1
|
Di Rienzi SC, Danhof HA, Forshee MD, Roberts A, Britton RA. Limosilactobacillus reuteri promotes the expression and secretion of enteroendocrine- and enterocyte-derived hormones. FASEB J 2025; 39:e70408. [PMID: 40098558 PMCID: PMC11914943 DOI: 10.1096/fj.202401669r] [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: 07/21/2024] [Revised: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 03/19/2025]
Abstract
Intestinal microbes can beneficially impact host physiology, prompting investigations into the therapeutic usage of such microbes in a range of diseases. For example, human intestinal microbe Limosilactobacillus reuteri strains ATCC PTA 6475 and DSM 17938 are being considered for use for intestinal ailments, including colic, infection, and inflammation, as well as for non-intestinal ailments, including osteoporosis, wound healing, and autism spectrum disorder. While many of their beneficial properties are attributed to suppressing inflammatory responses, we postulated that L. reuteri may also regulate intestinal hormones to affect physiology within and outside of the gut. To determine if L. reuteri secreted factors impact the secretion of enteric hormones, we treated an engineered jejunal organoid line, NGN3-HIO, which can be induced to be enriched in enteroendocrine cells, with L. reuteri 6475 or 17938 conditioned medium and performed transcriptomics. Our data suggest that these L. reuteri strains affect the transcription of many gut hormones, including vasopressin and luteinizing hormone subunit beta, which have not been previously recognized as produced in the gut epithelium. Moreover, we find that these hormones appear to be produced in enterocytes, in contrast to canonical gut hormones produced in enteroendocrine cells. Finally, we show that L. reuteri conditioned media promote the secretion of enteric hormones, including serotonin, GIP, PYY, vasopressin, and luteinizing hormone subunit beta, and identify by metabolomics metabolites potentially mediating these effects on hormones. These results support L. reuteri affecting host physiology through intestinal hormone secretion, thereby expanding our understanding of the mechanistic actions of this microbe.
Collapse
Affiliation(s)
- Sara C. Di Rienzi
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Heather A. Danhof
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Micah D. Forshee
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Ari Roberts
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| | - Robert A. Britton
- Department of Molecular Virology and MicrobiologyBaylor College of MedicineHoustonTexasUSA
- Alkek Center for Metagenomics and Microbiome ResearchBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
2
|
Brooke AK, Ojha S, Murrow DP, Ross AE. Purinergic Receptor P2Y1 Modulates Catecholamine Signaling in Murine Mesenteric Lymph Nodes. ACS Chem Neurosci 2025; 16:772-780. [PMID: 39988830 DOI: 10.1021/acschemneuro.4c00435] [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] [Indexed: 02/25/2025] Open
Abstract
Neuroimmune communication is crucial for the body's response to physiological challenges, homeostasis, and immune stress response. Adrenergic and purinergic neurotransmission in the sympathetic nervous system is vital for this communication. This study achieves the first co-detection of adenine-based purines and catecholamines in mesenteric lymph nodes via fast-scan cyclic voltammetry. Additionally, we reveal that manipulating an ATP receptor can impact catecholamine signaling in the lymph node for the first time. The G-protein-coupled receptor P2Y1, which controls intracellular Ca2+ levels, was targeted with the antagonist MRS2179. MRS2179 decreased catecholamine concentrations, increased inter-event times, and prolonged event durations. These results suggest that events became smaller, less frequent, and longer-lasting, possibly attributable to decreased intracellular Ca2+ levels. These findings indicate that ATP release in the lymph node can partially regulate norepinephrine signaling, providing mechanistic insight into sympathetic neuronal neurotransmitter control. A deeper understanding of more complicated neuroimmune mechanisms could potentially influence the development of therapeutic strategies in immunology and neurobiology.
