1
|
Halder N, Yadav S, Lal G. Neuroimmune communication of the cholinergic system in gut inflammation and autoimmunity. Autoimmun Rev 2024; 23:103678. [PMID: 39500481 DOI: 10.1016/j.autrev.2024.103678] [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: 10/01/2024] [Revised: 11/01/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Neuroimmune communication in the body forms a bridge between two central regulatory systems of the body, i.e., nervous and immune systems. The cholinergic system is a crucial modulatory neurotransmitter in the central and peripheral nervous system. It includes the neurotransmitter acetylcholine (ACh), the enzyme required for the synthesis of ACh (choline acetyltransferase, ChAT), the enzyme required for its degradation (acetylcholinesterase, AChE), and cholinergic receptors (Nicotinic acetylcholine receptors and muscarinic acetylcholine receptors). The cholinergic system in neurons is well known for its role in cognitive function, sensory perception, motor control, learning, and memory processes. It has been shown that the non-neuronal cholinergic system (NNCS) is present in various tissues and immune cells and forms a neuroimmune communications system. In the present review, we discussed the NNCS on immune cells, its role in homeostasis and inflammatory reactions in the gut, and how it can be exploited in treating inflammatory responses.
Collapse
Affiliation(s)
- Namrita Halder
- Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), SPPU campus, Ganeshkhind, Pune, MH-411007, India
| | - Sourabh Yadav
- Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), SPPU campus, Ganeshkhind, Pune, MH-411007, India
| | - Girdhari Lal
- Biotechnology Research and Innovation Council-National Centre for Cell Science (BRIC-NCCS), SPPU campus, Ganeshkhind, Pune, MH-411007, India.
| |
Collapse
|
2
|
Lawson CM, Jones C, Herman M, Kim C, Mannino E, Omer E, Venegas C. Does Ileus Represent the Forgotten End Organ Failure in Critical Illness? Curr Gastroenterol Rep 2024; 26:166-171. [PMID: 38558135 DOI: 10.1007/s11894-023-00910-8] [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] [Accepted: 12/12/2023] [Indexed: 04/04/2024]
Abstract
PURPOSE OF REVIEW This review evaluates the current literature on ileus, impaired gastrointestinal transit (IGT), and acute gastrointestinal injury (AGI) and its impact on multiple organ dysfunction syndrome. RECENT FINDINGS Ileus is often under recognized in critically ill patients and is associated with significant morbidity and is potentially a marker of disease severity as seen in other organs like kidneys (ATN).
Collapse
Affiliation(s)
- Christy M Lawson
- Department of Trauma Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA.
| | - Chris Jones
- Department of Internal Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Michael Herman
- Department of Gastroenterology, Borland Groover Clinic, Fleming Island, FL, USA
| | - Cecilia Kim
- Department of Trauma Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Elizabeth Mannino
- Department of Trauma Surgery, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, USA
| | - Endashaw Omer
- Department of Gastroenterology, University of Louisville, Louisville, KY, USA
| | - Carlas Venegas
- Department of Neurology, Mayo Clinc, Jacksonville, FL, USA
| |
Collapse
|
3
|
Dvornikova KA, Platonova ON, Bystrova EY. The Role of TRP Channels in Sepsis and Colitis. Int J Mol Sci 2024; 25:4784. [PMID: 38731999 PMCID: PMC11084600 DOI: 10.3390/ijms25094784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/20/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
To date, several members of the transient receptor potential (TRP) channels which provide a wide array of roles have been found in the gastrointestinal tract (GI). The goal of earlier research was to comprehend the intricate signaling cascades that contribute to TRP channel activation as well as how these receptors' activity affects other systems. Moreover, there is a large volume of published studies describing the role of TRP channels in a number of pathological disorders, including inflammatory bowel disease (IBD) and sepsis. Nevertheless, the generalizability of these results is subject to certain limitations. For instance, the study of IBD relies on various animal models and experimental methods, which are unable to precisely imitate the multifactorial chronic disease. The diverse pathophysiological mechanisms and unique susceptibility of animals may account for the inconsistency of the experimental data collected. The main purpose of this study was to conduct a comprehensive review and analysis of existing studies on transient receptor potential (TRP) channels implicating specific models of colitis and sepsis, with particular emphasis on their involvement in pathological disorders such as IBD and sepsis. Furthermore, the text endeavors to evaluate the generalizability of experimental findings, taking into consideration the limitations posed by animal models and experimental methodologies. Finally, we also provide an updated schematic of the most important and possible molecular signaling pathways associated with TRP channels in IBD and sepsis.
Collapse
Affiliation(s)
| | | | - Elena Y. Bystrova
- I.P. Pavlov Institute of Physiology RAS, 199034 St. Petersburg, Russia; (K.A.D.); (O.N.P.)
| |
Collapse
|
4
|
Liang Q, Wang JW, Bai YR, Li RL, Wu CJ, Peng W. Targeting TRPV1 and TRPA1: A feasible strategy for natural herbal medicines to combat postoperative ileus. Pharmacol Res 2023; 196:106923. [PMID: 37709183 DOI: 10.1016/j.phrs.2023.106923] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Abstract
Under physiological or pathological conditions, transient receptor potential (TRP) channel vanilloid type 1 (TRPV1) and TRP ankyrin 1 (TRPA1) possess the ability to detect a vast array of stimuli and execute diverse functions. Interestingly, increasing works have reported that activation of TRPV1 and TRPA1 could also be beneficial for ameliorating postoperative ileus (POI). Increasing research has revealed that the gastrointestinal (GI) tract is rich in TRPV1/TRPA1, which can be stimulated by capsaicin, allicin and other compounds. This activation stimulates a variety of neurotransmitters, leading to increased intestinal motility and providing protective effects against GI injury. POI is the most common emergent complication following abdominal and pelvic surgery, and is characterized by postoperative bowel dysfunction, pain, and inflammatory responses. It is noteworthy that natural herbs are gradually gaining recognition as a potential therapeutic option for POI due to the lack of effective pharmacological interventions. Therefore, the focus of this paper is on the TRPV1/TRPA1 channel, and an analysis and summary of the processes and mechanism by which natural herbs activate TRPV1/TRPA1 to enhance GI motility and relieve pain are provided, which will lay the foundation for the development of natural herb treatments for this disease.
Collapse
Affiliation(s)
- Qi Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Jing-Wen Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Yu-Ru Bai
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Ruo-Lan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China
| | - Chun-Jie Wu
- Institute of Innovation, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| | - Wei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, PR China.
| |
Collapse
|
5
|
Mahran YF, Al-Kharashi LA, Atawia RT, Alanazi RT, Dhahi AMB, Alsubaie R, Badr AM. Radioprotective Effects of Carvacrol and/or Thymol against Gamma Irradiation-Induced Acute Nephropathy: In Silico and In Vivo Evidence of the Involvement of Insulin-like Growth Factor-1 (IGF-1) and Calcitonin Gene-Related Peptide. Biomedicines 2023; 11:2521. [PMID: 37760962 PMCID: PMC10526293 DOI: 10.3390/biomedicines11092521] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/27/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Radiotherapy (RT) is an effective curative cancer treatment. However, RT can seriously damage kidney tissues resulting in radiotherapy nephropathy (RN) where oxidative stress, inflammation, and apoptosis are among the common pathomechanisms. Carvacrol and thymol are known for their antioxidative, anti-inflammatory, and radioprotective activities. Therefore, this study investigated the nephroprotective potentials of carvacrol and/or thymol against gamma (γ) irradiation-induced nephrotoxicity in rats along with the nephroprotection mechanisms, particularly the involvement of insulin-like growth factor-1 (IGF-1) and calcitonin gene-related peptide (CGRP). Methods: Male rats were injected with carvacrol and/or thymol (80 and 50 mg/kg BW in the vehicle, respectively) for five days and exposed to a single dose of irradiation (6 Gy). Then, nephrotoxicity indices, oxidative stress, inflammatory, apoptotic biomarkers, and the histopathological examination were assessed. Also, IGF-1 and CGRP renal expressions were measured. Results: Carvacrol and/or thymol protected kidneys against γ-irradiation-induced acute RN which might be attributed to their antioxidative, anti-inflammatory, and antiapoptotic activities. Moreover, both reserved the γ -irradiation-induced downregulation of CGRP- TNF-α loop in acute RN that might be involved in the pathomechanisms of acute RN. Additionally, in Silico molecular docking simulation of carvacrol and thymol demonstrated promising fitting and binding with CGRP, IGF-1, TNF-α and NF-κB through the formation of hydrogen, hydrophobic and alkyl bonds with binding sites of target proteins which supports the reno-protective properties of carvacrol and thymol. Collectively, our findings open a new avenue for using carvacrol and/or thymol to improve the therapeutic index of γ-irradiation.
