with strain gauge transducers in awake rats. J Surg Res 1998; 74:112-8. [PMID: 9587348 DOI: 10.1006/jsre.1997.5245] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Postoperative ileus influences patients well-being, hospital stay, and health cost, and postoperative inhibition of colonic motility is a major contributor to postoperative ileus. Experimental models for investigating postoperative ileus are needed. In particular, recording of postoperative colonic motility in awake rats has not been described yet. MATERIAL AND METHODS Gastric, small intestinal, and colonic motility were recorded with strain gauge transducers in awake rats, and the effects of anesthesia and abdominal surgery on gastrointestinal motility were investigated. RESULTS Ether anesthesia increased gastric motility and inhibited small intestinal motility, while enflurane anesthesia had only minor effects on gastrointestinal motility. Abdominal surgery inhibited gastric, small intestinal, and colonic motility, and a detailed analysis of gastrointestinal motility in our postoperative ileus model is given. CONCLUSIONS We established a model to record gastric, small intestinal, and colonic motility in awake rats postoperatively. We could demonstrate that enflurane anesthesia had little effect on gastrointestinal motility, while laparotomy and short manipulation of the cecum produced a prolonged inhibition of gastrointestinal motility. Our model could be used to investigate postoperative ileus, particularly of the colon, in awake rats.
Collapse
Affiliation(s)
- A Huge
- University Hospital, Department of General and Transplantation Surgery, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
van Berge Henegouwen MI, van Gulik TM, Moojen TM, Boeckxstaens GE, Gouma DJ. Gastrointestinal motility after pancreatoduodenectomy. SCANDINAVIAN JOURNAL OF GASTROENTEROLOGY. SUPPLEMENT 1998. [PMID: 9515753 DOI: 10.1080/003655298750026552] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Pancreatoduodenectomy (PD) is a major surgical procedure which is accompanied by a high morbidity of between 30 and 50%. A large part of this morbidity is caused by delayed gastric emptying (DGE), which is reported to have an incidence of between 30 and 40% and is associated with prolonged hospital stay. Several pathophysiological mechanisms are thought to cause this complication. Peroperative trauma of the pylorus and the occurrence of intra-abdominal abscesses play a role. Neuronal changes and disruption of the gastrointestinal (GI) intramural nervous plexus may be especially important regarding the pivotal role of the duodenum in the initiation and coordination of antroduodenal motor activity. Another important factor is the postoperative administration of enteral nutrition. Recently, it was demonstrated that cyclic enteral nutrition through a catheter jejunostomy led to a faster return to normal diet and shorter hospital stay than patients on continuous enteral nutrition; this might be partly caused by continuously elevated cholecystokinin levels. The effect of prokinetic agents has not been studied extensively, but a beneficial action on the return of postoperative gastric function after gastrointestinal surgery seems limited.
Collapse
|
46
|
Friese N, Diop L, Chevalier E, Angel F, Rivière PJ, Dahl SG. Involvement of prostaglandins and CGRP-dependent sensory afferents in peritoneal irritation-induced visceral pain. REGULATORY PEPTIDES 1997; 70:1-7. [PMID: 9250575 DOI: 10.1016/s0167-0115(97)02141-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
This study investigates the contribution of prostaglandins (PG) and calcitonin gene-related peptide (CGRP) pathways in visceral pain induced by peritoneal irritation in rats. Peritoneal irritation was produced by i.p. administration of acetic acid (AA: 0.06-1.0%, 10 ml/kg). Visceral pain was scored by counting abdominal contractions. The effect of CGRP (3-100 microg/kg, i.p.) was also evaluated. Like AA, CGRP induced abdominal pain. Neonatal pretreatment with capsaicin reduced abdominal contractions produced by AA (0.6%) and CGRP (20 microg/kg) with 64.6% and 45.6%, respectively. Abdominal contractions induced by AA and CGRP were blocked by two antinociceptive drugs, mu-and kappa-opioid agonists, morphine and (+/-)-U-50,488H, respectively. Indomethacin (3 mg/kg, s.c.) reduced the number of abdominal contractions produced by AA by 78.1%+/-6.4% but did not inhibit abdominal contractions produced by CGRP. The CGRP, receptor antagonist, hCGRP(8-37) (300 microg/kg, i.v.) inhibited AA- and CGRP-induced abdominal contractions with 57.5%+/-12.4% and 51.6%+/-11.3%, respectively. Concomitant i.p. administration of PGE1 and PGE2 (0.3 mg/kg of each) produced abdominal contractions which were inhibited 45.6%+/-9.3% by hCGRP(8-37) (300 microg/kg i.v.). Taken together, these results suggest that peritoneal irritation is likely to trigger the release of prostaglandins, which in turn produces a release of CGRP from primary sensory afferents.
Collapse
Affiliation(s)
- N Friese
- Institut de Recherche Jouveinal, Fresnes, France
| | | | | | | | | | | |
Collapse
|
47
|
Raybould HE, Zittel TT. Inhibition of gastric motility induced by intestinal glucose in awake rats: role of Na(+)-glucose co-transporter. Neurogastroenterol Motil 1995; 7:9-14. [PMID: 7627868 DOI: 10.1111/j.1365-2982.1995.tb00203.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Carbohydrates are a component of chyme that initiate feedback control of gastric emptying. The aim of the study was to investigate the mechanism by which sensors in the intestine are activated by carbohydrate to initiate intestinal feedback of gastric motor function. Intestinal perfusion with D-glucose inhibited gastric motility in awake rats. This response was reproduced by 3-O-methyl glucose, a non-metabolizable analogue of glucose that is absorbed by the Na(+)-glucose co-transporter, but not by 2-deoxy-D-glucose, and was attenuated by perfusion of the intestine with phloridzin, a competitive blocker of the Na(+)-glucose co-transporter. Feeding a high carbohydrate diet to increase the number of co-transporters reduced the response to intestinal glucose. It was concluded that activation of sensors to initiate feedback inhibition of gastric motility may be dependent either on rapid accumulation of glucose within epithelial cells or on activation of the Na(+)-glucose co-transporter.
Collapse
Affiliation(s)
- H E Raybould
- CURE: VA/UCLA Gastroenteric Biology Research Center, VA Medical Center West Los Angeles, Department of Medicine, California, USA
| | | |
Collapse
|