Collapse
Affiliation(s)
- Alexandra K Brooke
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221. United States
| | - Sarbeshwar Ojha
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221. United States
| | - Daniel P Murrow
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221. United States
| | - Ashley E Ross
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221. United States
| |
Collapse
|
3
|
Lu C, Zhu H, Lu H, Xie X, Tong L, Li Y, Qian Z. Colonic dysmotility regulated by downregulation of PDGFRα+ cells / SK3 channel in DSS-induced colitis mice. PLoS One 2024; 19:e0312413. [PMID: 39689119 DOI: 10.1371/journal.pone.0312413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 10/05/2024] [Indexed: 12/19/2024] Open
Abstract
Colitis is a complex multifactorial disease with an unknown aetiology that mainly manifests as chronic refractory colon transmission disorders. Smooth muscle, the main source of colon transmission power, consists of not only smooth muscle cells (SMCs) but also PDGFRα+ cells that mediate smooth muscle relaxation and ICCs that mediate contraction. PDGFRα+ cells and their unique small conductance Ca2+-activated K (SK3) channels are crucial in regulating colonic transit by exerting inhibitory effects. In this study, the contributions of the SK3 signalling pathway in PDGFRα+ cells to colitis-induced colonic transit dysmotility were investigated in DSS-induced colitis mice. An experiment was conducted to record the transmission of waves during smooth muscle contraction in the colon, using a colonic migrating motor complex(CMMC). Western blotting was utilized for protein expression detection, while PCR was employed for gene expression analysis. Immunofluorescence staining was used to detect the co-localization of SK3 channels with PDGFRα+ cells. In the colitis group, the weight of mice was reduced, the length of colon was shortened, and the disease activity index (DAI) was significantly increased. In the CMMC experiment, colon transmission was significantly disrupted in the colitis group compared to the control group, with a consistent colonic transmission amplitude and frequency. The sensitivity of mice with colitis to SK3 antagonists and agonists (apamin and CyPPA) was lower than that of the control group in CMMC experiment. The expression levels of mRNA and protein of PDGFRα and SK3 channels in colon of mice with colitis were decreased. Less SK3 channel colocalization with PDGFRα+ cells was observed in the colitis mouse group than in the control group. The findings indicated that colonic transit disorder in DSS-induced colitis mice is caused by the down-regulation of PDGFRα+ cells / SK3 channel expression.
Collapse
Affiliation(s)
- Chen Lu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Hongxia Zhu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Haiqian Lu
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xianjing Xie
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Ling Tong
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yujia Li
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhida Qian
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| |
Collapse
|
4
|
Koh SD, Lee JY, Ryoo SB, Drumm BT, Kim HJ, Baker SA, Sanders KM. Integrated responses of the SIP syncytium generate a major motility pattern in the colon. J Physiol 2024; 602:6659-6682. [PMID: 39572771 PMCID: PMC11908618 DOI: 10.1113/jp287315] [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: 07/23/2024] [Accepted: 10/28/2024] [Indexed: 12/18/2024] Open
Abstract
The peristaltic reflex has been a central concept in gastrointestinal motility; however, evidence was published recently suggesting that post-stimulus responses that follow inhibitory neural responses provide the main propulsive force in colonic motility. This new concept was based on experiments on proximal colon where enteric inhibitory neural inputs are mainly nitrergic. However, the nature of inhibitory neural inputs changes from proximal to distal colon where purinergic inhibitory regulation dominates. In spite of the transition from nitrergic to purinergic regulation, post-stimulus responses and propulsive contractions were both blocked by antagonists of a conductance (ANO1) exclusive to interstitial cells of Cajal (ICC). How purinergic neurotransmission, transduced by PDGFRα+ cells, can influence ANO1 in ICC is unknown. We compared neural responses in proximal and distal colon. Post-stimulus responses were blocked by inhibition of nitrergic neurotransmission in proximal colon, but P2Y1 receptor antagonists were more