Collapse
Affiliation(s)
- Yasmen F. Mahran
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; (R.T.A.); (A.M.B.)
| | - Layla A. Al-Kharashi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11211, Saudi Arabia;
| | - Reem T. Atawia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; (R.T.A.); (A.M.B.)
- Department of Pharmaceutical Sciences, College of Pharmacy, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Rawan Turki Alanazi
- Student, Pharmacy College, King Saud University, Riyadh 11211, Saudi Arabia; (R.T.A.); (A.M.B.D.); (R.A.)
| | - Amal M. Bin Dhahi
- Student, Pharmacy College, King Saud University, Riyadh 11211, Saudi Arabia; (R.T.A.); (A.M.B.D.); (R.A.)
| | - Rawd Alsubaie
- Student, Pharmacy College, King Saud University, Riyadh 11211, Saudi Arabia; (R.T.A.); (A.M.B.D.); (R.A.)
| | - Amira M. Badr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt; (R.T.A.); (A.M.B.)
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11211, Saudi Arabia;
| |
Collapse
|
6
|
Xu S, Liang S, Pei Y, Wang R, Zhang Y, Xu Y, Huang B, Li H, Li J, Tan B, Cao H, Guo S. TRPV1 Dysfunction Impairs Gastric Nitrergic Neuromuscular Relaxation in High-Fat Diet-Induced Diabetic Gastroparesis Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:548-557. [PMID: 36740184 DOI: 10.1016/j.ajpath.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/30/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023]
Abstract
Diabetic gastroparesis (DGP) is characterized by delayed gastric emptying of solid food. Nitrergic neuron-mediated fundus relaxation and intragastric peristalsis are pivotal for gastric emptying and are impaired in DGP. Transient receptor potential vanilloid 1 (TRPV1) ion channels are expressed in gastrointestinal vagal afferent nerves and have a potential role in relevant gastrointestinal disorders. In this study, mice with high-fat diet (HFD)-induced type 2 diabetes mellitus (T2DM), associated with gastroparesis, were used to determine the role of TRPV1 in DGP. After feeding with HFD, mice exhibited obesity, hyperglycemia, insulin resistance, and delayed gastric emptying. Cholinergic- and nitrergic neuron-mediated neuromuscular contractions and relaxation were impaired. The antral tone of the DGP mice was attenuated. Interestingly, activating or suppressing TRPV1 facilitated or inhibited gastric fundus relaxation in normal mice. These effects were neutralized by using a nitric oxide synthase (NOS) inhibitor. Activation or suppression of TRPV1 also increased or reduced NO release. TRPV1 was specifically localized with neuronal NOS in the gastric fundus. These data suggest that TRPV1 activation facilitates gastric fundus relaxation by regulating neuronal NOS and promoting NO release. However, these effects and mechanisms disappeared in mice with DGP induced by HFD diet. TRPV1 expression was only marginally decreased in the fundus of DGP mice. TRPV1 dysfunction may be a potential mechanism underlying the dysfunction of DGP gastric nitrergic neuromuscular relaxation.
Collapse
Affiliation(s)
- Siyuan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China; Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Shaochan Liang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Pei
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Rui Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yao Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yifei Xu
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bin Huang
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Haiwen Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Juanjuan Li
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Bo Tan
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongying Cao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Shaoju Guo
- Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China.
| |
Collapse
|
7
|
Russo AF, Hay DL. CGRP physiology, pharmacology, and therapeutic targets: migraine and beyond. Physiol Rev 2023; 103:1565-1644. [PMID: 36454715 PMCID: PMC9988538 DOI: 10.1152/physrev.00059.2021] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 11/23/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
Calcitonin gene-related peptide (CGRP) is a neuropeptide with diverse physiological functions. Its two isoforms (α and β) are widely expressed throughout the body in sensory neurons as well as in other cell types, such as motor neurons and neuroendocrine cells. CGRP acts via at least two G protein-coupled receptors that form unusual complexes with receptor activity-modifying proteins. These are the CGRP receptor and the AMY1 receptor; in rodents, additional receptors come into play. Although CGRP is known to produce many effects, the precise molecular identity of the receptor(s) that mediates CGRP effects is seldom clear. Despite the many enigmas still in CGRP biology, therapeutics that target the CGRP axis to treat or prevent migraine are a bench-to-bedside success story. This review provides a contextual background on the regulation and sites of CGRP expression and CGRP receptor pharmacology. The physiological actions of CGRP in the nervous system are discussed, along with updates on CGRP actions in the cardiovascular, pulmonary, gastrointestinal, immune, hematopoietic, and reproductive systems and metabolic effects of CGRP in muscle and adipose tissues. We cover how CGRP in these systems is associated with disease states, most notably migraine. In this context, we discuss how CGRP actions in both the peripheral and central nervous systems provide a basis for therapeutic targeting of CGRP in migraine. Finally, we highlight potentially fertile ground for the development of additional therapeutics and combinatorial strategies that could be designed to modulate CGRP signaling for migraine and other diseases.
Collapse
Affiliation(s)
- Andrew F Russo
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa
- Department of Neurology, University of Iowa, Iowa City, Iowa
- Center for the Prevention and Treatment of Visual Loss, Department of Veterans Affairs Health Center, Iowa City, Iowa
| | - Debbie L Hay
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, School of Biological Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
8
|
Brodersen K, Mose M, Ramer Mikkelsen U, Jørgensen JOL, Festersen Nielsen M, Møller N, Wegeberg A, Brock C, Hartmann B, Holst JJ, Rittig N. Prolonged lipopolysaccharide-induced illness elevates glucagon-like peptide-1 and suppresses peptide YY: A human-randomized cross-over trial. Physiol Rep 2022; 10:e15462. [PMID: 36117310 PMCID: PMC9483438 DOI: 10.14814/phy2.15462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/15/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023] Open
Abstract
Severe systemic inflammation is associated with nausea, loss of appetite, and delayed gastric emptying, which increases hospitalization admission length and mortality rate. There is a lack of human controlled studies exploring gastric emptying rates and underlying mechanisms during inflammatory conditions. We aimed to investigate if systemic inflammation in young men delays gastro-intestinal transit times, lowers motility, and affects gastrointestinal hormone secretion. This substudy of a randomized crossover trial investigated eight healthy young men on two separate occasions; (I) following an overnight fast (healthy conditions/HC) and (II) fasting and bedrest combined with two lipopolysaccharide (LPS) injections of 1 ng kg-1 following an overnight fast and 0.5 ng kg-1 following another 24 h (systemic inflammation/SI). A standardized protein beverage and a SmartPill capsule (a wireless gastrointestinal monitoring system) were swallowed during each occasion. Whole gut transit time was comparable between HC and SI. SI decreased gastric mean pressure peak amplitude (p = 0.04) and increased pH rise across the pylorus and small bowel pH (p = 0.02) compared with HC. Glucagon-like peptide-1 was elevated during SI compared with HC (p = 0.04). Peptide YY was lower during SI compared with HC (p = 0.007). Prolonged LPS exposure combined with fasting and bedrest elevated glucagon-like peptide 1 concentrations, which may play a role for the nausea and loss of appetite typically associated with SI.
Collapse
Affiliation(s)
- Katrine Brodersen
- Department of SurgeryViborg Regional HospitalViborgDenmark
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
- Steno Diabetes Center AarhusAarhus University HospitalAarhusDenmark
| | - Maike Mose
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
| | | | - Jens Otto Lunde Jørgensen
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
- Department of Endocrinology and Internal MedicineAarhus University HospitalAarhusDenmark
| | | | - Niels Møller
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
| | - Anne‐Marie Wegeberg
- Mech‐Sense, Department of Gastroenterology and HepatologyAalborg University HospitalAalborgDenmark
| | - Christina Brock
- Mech‐Sense, Department of Gastroenterology and HepatologyAalborg University HospitalAalborgDenmark
- Steno Diabetes Center North DenmarkAalborg University HospitalAalborgDenmark
| | - Bolette Hartmann
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenKøbenhavnDenmark
| | - Jens Juul Holst
- Department of Biomedical Sciences and Novo Nordisk Foundation Center for Basic Metabolic ResearchUniversity of CopenhagenKøbenhavnDenmark
| | - Nikolaj Rittig
- Medical/Steno Aarhus Research LaboratoryAarhus University Hospital, Aarhus UniversityAarhusDenmark
- Steno Diabetes Center AarhusAarhus University HospitalAarhusDenmark
| |
Collapse
|
9
|
Role of Ion Channels in the Chemotransduction and Mechanotransduction in Digestive Function and Feeding Behavior. Int J Mol Sci 2022; 23:ijms23169358. [PMID: 36012643 PMCID: PMC9409042 DOI: 10.3390/ijms23169358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022] Open
Abstract
The gastrointestinal tract constantly communicates with the environment, receiving and processing a wide range of information. The contents of the gastrointestinal tract and the gastrointestinal tract generate mechanical and chemical signals, which are essential for regulating digestive function and feeding behavior. There are many receptors here that sense intestinal contents, including nutrients, microbes, hormones, and small molecule compounds. In signal transduction, ion channels are indispensable as an essential component that can generate intracellular ionic changes or electrical signals. Ion channels generate electrical activity in numerous neurons and, more importantly, alter the action of non-neurons simply and effectively, and also affect satiety, molecular secretion, intestinal secretion, and motility through mechanisms of peripheral sensation, signaling, and altered cellular function. In this review, we focus on the identity of ion channels in chemosensing and mechanosensing in the gastrointestinal tract.
Collapse
|
10
|
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
|
11
|
Montalbetti N, Dalghi MG, Bastacky SI, Clayton DR, Ruiz WG, Apodaca G, Carattino MD. Bladder infection with uropathogenic Escherichia coli increases the excitability of afferent neurons. Am J Physiol Renal Physiol 2022; 322:F1-F13. [PMID: 34779263 PMCID: PMC8698541 DOI: 10.1152/ajprenal.00167.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 11/05/2021] [Indexed: 01/03/2023] Open
Abstract
Urinary tract infections (UTIs) cause bladder hyperactivity and pelvic pain, but the underlying causes of these symptoms remain unknown. We investigated whether afferent sensitization contributes to the bladder overactivity and pain observed in mice suffering from experimentally induced bacterial cystitis. Inoculation of mouse bladders with the uropathogenic Escherichia coli strain UTI89 caused pelvic allodynia, increased voiding frequency, and prompted an acute inflammatory process marked by leukocytic infiltration and edema of the mucosa. Compared with controls, isolated bladder sensory neurons from UTI-treated mice exhibited a depolarized resting membrane potential, lower action potential threshold and rheobase, and increased firing in response to suprathreshold stimulation. To determine whether bacterial virulence factors can contribute to the sensitization of bladder afferents, neurons isolated from naïve mice were incubated with supernatants collected from bacterial cultures with or depleted of lipopolysaccharide (LPS). Supernatants containing LPS prompted the sensitization of bladder sensory neurons with both tetrodotoxin (TTX)-resistant and TTX-sensitive action potentials. However, bladder sensory neurons with TTX-sensitive action potentials were not affected by bacterial supernatants depleted of LPS. Unexpectedly, ultrapure LPS increased the excitability only of bladder sensory neurons with TTX-resistant action potentials, but the supplementation of supernatants depleted of LPS with ultrapure LPS resulted in the sensitization of both population of bladder sensory neurons. In summary, the results of our study indicate that multiple virulence factors released from UTI89 act on bladder sensory neurons to prompt their sensitization. These sensitized bladder sensory neurons mediate, at least in part, the bladder hyperactivity and pelvic pain seen in mice inoculated with UTI89.NEW & NOTEWORTHY Urinary tract infection (UTI) produced by uropathogenic Escherichia coli (UPEC) promotes sensitization of bladder afferent sensory neurons with tetrodotoxin-resistant and tetrodotoxin-sensitive action potentials. Lipopolysaccharide and other virulence factors produced by UPEC contribute to the sensitization of bladder afferents in UTI. In conclusion, sensitized afferents contribute to the voiding symptoms and pelvic pain present in mice bladder inoculated with UPEC.
Collapse
Affiliation(s)
- Nicolas Montalbetti
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marianela G Dalghi
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sheldon I Bastacky
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Dennis R Clayton
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wily G Ruiz
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gerard Apodaca
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Marcelo D Carattino
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
12
|
Sensory nerves promote corneal inflammation resolution via CGRP mediated transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway. Int Immunopharmacol 2021; 102:108426. [PMID: 34906854 DOI: 10.1016/j.intimp.2021.108426] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To explore the role of the corneal sensory nerves in Pseudomonas aeruginosa (P. aeruginosa) keratitis, the synergistic effect between the sensory neurons and macrophages in calcitonin gene-related peptide (CGRP) release, and the functional mechanisms of CGRP-mediated transformation of macrophages to the M2 phenotype. METHODS Corneal nerve loss, macrophage recruitment, and CGRP expression were evaluated. To explore the synergistic effect between the sensory neurons and macrophages, RAW 264.7 cells were challenged with lipopolysaccharide (LPS), then trigeminal ganglion (TG) sensory neurons were isolated and co-incubated with macrophages, and CGRP expression was tested. To investigate the biological function of cornea neuron-initiated immune responses mediated by CGRP, BIBN 4096BS was used to inhibit CGRP in vivo and α-CGRP was used to simulate CGRP in vitro. The expressions of inflammatory cytokines (IL-1β, IL-6, TNF-α, and IL-10), M1 (CD80/CD86), M2 (CD163/CD206) macrophage markers, and signal transducers (PI3K/AKT) were detected. RESULTS P. aeruginosa infection induced corneal nerve loss, macrophage recruitment, and CGRP up-expression. CGRP was co-localized with macrophages. Co-culture showed that sensory neurons and macrophages can mediate CGRP release. More CGRP was released when the two types of cells were combined to respond to LPS. BIBN 4096BS promoted pro-inflammatory cytokines and inhibited the anti-inflammatory cytokines and signal transducers, while, α-CGRP inhibited the pro-inflammatory cytokines and M1 markers and promoted the anti-inflammatory cytokine, M2 markers, and signal transducers. CONCLUSIONS P. aeruginosa infection induces corneal sensory neuron activation, macrophage recruitment, and CGRP up-expression. The synergistic effect between the sensory neurons and macrophages promotes CGRP release. CGRP inhibits corneal inflammation and promotes the transformation of macrophages to the M2 phenotype through the PI3K/AKT signaling pathway.
Collapse
|
13
|
Li Y, Wang Y, Chang H, Cheng B, Miao J, Li S, Hu H, Huang L, Wang Q. Inhibitory Effects of Dexmedetomidine and Propofol on Gastrointestinal Tract Motility Involving Impaired Enteric Glia Ca 2+ Response in Mice. Neurochem Res 2021; 46:1410-1422. [PMID: 33656693 DOI: 10.1007/s11064-021-03280-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 02/20/2021] [Accepted: 02/20/2021] [Indexed: 12/31/2022]
Abstract
Propofol and dexmedetomidine are popular used for sedation in ICU, however, inadequate attention has been paid to their effect on gastrointestinal tract (GIT) motility. Present study aimed to compare the effect of propofol and dexmedetomidine on GIT motility at parallel level of sedation and explore the possible mechanism. Male C57BL/6 mice (8-10 weeks) were randomly divided into control, propofol and dexmedetomidine group. After intraperitoneal injection of propofol or dexmedetomidine, comparable sedative level was confirmed by sedative score, physiological parameters and electroencephalogram (EEG). Different segments of GIT motility in vivo (gastric emptying, small intestine transit, distal colon bead expulsion, stool weight and number of fecal pellets, gastrointestinal transit and whole gut transit time) and colonic migrating motor complexes (CMMCs) pattern in vitro were evaluated. The Ca2+ response of primary enteric glia was examined under the treatment of propofol or dexmedetomidine. There is little difference in physiological parameters and composite permutation entropy index (CPEI) between administration of 50 mg/kg propofol and 40 μg/kg dexmedetomidine, indicated that parallel level of sedation was reached. Data showed that propofol and dexmedetomidine had significantly inhibitory effect on GIT motility while dexmedetomidine was stronger. Also, the amplitude (ΔF/F0) of Ca2+ response in primary enteric glia was attenuated after treated with the sedatives while the effect of dexmedetomidine was greater than propofol. These findings demonstrated that dexmedetomidine caused stronger inhibitory effects on GIT motility in sedative mice, which may involve impaired Ca2+ response in enteric glia. Hence, dexmedetomidine should be carefully applied especially for potential GIT dysmotility patient.