effective in distal colon. Ca2+ entry through voltage-dependent channels (CaV3) enhances Ca2+ release in ICC. Thus, we reasoned that hyperpolarization caused by purinergic responses in PDGFRα+ cells, which are electrically coupled to ICC, might decrease inactivation of CaV3 channels and activate Ca2+ entry into ICC via anode-break upon cessation of inhibitory responses. Post-stimulus responses in distal colon were blocked by MRS2500 (P2Y1 receptor antagonist), apamin (SK channel antagonist) and NNC55-0396 (CaV3 antagonist). These compounds also blocked propagating contractions in mid and distal colon. These data provide the first clear demonstration that integration of functions in the smooth muscle-ICC-PDGFRα+ cell (SIP) syncytium generates a major motility behaviour. KEY POINTS: Propagating propulsive contractions initiated by the enteric nervous system are a major motility behaviour in the colon. A major component of contractions, necessary for propulsive contractions, occurs at cessation of enteric inhibitory neurotransmission (post-stimulus response) and is generated by interstitial cells of Cajal (ICC), which are electrically coupled to smooth muscle cells. The nature of enteric inhibitory neurotransmission shifts from proximal colon, where it is predominantly due to nitric oxide, to distal colon, where it is predominantly due to purine neurotransmitters. Different cells transduce nitric oxide and purines in the colon. ICC transduce nitric oxide, but another type of interstitial cell, PDGFRα+ cells, transduces input from purinergic neurons. However, the post-stimulus responses in proximal and distal colon are still generated in ICC. This paper explores how integrated behaviours of ICC, PDGFRα+ cells and smooth muscle cells accomplish propulsive motility in the colon.
Collapse
Affiliation(s)
- Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Ji Yeon Lee
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Seung-Bum Ryoo
- Department of Surgery, Seoul National University Hospital, Seoul National University, Seoul, South Korea
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Hyun Jin Kim
- Department of Internal Medicine, Gyeongsang National University Changwon Hospital, Gyeongsang National University, College of Medicine: Changwon, Gyeongnam-do, South Korea
| | - Sal A Baker
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| |
Collapse
|
5
|
Hendler BJ, McClain JL, Zilli A, Seguella L, Gulbransen BD. Purinergic P2Y 1 and P2Y 12 receptors control enteric nervous system activity through neuro-glia-macrophage crosstalk. Purinergic Signal 2024:10.1007/s11302-024-10060-9. [PMID: 39612055 DOI: 10.1007/s11302-024-10060-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/25/2024] [Indexed: 11/30/2024] Open
Abstract
Purines are important mediators of intercellular communication in the enteric nervous system (ENS) that participate in physiological gut functions and disease. Purinergic transmission is prominent in mechanisms of crosstalk between enteric neurons and glia where enteric glia exhibit high responsiveness to adenosine diphosphate (ADP) through P2Y1 receptors and neurons to adenosine triphosphate (ATP) through P2X3 receptors. Despite functional data suggesting that enteric glia are the primary site of P2Y1 expression in the ENS, gene sequencing suggests that P2Y1 expression is more enriched in neurons than glia. The reason for the mismatch between genomic and functional data is unclear but could involve co-expression of inhibitory P2Y12 receptors in neurons. We addressed this issue by studying the expression and function of P2Y1 and P2Y12 receptors in the mouse ENS using live immunolabeling and calcium imaging techniques. The data show that ADP drives activity among enteric glia and neurons in the myenteric plexus. Interestingly, inhibiting P2Y12 activity increased neuron responses to ADP and overall spontaneous activity among enteric neurons and glia while decreasing the magnitude of glial responses to ADP. Investigating the location of the receptors involved revealed P2Y1 receptor expression by both neurons and glia, while P2Y12 receptor expression was minimal in the ENS. Instead, P2Y12 expression was enriched in the surrounding muscularis macrophages. Macrophages positive for P2Y12 overlapped with CD163 positive subsets that have known inhibitory influences over myenteric neurocircuits. Together, these data suggest that macrophage P2Y12 pathways act to constrain activity in the ENS, which could have implications in mechanisms that contribute to enteric hyperexcitability following disease.