Collapse
Affiliation(s)
- Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yubo Wang
- School of Life Science and Technology, Xidian University, Xi'an, 710061, Shaanxi, China
| | - Haiqing Chang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiwen Miao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shuang Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Liyu Huang
- School of Life Science and Technology, Xidian University, Xi'an, 710061, Shaanxi, China
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
14
|
Chang H, Li S, Li Y, Hu H, Cheng B, Miao J, Gao H, Ma H, Gao Y, Wang Q. Effect of sedation with dexmedetomidine or propofol on gastrointestinal motility in lipopolysaccharide-induced endotoxemic mice. BMC Anesthesiol 2020; 20:227. [PMID: 32894042 PMCID: PMC7487735 DOI: 10.1186/s12871-020-01146-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/02/2020] [Indexed: 12/18/2022] Open
Abstract
Background Sepsis often accompanies gastrointestinal motility disorder that contributes to the development of sepsis in turn. Propofol and dexmedetomidine, as widely used sedatives in patients with sepsis, are likely to depress gastrointestinal peristalsis. We queried whether propofol or dexmedetomidine, at sedative doses, aggravated sepsis-induced ileus. Methods Sedative/Anesthetic Scores and vital signs of lipopolysaccharide (LPS)-induced endotoxemic mice were measured during sedation with propofol or dexmedetomidine. Endotoxemic mice were divided into 10% fat emulsion, propofol, saline, and dexmedetomidine group. The gastric emptying, small intestinal transit, tests of colonic motility, gastrointestinal transit and whole gut transit were evaluated at 15 mins and 24 h after intraperitoneal injection of sedatives/vehicles respectively. Results 40 mg·kg− 1propofol and 80 μg·kg− 1 dexmedetomidine induced a similar depth of sedation with comparable vital signs except that dexmedetomidine strikingly decreased heart rate in endotoxemic mice. Dexmedetomidine markedly inhibited gastric emptying (P = 0.006), small intestinal transit (P = 0.006), colonic transit (P = 0.0006), gastrointestinal transit (P = 0.0001) and the whole gut transit (P = 0.034) compared with the vehicle, whereas propofol showed no depression on all parts of gastrointestinal motility 15 mins after administration. The inhibitive effects of dexmedetomidine in these tests vanished 24 h after the administration. Conclusions Deep sedation with dexmedetomidine, but not propofol, significantly inhibited gastrointestinal peristalsis in endotoxemic mice while the inhibitory effect disappeared 24 h after sedation. These data suggested that both propofol and dexmedetomidine could be applied in septic patients while dexmedetomidine should be used cautiously in patients with cardiac disease or ileus.
Collapse
Affiliation(s)
- Haiqing Chang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Shuang Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yansong Li
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hao Hu
- Department of Pharmacology, School of Basic Medical Sciences, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Bo Cheng
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jiwen Miao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hui Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Hongli Ma
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yanfeng Gao
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| | - Qiang Wang
- Department of Anesthesiology & Center for Brain Science, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China.
| |
Collapse
|
15
|
Chen Y, Mu J, Zhu M, Mukherjee A, Zhang H. Transient Receptor Potential Channels and Inflammatory Bowel Disease. Front Immunol 2020; 11:180. [PMID: 32153564 PMCID: PMC7044176 DOI: 10.3389/fimmu.2020.00180] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/23/2020] [Indexed: 02/05/2023] Open
Abstract
The transient receptor potential (TRP) cation channels are present in abundance across the gastrointestinal (GI) tract, serving as detectors for a variety of stimuli and secondary transducers for G-protein coupled receptors. The activation of TRP channels triggers neurogenic inflammation with related neuropeptides and initiates immune reactions by extra-neuronally regulating immune cells, contributing to the GI homeostasis. However, under pathological conditions, such as inflammatory bowel disease (IBD), TRP channels are involved in intestinal inflammation. An increasing number of human and animal studies have indicated that TRP channels are correlated to the visceral hypersensitivity (VHS) and immune pathogenesis in IBD, leading to an exacerbation or amelioration of the VHS or intestinal inflammation. Thus, TRP channels are a promising target for novel therapeutic methods for IBD. In this review, we comprehensively summarize the functions of TRP channels, especially their potential roles in immunity and IBD. Additionally, we discuss the contradictory findings of prior studies and offer new insights with regard to future research.
Collapse
Affiliation(s)
- Yiding Chen
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Jingxi Mu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Min Zhu
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| | | | - Hu Zhang
- Department of Gastroenterology, West China Hospital, Sichuan University, Chengdu, China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Nullens S, De Man J, Bridts C, Ebo D, Francque S, De Winter B. Identifying Therapeutic Targets for Sepsis Research: A Characterization Study of the Inflammatory Players in the Cecal Ligation and Puncture Model. Mediators Inflamm 2018; 2018:5130463. [PMID: 30174555 PMCID: PMC6098915 DOI: 10.1155/2018/5130463] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/23/2018] [Accepted: 05/29/2018] [Indexed: 01/04/2023] Open
Abstract
During sepsis, disturbed gastrointestinal motility and increased mucosal permeability can aggravate sepsis due to the increased risk of bacterial translocation. To help identify new therapeutic targets, there is a need for animal models that mimic the immunological changes in the gastrointestinal tract as observed during human sepsis. We therefore characterized in detail the gastrointestinal neuroimmune environment in the cecal ligation and puncture (CLP) model, which is the gold standard animal model of microbial sepsis. Mice were sacrificed at day 2 and day 7, during which gastrointestinal motility was assessed and cytokines were measured in the serum and the colon. In the spleen, lymph nodes, ileum, and colon, subsets of leukocyte populations were identified by flow cytometry. Septic animals displayed an impaired gastrointestinal motility at day 2 and day 7. Two days post-CLP, increased serum and colonic levels of proinflammatory cytokines were measured. Flow cytometry revealed an influx of neutrophils in the colon and ileum, increased numbers of macrophages in the spleen and mesenteric lymph nodes, and an enhanced number of mast cells in all tissues. At day 7 post-CLP, lymphocyte depletion was observed in all tissues coinciding with increased IL-10 and TGF-β levels, as well as increased colonic levels of IL-17A and IFN-γ. Thus, CLP-induced sepsis in mice results in simultaneous activation of pro- and anti-inflammatory players at day 2 and day 7 in different tissues, mimicking human sepsis.
Collapse
Affiliation(s)
- Sara Nullens
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Joris De Man
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Chris Bridts
- Immunology-Allergology-Rheumatology Department, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Didier Ebo
- Immunology-Allergology-Rheumatology Department, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Sven Francque
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Edegem, Belgium
| | - Benedicte De Winter
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
17
|
Zanos TP, Silverman HA, Levy T, Tsaava T, Battinelli E, Lorraine PW, Ashe JM, Chavan SS, Tracey KJ, Bouton CE. Identification of cytokine-specific sensory neural signals by decoding murine vagus nerve activity. Proc Natl Acad Sci U S A 2018; 115:E4843-E4852. [PMID: 29735654 PMCID: PMC6003492 DOI: 10.1073/pnas.1719083115] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The nervous system maintains physiological homeostasis through reflex pathways that modulate organ function. This process begins when changes in the internal milieu (e.g., blood pressure, temperature, or pH) activate visceral sensory neurons that transmit action potentials along the vagus nerve to the brainstem. IL-1β and TNF, inflammatory cytokines produced by immune cells during infection and injury, and other inflammatory mediators have been implicated in activating sensory action potentials in the vagus nerve. However, it remains unclear whether neural responses encode cytokine-specific information. Here we develop methods to isolate and decode specific neural signals to discriminate between two different cytokines. Nerve impulses recorded from the vagus nerve of mice exposed to IL-1β and TNF were sorted into groups based on their shape and amplitude, and their respective firing rates were computed. This revealed sensory neural groups responding specifically to TNF and IL-1β in a dose-dependent manner. These cytokine-mediated responses were subsequently decoded using a Naive Bayes algorithm that discriminated between no exposure and exposures to IL-1β and TNF (mean successful identification rate 82.9 ± 17.8%, chance level 33%). Recordings obtained in IL-1 receptor-KO mice were devoid of IL-1β-related signals but retained their responses to TNF. Genetic ablation of TRPV1 neurons attenuated the vagus neural signals mediated by IL-1β, and distal lidocaine nerve block attenuated all vagus neural signals recorded. The results obtained in this study using the methodological framework suggest that cytokine-specific information is present in sensory neural signals within the vagus nerve.
Collapse
Affiliation(s)
- Theodoros P Zanos
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Harold A Silverman
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030
- Center for Biomedical Sciences, Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Todd Levy
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Tea Tsaava
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030
- Center for Biomedical Sciences, Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Emily Battinelli
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030
- Center for Biomedical Sciences, Feinstein Institute for Medical Research, Manhasset, NY 11030
| | | | - Jeffrey M Ashe
- General Electric Global Research US, Niskayuna, NY 12309
| | - Sangeeta S Chavan
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030
- Center for Biomedical Sciences, Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Kevin J Tracey
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030;
- Center for Biomedical Sciences, Feinstein Institute for Medical Research, Manhasset, NY 11030
| | - Chad E Bouton
- Center for Bioelectronic Medicine, Feinstein Institute for Medical Research, Manhasset, NY 11030;
| |
Collapse
|
18
|
Chen J, Hamers AJP, Finsterbusch M, Massimo G, Zafar M, Corder R, Colas RA, Dalli J, Thiemermann C, Ahluwalia A. Endogenously generated arachidonate-derived ligands for TRPV1 induce cardiac protection in sepsis. FASEB J 2018; 32:3816-3831. [PMID: 29465314 DOI: 10.1096/fj.201701303r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The severity of cardiac dysfunction predicts mortality in sepsis. Activation of transient receptor potential vanilloid receptor type (TRPV)-1, a predominantly neuronal nonselective cation channel, has been shown to improve outcome in sepsis and endotoxemia. However, the role of TRPV1 and the identity of its endogenous ligands in the cardiac dysfunction caused by sepsis and endotoxemia are unknown. Using TRPV1-/- and TRPV1+/+ mice, we showed that endogenous activation of cardiac TRPV1 during sepsis is key to limiting the ensuing cardiac dysfunction. Use of liquid chromatography-tandem mass spectrometry lipid analysis and selective inhibitors of arachidonic metabolism suggest that the arachidonate-derived TRPV1 activator, 20-hydroxyeicosateraenoic acid (20-HETE), underlies a substantial component of TRPV1-mediated cardioprotection in sepsis. Moreover, using selective antagonists for neuropeptide receptors, we show that this effect of TRPV1 relates to the activity of neuronally released cardiac calcitonin gene-related peptide (CGRP) and that, accordingly, administration of CGRP can rescue cardiac dysfunction in severe endotoxemia. In sum activation of TRPV1 by 20-HETE leads to the release of CGRP, which protects the heart against the cardiac dysfunction in endotoxemia and identifies both TRPV1 and CGRP receptors as potential therapeutic targets in endotoxemia.-Chen, J., Hamers, A. J. P., Finsterbusch, M., Massimo, G., Zafar, M., Corder, R., Colas, R. A., Dalli, J., Thiemermann, C., Ahluwalia, A. Endogenously generated arachidonate-derived ligands for TRPV1 induce cardiac protection in sepsis.
Collapse
Affiliation(s)
- Jianmin Chen
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Alexander J P Hamers
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Michaela Finsterbusch
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Gianmichele Massimo
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Maleeha Zafar
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Roger Corder
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Romain A Colas
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Amrita Ahluwalia
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
19
|
Calzetta L, Rogliani P, Pistocchini E, Mattei M, Cito G, Alfonsi P, Page C, Matera MG. Effect of lipopolysaccharide on the responsiveness of equine bronchial tissue. Pulm Pharmacol Ther 2018; 49:88-94. [PMID: 29408044 DOI: 10.1016/j.pupt.2018.01.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/09/2023]
Abstract
Recurrent airway obstruction (RAO) is a main characteristic of horses with severe equine asthma syndrome. The presence of bacterial lipopolysaccharide (LPS) in the airways of horses is thought to play a crucial role in the clinical expression of this disorder. This study pharmacologically characterized the effect of LPS on the responsiveness of equine bronchial tissue. Equine isolated bronchi were incubated overnight with LPS (0.1-100 ng/ml) and then stimulated by electrical field stimulation (EFS). The role of capsaicin sensitive-sensory nerves (capsaicin desensitization treatment), neurokinin-2 (NK2) receptors (blocked by GR159897), transient receptor potential vanilloid type 1 receptors (TRPV1; blocked by SB366791), and neurokinin A (NKA) were investigated. Untreated bronchi were used as control tissues. LPS (1 ng/ml) significantly increased the EFS-evoked contractility of equine bronchi compared with control tissues (+742 ± 123 mg; P < 0.001). At higher concentrations LPS induced desensitization to airways hyperresponsiveness (AHR; EC50: 5.9 ± 2.6 ng/ml). Capsaicin desensitization and GR159897 significantly prevented AHR induced by LPS at EFS1-50Hz (-197 ± 25%; P < 0.01). SB366791 inhibited AHR at very low EFS frequency (EFS1Hz -193 ± 29%; P < 0.01 vs. LPS-treated bronchi). LPS (1 ng/ml) significantly (P < 0.01) increased 3.7 ± 0.7 fold the release of NKA compared with control bronchi. LPS induces biphasic dysfunctional bronchial contractility due to the stimulation of capsaicin sensitive-sensory nerves, increased release of NKA, and activation of NK2 receptors, whereas TRPV1 receptors appear to play a marginal role in this response. The overnight challenge with low concentrations of LPS represents a suitable model to investigate pharmacological options that may be of value in the treatment of equine RAO.
Collapse
Affiliation(s)
- Luigino Calzetta
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy.
| | - Paola Rogliani
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Rome, Italy
| | | | - Maurizio Mattei
- Department of Biology, Centro Servizi Interdipartimentale-STA, University of Rome Tor Vergata, Rome, Italy
| | - Giuseppe Cito
- ASL Roma 2, UOC Tutela Igienico Sanitaria Degli Alimenti di Origine Animale, Rome, Italy
| | - Pietro Alfonsi
- ASL Roma 2, UOC Igiene Degli Allevamenti e Delle Produzioni Zootecniche, Rome, Italy
| | - Clive Page
- The Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - Maria Gabriella Matera
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
20
|
Lu CX, Qiu T, Liu ZF, Su L, Cheng B. Calcitonin gene-related peptide has protective effect on brain injury induced by heat stroke in rats. Exp Ther Med 2017; 14:4935-4941. [PMID: 29201197 PMCID: PMC5704302 DOI: 10.3892/etm.2017.5126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 06/22/2017] [Indexed: 11/13/2022] Open
Abstract
Heat stroke often leads to multiple organ dysfunction syndrome (MODS) with a neurological morbidity of 30%. Current studies suggested that pathophysiological responses to heat stroke may be due to a systemic inflammatory response syndrome and a series of peptidergic nerve reactions. The mechanisms underlying the high neurological morbidity in heat stroke have remained largely elusive. In recent years, calcitonin gene-related peptide (CGRP) has been considered to have a positive role in central nervous system injury. The present study investigated the influence of CGRP on brain injury induced by heat stroke. A rat model of heat stroke was established in a pre-warmed artificial climate chamber with a temperature of 35.5±0.5°C and a relative humidity of 60±5%. The rectal core temperature (Tc) was monitored. Heat stress was halted at a Tc of no more than 41°C A bolus injection of CGRP was administered to each rat in the HS+CGRP group and a bolus injection of CGRP8-37 was administered to each rat in the HS+CGRP8-37 group after heat stress. After 2 h, electroencephalograms were recorded and the pathological morphology of brain tissue as well as brain cell apoptosis and caspase-3 protein levels in the brain were measured. The EEG of rats in the HS+CGRP group was characterized by a short- to long-term α-wave and low-voltage β-waves as well as a large amount of intermittent δ- and θ-waves. Compared with the HS group, the θ-wave decreased and the α-wave increased significantly (P<0.05). Slight pathological damage of nerve cells appeared in the HS+CGRP group. Greater damage was observed in HS+CGRP8-37 group with neural cell shrinkage, volume reduction, nuclear pyknosis, disappearance of part of the nuclear membrane and cell necrosis. In the HS+CGRP group, apoptotic cells and caspase-3 protein in the brain were significantly decreased when compared with those in the HS group (P<0.05), while they were significantly increased in the HS+CGRP8-37 group (P<0.05 vs. HS group). The results of the present study reflected that CGRP has a protective effect on early-stage brain injury induced by heat stroke in rats.