Collapse
Affiliation(s)
- Blake J Hendler
- Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824, USA
| | - Jonathon L McClain
- Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824, USA
| | - Aurora Zilli
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Luisa Seguella
- Department of Physiology and Pharmacology "V. Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, 567 Wilson Road, East Lansing, MI, 48824, USA.
| |
Collapse
|
6
|
Hannigan KI, Ni Bhraonain EP, Gould TW, Keef KD, Cobine CA. Modulation of intracellular calcium activity in interstitial cells of Cajal by inhibitory neural pathways within the internal anal sphincter. Am J Physiol Gastrointest Liver Physiol 2024; 327:G382-G404. [PMID: 38860285 PMCID: PMC11427099 DOI: 10.1152/ajpgi.00309.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/12/2024]
Abstract
The internal anal sphincter (IAS) functions to maintain continence. Previous studies utilizing mice with cell-specific expression of GCaMP6f revealed two distinct subtypes of intramuscular interstitial cells of Cajal (ICC-IM) with differing Ca2+ activities in the IAS. The present study further examined Ca2+ activity in ICC-IM and its modulation by inhibitory neurotransmission. The spatiotemporal properties of Ca2+ transients in Type II ICC-IM mimicked those of smooth muscle cells (SMCs), indicating their joint participation in the "SIP" syncytium. Electrical field stimulation (EFS; atropine present) abolished localized and whole cell Ca2+ transients in Type I and II ICC-IM. The purinergic antagonist MRS2500 did not abolish EFS responses in either cell type, whereas the nitric oxide synthase (NOS) inhibitor NG-nitro-l-arginine (l-NNA) abolished responses in Type I but not Type II ICC-IM. Combined antagonists abolished EFS responses in Type II ICC-IM. In both ICC-IM subtypes, the ability of EFS to inhibit Ca2+ release was abolished by l-NNA but not MRS2500, suggesting that the nitrergic pathway directly inhibits ICC-IM by blocking Ca2+ release from intracellular stores. Since inositol (1,4,5)-trisphosphate receptor-associated cGMP kinase substrate I (IRAG1) is expressed in ICC-IM, it is possible that it participates in the inhibition of Ca2+ release by nitric oxide. Platelet-derived growth factor receptor α (PDGFRα)+ cells but not ICC-IM expressed P2Y1 receptors (P2Y1R) and small-conductance Ca2+-activated K+ channels (SK3), suggesting that the purinergic pathway indirectly blocks whole cell Ca2+ transients in Type II ICC-IM via PDGFRα+ cells. This study provides the first direct evidence for functional coupling between inhibitory motor neurons and ICC-IM subtypes in the IAS, with contractile inhibition ultimately dependent upon electrical coupling between SMCs, ICC, and PDGFRα+ cells via the SIP syncytium.NEW & NOTEWORTHY Two intramuscular interstitial cells of Cajal (ICC-IM) subtypes exist within the internal anal sphincter (IAS). This study provides the first evidence for direct coupling between nitrergic motor neurons and both ICC-IM subtypes as well as indirect coupling between purinergic inputs and Type II ICC-IM. The spatiotemporal properties of whole cell Ca2+ transients in Type II ICC-IM mimic those of smooth muscle cells (SMCs), suggesting that ICC-IM modulate the activity of SMCs via their joint participation in a SIP syncytium (SMCs, ICC, and PDGFRα+ cells).