Collapse
Affiliation(s)
- Cheng-Xiang Lu
- Department of Intensive Care Unit, Affiliated General Hospital of Guangzhou Military Command of Southern Medical University, Guangzhou, Guangdong 510010, P.R. China.,Department of Intensive Care Unit, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Ting Qiu
- Department of Neurology, Zhongshan Hospital Xiamen University, Xiamen, Fujian 361004, P.R. China
| | - Zhi-Feng Liu
- Department of Intensive Care Unit, General Hospital of Guangzhou Military Command, Guangzhou, Guangdong 510010, P.R. China
| | - Lei Su
- Department of Intensive Care Unit, Affiliated General Hospital of Guangzhou Military Command of Southern Medical University, Guangzhou, Guangdong 510010, P.R. China
| | - Biao Cheng
- Department of Plastic Surgery, Affiliated General Hospital of Guangzhou Military Command of Southern Medical University, Guangzhou, Guangdong 510010, P.R. China
| |
Collapse
|
21
|
Lian YL, Cheng MJ, Zhang XX, Wang L. Elevated expression of transient receptor potential vanilloid type 1 in dorsal root ganglia of rats with endometriosis. Mol Med Rep 2017; 16:1920-1926. [PMID: 28627595 PMCID: PMC5561994 DOI: 10.3892/mmr.2017.6783] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 04/06/2017] [Indexed: 12/03/2022] Open
Abstract
Pain is the most pronounced complaint of women with endometriosis, however the underlying mechanism is still poorly understood. In the present study, the authors evaluate the effect of transient receptor potential vanilloid type 1 (TRPV1) of dorsal root ganglia (DRG) on endometriosis-associated pain. A total of 36 SD rats were randomly divided into a sham group (n=9) and a Model group (n=27), accepted auto‑transplanted pieces of fat or uterus to the pelvic cavity. At 4 weeks, the Model group was randomly subdivided into the following groups: ENDO group (no treatment, n=9), BCTC group (Model + BCTC, an antagonist of TRPV1, n=9), Vehicle group (Model + cyclodextrin, the vehicle of BCTC, n=9). Tail‑flick test was performed prior to surgery, 1 h prior to and following treatment of BCTC or cyclodextrin. The expression of TRPV1, substance P (SP), calcitonin gene‑related peptide (CGRP) in L1‑L6 DRG was measured via immunohistochemistry, western blotting and RT‑qPCR. The results indicated that the Model group exhibited a significant decrease in tail flick latency compared to pre‑surgical baseline, and the expression of TRPV1, SP, CGRP protein and mRNA in L1‑L6 DRG significantly increased compared to the sham group. BCTC significantly improved tail flick latency, and downregulated the expression of TRPV1, SP and CGRP protein and mRNA levels in L1‑L6 DRG compared to ENDO group. However, there were no significant differences of those in Vehicle group compared with the ENDO group. Taken together, the current study provides evidence that TRPV1 expressed in DRG may serve an important role in endometriosis-associated pain.
Collapse
Affiliation(s)
- Yu-Ling Lian
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, P.R. China
| | - Ming-Jun Cheng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, P.R. China
| | - Xian-Xia Zhang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, P.R. China
| | - Li Wang
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, P.R. China
- Department of Obstetrics and Gynecology of Shanghai Medical College, Fudan University, Shanghai 200032, P.R. China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai 200011, P.R. China
| |
Collapse
|
22
|
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
|
23
|
Farro G, Gomez-Pinilla PJ, Di Giovangiulio M, Stakenborg N, Auteri M, Thijs T, Depoortere I, Matteoli G, Boeckxstaens GE. Smooth muscle and neural dysfunction contribute to different phases of murine postoperative ileus. Neurogastroenterol Motil 2016; 28:934-47. [PMID: 26891411 DOI: 10.1111/nmo.12796] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/15/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND Postoperative ileus (POI) is characterized by a transient inhibition of gastrointestinal (GI) motility after abdominal surgery mediated by the inflammation of the muscularis externa (ME). The aim of this study was to identify alterations in the enteric nervous system that may contribute to the pathogenesis of POI. METHODS Gastrointestinal transit, contractility of isolated smooth muscle strips and inflammatory parameters were evaluated at different time points (1.5 h to 10 days) after intestinal manipulation (IM) in mice. Immune-labeling was used to visualize changes in myenteric neurons. KEY RESULTS Intestinal manipulation resulted in an immediate inhibition of GI transit recovering between 24 h and 5 days. In vitro contractility to K(+) (60 mM) or carbachol (10(-9) to 10(-4) M) was biphasically suppressed over 24 h after IM (with transient recovery at 6 h). The first phase of impaired myogenic contractility was associated with increased expression of TNF-α, IL-6 and IL-1α. After 24 h, we identified a significant reduction in electrical field stimulation-evoked contractions and relaxations, lasting up to 10 days after IM. This was associated with a reduced expression of chat and nos1 genes. CONCLUSIONS & INFERENCES Intestinal manipulation induces two waves of smooth muscle inhibition, most likely mediated by inflammatory cytokines, lasting up to 3 days after IM. Further, we here identify a late third phase (>24 h) characterized by impaired cholinergic and nitrergic neurotransmission persisting after recovery of muscle contractility. These findings illustrate that POI results from inflammation-mediated impaired smooth muscle contraction, but also involves a long-lasting impact of IM on the enteric nervous system.
Collapse
Affiliation(s)
- G Farro
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - P J Gomez-Pinilla
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - M Di Giovangiulio
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - N Stakenborg
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - M Auteri
- Division of Physiology, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| | - T Thijs
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - I Depoortere
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G Matteoli
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - G E Boeckxstaens
- Division of Gastroenterology, Department of Clinical and Experimental Medicine, Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| |
Collapse
|
24
|
Yu X, Yu M, Liu Y, Yu S. TRP channel functions in the gastrointestinal tract. Semin Immunopathol 2015; 38:385-96. [PMID: 26459157 DOI: 10.1007/s00281-015-0528-y] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 09/07/2015] [Indexed: 12/14/2022]
Abstract
Transient receptor potential (TRP) channels are predominantly distributed in both somatic and visceral sensory nervous systems and play a crucial role in sensory transduction. As the largest visceral organ system, the gastrointestinal (GI) tract frequently accommodates external inputs, which stimulate sensory nerves to initiate and coordinate sensory and motor functions in order to digest and absorb nutrients. Meanwhile, the sensory nerves in the GI tract are also able to detect potential tissue damage by responding to noxious irritants. This nocifensive function is mediated through specific ion channels and receptors expressed in a subpopulation of spinal and vagal afferent nerve called nociceptor. In the last 18 years, our understanding of TRP channel expression and function in GI sensory nervous system has been continuously improved. In this review, we focus on the expressions and functions of TRPV1, TRPA1, and TRPM8 in primary extrinsic afferent nerves innervated in the esophagus, stomach, intestine, and colon and briefly discuss their potential roles in relevant GI disorders.
Collapse
Affiliation(s)
- Xiaoyun Yu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Mingran Yu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Yingzhe Liu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Shaoyong Yu
- Division of Gastroenterology & Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Ross Research Building, Room 945, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
25
|
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
|
26
|
Calzetta L, Luongo L, Cazzola M, Page C, Rogliani P, Facciolo F, Maione S, Capuano A, Rinaldi B, Matera MG. Contribution of sensory nerves to LPS-induced hyperresponsiveness of human isolated bronchi. Life Sci 2015; 131:44-50. [PMID: 25914087 DOI: 10.1016/j.lfs.2015.03.023] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/11/2015] [Accepted: 03/29/2015] [Indexed: 10/23/2022]
Abstract
AIMS Bacterial lipopolysaccharide (LPS) can induce bronchial hyperresponsiveness (BHR), but the underlying mechanisms remain to be determined. Here, the possible contribution of sensory nerves to LPS-induced BHR was examined in human isolated bronchi to pharmacologically identify the mechanisms underlying this phenomenon. MAIN METHODS Human isolated bronchial tone was induced by electrical field stimulation (EFS). The responses of airways to LPS, with or without capsaicin desensitization or thiorphan treatment were studied and the transient receptor potential vanilloid type 1 (TRPV1) expression was assessed. We performed similar experiments in the presence of a TRPV1 or a neurokinin (NK) 2 receptor antagonist using SB366791 and GR159897, respectively. KEY FINDINGS LPS increased (≃2.3-fold, P<0.001) the contraction induced by EFS, compared to control tissues. Acute administration of capsaicin enhanced (≃2.3-fold, P<0.001) the EFS-mediated contraction, but did not potentiate the effect of LPS. Thiorphan increased (≃1.3-fold, P<0.05) the contractile response of LPS treated tissues and, at lower frequencies, it enhanced (≃1.7-fold, P<0.001) the capsaicin-induced contraction. In capsaicin-desensitized bronchi, LPS did not modify (P>0.05) the EFS contractile response, nor after treatment with thiorphan. Capsaicin desensitization reduced (≃0.4-fold, P<0.001) the LPS-induced BHR. SB366791 and GR159897 prevented the LPS-induced BHR and the release of NKA. LPS increased (+85.3±9.5%, P<0.01) the surface membrane expression of TRPV1 in parasympathetic ganglia. SIGNIFICANCE Our results demonstrate the involvement of capsaicin-sensitive sensory nerves and neutral endopeptidases in LPS-induced BHR of the human bronchi, associated with an upregulation of TRPV1 and release of NKA.
Collapse
Affiliation(s)
- Luigino Calzetta
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Livio Luongo
- Unit of Pharmacology, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Mario Cazzola
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.
| | - Clive Page
- The Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Paola Rogliani
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | | | - Sabatino Maione
- Unit of Pharmacology, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Annalisa Capuano
- Unit of Pharmacology, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Barbara Rinaldi
- Unit of Pharmacology, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, Second University of Naples, Naples, Italy
| |
Collapse
|
27
|
Toll-like receptor 4 contributes to the inhibitory effect of morphine on colonic motility in vitro and in vivo. Sci Rep 2015; 5:9499. [PMID: 25962524 PMCID: PMC5386182 DOI: 10.1038/srep09499] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 03/10/2015] [Indexed: 12/13/2022] Open
Abstract
Opioids rank among the most potent analgesic drugs but gastrointestinal side effects, especially constipation, limit their therapeutic utility. The adverse effects of opioids have been attributed to stimulation of opioid receptors, but emerging evidence suggests that opioids interact with the innate immune receptor Toll-like receptor 4 (TLR4) and its signalling pathway. As TLR4 signalling affects gastrointestinal motility, we examined the involvement of TLR4 in morphine-induced depression of peristaltic motility in the guinea-pig intestine in vitro and male C57BL/6N mice in vivo. While the TLR4 antagonist TAK-242 (0.1 μM and 1 μM) did not alter the morphine-induced inhibition of peristalsis in the isolated guinea-pig small intestine, the morphine-induced decrease in pellet propulsion velocity in colonic segments was attenuated by TAK-242 (0.1 μM). The ability of TAK-242 (4 mg/kg) to mitigate the morphine-induced suppression of colonic motility was replicated in mice in vivo by measuring the expulsion time of beads inserted in the distal colon. The inhibition of upper gastrointestinal transit of mice by morphine was not affected by pre-treatment with TAK-242 (4 mg/kg) in vivo. This is the first report that morphine-induced inhibition of colonic peristalsis is alleviated by TLR4 antagonism. We therefore conclude that TLR4 may contribute to opioid-induced constipation.
Collapse
|
28
|
Assas BM, Pennock JI, Miyan JA. Calcitonin gene-related peptide is a key neurotransmitter in the neuro-immune axis. Front Neurosci 2014; 8:23. [PMID: 24592205 PMCID: PMC3924554 DOI: 10.3389/fnins.2014.00023] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 01/27/2014] [Indexed: 12/26/2022] Open
Abstract
The question of how the neural and immune systems interact in host defense is important, integrating a system that senses the whole body with one that protects. Understanding the mechanisms and routes of control could produce novel and powerful ways of promoting and enhancing normal functions as well as preventing or treating abnormal functions. Fragmentation of biological research into specialities has resulted in some failures in recognizing and understanding interactions across different systems and this is most striking across immunology, hematology, and neuroscience. This reductionist approach does not allow understanding of the in vivo orchestrated response generated through integration of all systems. However, many factors make the understanding of multisystem cross-talk in response to a threat difficult, for instance the nervous and immune systems share communication molecules and receptors for a wide range of physiological signals. But, it is clear that physical, hard-wired connections exist between the two systems, with the key link involving sensory, unmyelinated nerve fibers (c fibers) containing the neuropeptide calcitonin gene-related peptide (CGRP), and modified macrophages, mast cells and other immune and host defense cells in various locations throughout the body. In this review we will therefore focus on the induction of CGRP and its key role in the neuroimmune axis.
Collapse
Affiliation(s)
- Bakri M Assas
- Translational Medicine, Faculty of Medical and Human Sciences, The University of Manchester Manchester, UK ; Department of Immunology, Faculty of Applied Sciences, King Abdulaziz University Jeddah, Saudi Arabia
| | - Joanne I Pennock
- Translational Medicine, Faculty of Medical and Human Sciences, The University of Manchester Manchester, UK
| | - Jaleel A Miyan
- Neurosciences, Faculty of Life Sciences, The University of Manchester Manchester, UK
| |
Collapse
|
29
|
Buckinx R, Van Nassauw L, Avula LR, Alpaerts K, Adriaensen D, Timmermans JP. Transient receptor potential vanilloid type 1 channel (TRPV1) immunolocalization in the murine enteric nervous system is affected by the targeted C-terminal epitope of the applied antibody. J Histochem Cytochem 2013; 61:421-32. [PMID: 23482327 DOI: 10.1369/0022155413484764] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The expression of transient receptor potential vanilloid type 1 channel (TRPV1) in the enteric nervous system is still the subject of debate. Although a number of studies have reported that TRPV1 is limited to extrinsic afferent fibers, other studies argue for an intrinsic expression of TRPV1. In the present study, reverse transcriptase PCR was employed to establish the expression of TRPV1 mRNA throughout the gastrointestinal tract. Using two antibodies directed against different epitopes of TRPV1, we were able to show at the protein level that the observed distribution pattern of TRPV1 is dependent on the antibody used in the immunohistochemical staining. A first antibody indeed mainly stained neuronal fibers, whereas a second antibody exclusively stained perikarya of enteric neurons throughout the mouse gastrointestinal tract. We argue that these different distribution patterns are due to the antibodies discriminating between different modulated forms of TRPV1 that influence the recognition of the targeted immunogen and as such distinguish intracellular from plasmalemmal forms of TRPV1. Our study is the first to directly compare these two antibodies within the same species and in identical conditions. Our observations underline that detailed knowledge of the epitope that is recognized by the antibodies employed in immunohistochemical procedures is a prerequisite for correctly interpreting experimental results.
Collapse
Affiliation(s)
- Roeland Buckinx
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | | |
Collapse
|
30
|
Kimura Y, Yamauchi M, Inoue H, Kimura S, Yamakage M, Aimono M, Sumita S. Risk factors for gastric distension in patients with acute appendicitis: a retrospective cohort study. J Anesth 2012; 26:574-8. [PMID: 22349834 DOI: 10.1007/s00540-012-1353-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 01/30/2012] [Indexed: 01/07/2023]
Abstract
PURPOSE There has been no report on risk factors for gastric distension (GD) when inducing general anesthesia in an emergency situation. The aim of this study was to clarify the risk factors for GD in patients with acute appendicitis at their hospital visit. METHODS We reviewed medical records of patients from April 2007 to March 2010 who underwent open appendectomy for acute appendicitis and were diagnosed pathologically. GD was defined as a larger anteroposterior diameter and larger lateral diameter of the stomach than those of the left kidney in computed tomography (CT) imaging. The primary outcome was the presence of GD. Candidate variables such as patient characteristics, physical findings, and CT imaging findings associated with GD were assessed. Time after beginning of abdominal pain was categorized and compared. Determinants with significant univariate association (P < 0.20) with the primary outcome were used to construct multivariable logistic regression models. RESULTS We enrolled 121 patients and divided this cohort into a GD group (44 cases, 36%) and a non-GD group (77 cases, 64%). Results of univariate analysis showed longer duration of time after beginning of abdominal pain (P = 0.016), younger age (P < 0.001), and more frequent distended small bowel (P < 0.001) in the GD group than in the non-GD group. In multivariate analysis, age [odds ratio (OR) = 0.939, P = 0.002] and time after beginning of abdominal pain (OR = 1.807, P = 0.031) were shown to be independent risk factors. CONCLUSION Younger appendicitis patients with acute abdominal pain for 1 or more days should be treated as patients with high risk for GD.
Collapse
Affiliation(s)
- Yoshinobu Kimura
- Division of Anesthesia, Asahikawa Redcross Hospital, 1-1-1-1 Akebono, Asahikawa, Hokkaido, 070-8530, Japan.
| | | | | | | | | | | | | |
Collapse
|
31
|
Caddell KA, Martindale R, McClave SA, Miller K. Can the intestinal dysmotility of critical illness be differentiated from postoperative ileus? Curr Gastroenterol Rep 2011; 13:358-367. [PMID: 21626118 DOI: 10.1007/s11894-011-0206-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Gastrointestinal dysmotility is commonly noted in the intensive care unit and postoperative settings. Characterized by delayed passage of stool and flatus, nausea, vomiting, and abdominal distention, the condition is associated with nutritional deficiencies, risk of aspiration, and considerable allocation of health care resources. Knowledge of gastrointestinal function in health and illness continues to expand. While the factors that precipitate ileus differ between postoperative and critically ill patients, the two clinical scenarios seem to have similar mechanisms and share many of the same pathophysiologic patterns. By reviewing and comparing the literature on the respective mechanisms and contributing factors generated in these separate clinical settings, a common more comprehensive management strategy may be derived with the potential for newer innovative therapeutic options.
Collapse
Affiliation(s)
- Kirk A Caddell
- Department of Surgery, Oregon Health and Sciences University, Portland, OR 97239-3098, USA
| | | | | | | |
Collapse
|
32
|
Devesa I, Planells-Cases R, Fernández-Ballester G, González-Ros JM, Ferrer-Montiel A, Fernández-Carvajal A. Role of the transient receptor potential vanilloid 1 in inflammation and sepsis. J Inflamm Res 2011; 4:67-81. [PMID: 22096371 PMCID: PMC3218746 DOI: 10.2147/jir.s12978] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The transient receptor potential vanilloid 1 (TRPV1) is a thermoreceptor that responds to noxious temperatures, as well as to chemical agonists, such as vanilloids and protons. In addition, its channel activity is notably potentiated by proinflammatory mediators released upon tissue damage. The TRPV1 contribution to sensory neuron sensitization by proalgesic agents has signaled this receptor as a prime target for analgesic and anti-inflammatory drug intervention. However, TRPV1 antagonists have notably failed in clinical and preclinical studies because of their unwanted side effects. Recent reports have unveiled previously unrecognized anti-inflammatory and protective functions of TRPV1 in several diseases. For instance, this channel has been suggested to play an anti-inflammatory role in sepsis. Therefore, the use of potent TRPV1 antagonists as a general strategy to treat inflammation must be cautiously considered, given the deleterious effects that may arise from inhibiting the population of channels that have a protective function. The use of TRPV1 antagonists may be limited to treating those pathologies where enhanced receptor activity contributes to the inflamed state. Alternatively, therapeutic paradigms, such as reduction of inflammatory-mediated increase of receptor expression in the cell surface, may be a better strategy to prevent abrogation of the TRPV1 subpopulation involved in anti-inflammatory and protective processes.
Collapse
Affiliation(s)
- Isabel Devesa
- Instituto de Biología Molecular y Celular, Universidad Miguel Hernández, Alicante
| | | | | | | | | | | |
Collapse
|
33
|
De Winter BY, De Man JG. Interplay between inflammation, immune system and neuronal pathways: effect on gastrointestinal motility. World J Gastroenterol 2010; 16:5523-5535. [PMID: 21105185 PMCID: PMC2992670 DOI: 10.3748/wjg.v16.i44.5523] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Revised: 07/30/2010] [Accepted: 08/06/2010] [Indexed: 02/06/2023] Open
Abstract
Sepsis is a systemic inflammatory response representing the leading cause of death in critically ill patients, mostly due to multiple organ failure. The gastrointestinal tract plays a pivotal role in the pathogenesis of sepsis-induced multiple organ failure through intestinal barrier dysfunction, bacterial translocation and ileus. In this review we address the role of the gastrointestinal tract, the mediators, cell types and transduction pathways involved, based on experimental data obtained from models of inflammation-induced ileus and (preliminary) clinical data. The complex interplay within the gastrointestinal wall between mast cells, residential macrophages and glial cells on the one hand, and neurons and smooth muscle cells on the other hand, involves intracellular signaling pathways, Toll-like receptors and a plethora of neuroactive substances such as nitric oxide, prostaglandins, cytokines, chemokines, growth factors, tryptases and hormones. Multidirectional signaling between the different components in the gastrointestinal wall, the spinal cord and central nervous system impacts inflammation and its consequences. We propose that novel therapeutic strategies should target inflammation on the one hand and gastrointestinal motility, gastrointestinal sensitivity and even pain signaling on the other hand, for instance by impeding afferent neuronal signaling, by activation of the vagal anti-inflammatory pathway or by the use of pharmacological agents such as ghrelin and ghrelin agonists or drugs interfering with the endocannabinoid system.
Collapse
|
34
|
Li YY, Li YN, Ni JB, Chen CJ, Lv S, Chai SY, Wu RH, Yüce B, Storr M. Involvement of cannabinoid-1 and cannabinoid-2 receptors in septic ileus. Neurogastroenterol Motil 2010; 22:350-e88. [PMID: 19840270 DOI: 10.1111/j.1365-2982.2009.01419.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Cannabinoid (CB) receptors are involved in the regulation of gastrointestinal (GI) motility under physiological and pathophysiological conditions. We aimed to characterize the possible influence of CB(1) and CB(2) receptors on motility impairment in a model of septic ileus. METHODS Lipopolysaccharide (LPS) injections were used to mimic pathophysiological features of septic ileus. Spontaneous jejunal myoelectrical activity was measured in rats in vivo, and upper GI transit was measured in vivo by gavaging of a charcoal marker into the stomach of mice, in absence or presence of LPS, and CB(1) and CB(2) receptor agonists and antagonists. Tumour necrosis factor (TNF)-alpha and interleukin (IL)-6 levels were measured using enzyme-linked immunosorbent assay. Histology was performed with haematoxylin-eosin staining. KEY RESULTS Lipopolysaccharide treatment significantly reduced amplitude and frequency of myoelectric spiking activity and GI transit in vivo in a dose-dependent manner. TNF-alpha and IL-6 were increased in LPS-treated animals and histology showed oedema and cell infiltration. Both, the CB(1) agonist HU210 and the CB(2) agonist JWH133 reduced myoelectrical activity whereas the CB(1) antagonist AM251 caused an increase of myoelectrical activity. Pretreatment with AM251 or AM630 prevented against LPS-induced reduction of myoelectrical activity, and also against the delay of GI transit during septic ileus in vivo. CONCLUSIONS & INFERENCES The LPS model of septic ileus impairs jejunal myoelectrical activity and delays GI transit in vivo. Antagonists at the CB(1) receptor or the CB(2) receptor prevent the delay of GI transit and thus may be powerful tools in the future treatment of septic ileus.
Collapse
Affiliation(s)
- Y-Y Li
- Department of Pathophysiology, School of Medicine, Tongji University, Shanghai, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Buchholz BM, Bauer AJ. Membrane TLR signaling mechanisms in the gastrointestinal tract during sepsis. Neurogastroenterol Motil 2010; 22:232-45. [PMID: 20377787 PMCID: PMC2951022 DOI: 10.1111/j.1365-2982.2009.01464.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our bacterial residents are deadly Janus-faced indwellers that can lead to a sepsis-induced systemic inflammatory response syndrome and multiple organ failure. Over half of ICU patients suffer from infections and sepsis remains one of the top 10 causes of death worldwide. Severe ileus frequently accompanies sepsis setting up an insidious cycle of gut-derived microbial translocation and the copious intestinal production of potent systemic inflammatory mediators. Few therapeutic advances have occurred to prevent/treat the sequelae of sepsis. Here, we selectively review studies on cellular membrane-bound Toll-like receptor (TLR) mechanisms of ileus. Virtually, no data exist on Gram-positive/TLR2 signaling mechanisms of ileus; however, TLR2 is highly inducible by numerous inflammatory mediators and studies using clinically relevant scenarios of Gram-positive sepsis are needed. Specific Gram-negative/TLR4 signaling pathways are being elucidated using a 'reverse engineering' approach, which has revealed that endotoxin-induced ileus is dually mediated by classical leukocyte signaling and by a MyD88-dependent non-bone marrow-derived mechanism, but the specific roles of individual cell populations are still unknown. Like TLR2, little is also know of the role of flagellin/TLR5 signaling in ileus. But, much can be learned by understanding TLR signaling in other systems. Clearly, the use of polymicrobial models provides important clinical relevancy, but the simultaneous activation of virtually all pattern recognition receptors makes it impossible to discretely study specific pathways. We believe that the dissection of individual TLR pathways within the gastrointestinal tract, which can then be intelligently reassembled in a meaningful manner, will provide insight into treatments for sepsis.
Collapse
Affiliation(s)
- Bettina M. Buchholz
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA, Department of Surgery, Rheinische Friedrich-Wilhelms-Universität Bonn, Germany
| | - Anthony J. Bauer
- Department of Medicine/Gastroenterology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
36
|
The vanilloid receptor TRPV1: role in cardiovascular and gastrointestinal protection. Eur J Pharmacol 2009; 627:1-7. [PMID: 19879868 DOI: 10.1016/j.ejphar.2009.10.053] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Accepted: 10/26/2009] [Indexed: 12/21/2022]
Abstract
It has been shown that the transient receptor potential channel vanilloid type 1 (TRPV1) is able to sense a vast range of stimuli and exerts multiple functions under physiological or pathophysiological conditions. TRPV1 not only plays a fundamental role in pain signaling but also involves in many other physiological or pathophysiological functions including the beneficial effects on cardiovascular and gastrointestinal function. It has been found that TRPV1 could be activated by endogenous ligands such as anandamide, N-arachidonoyl dopamine and N-oleoyldopamine or by exogenous agonists such as capsaicin and rutaecarpine. Since capsaicin-sensitive sensory nerves (rich in TRPV1) are densely distributed in the cardiovascular and gastrointestinal system, activation of TRPV1 either by endogenous ligands or by exogenous agonists has been repeatedly reported to exert hypotensive effects or protective effects against cardiac or gastrointestinal injury through stimulating the synthesis and release of multiple neurotransmitters such as calcitonin gene-related peptide and substance P. Therefore, TRPV1 is not only a prime target for the pharmacological control of pain but also a useful target for drug development to treat various diseases including cardiovascular and gastrointestinal diseases. However, considering the contribution of TRPV1 to the development of inflammation in the gastrointestinal tract, the potential side effects of TRPV1 agonist cannot be neglected while in seeking and developing the novel TRPV1 agonists.
Collapse
|