Collapse
Affiliation(s)
- Karen I Hannigan
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Emer P Ni Bhraonain
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Thomas W Gould
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Kathleen D Keef
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| | - Caroline A Cobine
- Department of Physiology and Cell BiologyUniversity of NevadaRenoNevadaUnited States
| |
Collapse
|
7
|
Rienzi SCD, Danhof HA, Forshee MD, Roberts A, Britton RA. Limosilactobacillus reuteri promotes the expression and secretion of enteroendocrine- and enterocyte-derived hormones. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610555. [PMID: 39257733 PMCID: PMC11384013 DOI: 10.1101/2024.08.30.610555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Observations that intestinal microbes can beneficially impact host physiology have prompted investigations into the therapeutic usage of such microbes in a range of diseases. For example, the human intestinal microbe Limosilactobacillus reuteri strains ATCC PTA 6475 and DSM 17938 are being considered for use for intestinal ailments including colic, infection, and inflammation as well as non-intestinal ailments including osteoporosis, wound healing, and autism spectrum disorder. While many of their beneficial properties are attributed to suppressing inflammatory responses in the gut, we postulated that L. reuteri may also regulate hormones of the gastrointestinal tract to affect physiology within and outside of the gut. To determine if L. reuteri secreted factors impact the secretion of enteric hormones, we treated an engineered jejunal organoid line, NGN3-HIO, which can be induced to be enriched in enteroendocrine cells, with L. reuteri 6475 or 17938 conditioned medium and performed transcriptomics. Our data suggest that these L. reuteri strains affect the transcription of many gut hormones, including vasopressin and luteinizing hormone subunit beta, which have not been previously recognized as being produced in the gut epithelium. Moreover, we find that these hormones appear to be produced in enterocytes, in contrast to canonical gut hormones which are produced in enteroendocrine cells. Finally, we show that L. reuteri conditioned media promotes the secretion of several enteric hormones including serotonin, GIP, PYY, vasopressin, and luteinizing hormone subunit beta. These results support L. reuteri affecting host physiology through intestinal hormone secretion, thereby expanding our understanding of the mechanistic actions of this microbe.
Collapse
Affiliation(s)
- Sara C. Di Rienzi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Heather A. Danhof
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Micah D. Forshee
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Ari Roberts
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Robert A. Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
8
|
Sanders KM, Drumm BT, Cobine CA, Baker SA. Ca 2+ dynamics in interstitial cells: foundational mechanisms for the motor patterns in the gastrointestinal tract. Physiol Rev 2024; 104:329-398. [PMID: 37561138 PMCID: PMC11281822 DOI: 10.1152/physrev.00036.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/29/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal (GI) tract displays multiple motor patterns that move nutrients and wastes through the body. Smooth muscle cells (SMCs) provide the forces necessary for GI motility, but interstitial cells, electrically coupled to SMCs, tune SMC excitability, transduce inputs from enteric motor neurons, and generate pacemaker activity that underlies major motor patterns, such as peristalsis and segmentation. The interstitial cells regulating SMCs are interstitial cells of Cajal (ICC) and PDGF receptor (PDGFR)α+ cells. Together these cells form the SIP syncytium. ICC and PDGFRα+ cells express signature Ca2+-dependent conductances: ICC express Ca2+-activated Cl- channels, encoded by Ano1, that generate inward current, and PDGFRα+ cells express Ca2+-activated K+ channels, encoded by Kcnn3, that generate outward current. The open probabilities of interstitial cell conductances are controlled by Ca2+ release from the endoplasmic reticulum. The resulting Ca2+ transients occur spontaneously in a stochastic manner. Ca2+ transients in ICC induce spontaneous transient inward currents and spontaneous transient depolarizations (STDs). Neurotransmission increases or decreases Ca2+ transients, and the resulting depolarizing or hyperpolarizing responses conduct to other cells in the SIP syncytium. In pacemaker ICC, STDs activate voltage-dependent Ca2+ influx, which initiates a cluster of Ca2+ transients and sustains activation of ANO1 channels and depolarization during slow waves. Regulation of GI motility has traditionally been described as neurogenic and myogenic. Recent advances in understanding Ca2+ handling mechanisms in interstitial cells and how these mechanisms influence motor patterns of the GI tract suggest that the term "myogenic" should be replaced by the term "SIPgenic," as this review discusses.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| | - Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Caroline A Cobine
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Salah A Baker
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| |
Collapse
|
9
|
Huan Q, Peng J, Chang Y, Zhang Q, Xing T, Jiang D, Chen W, Shen X, Bian Z, Xiao H. Activation of P2Y1R impedes intestinal mucosa repair during colitis. Int J Biol Sci 2023; 19:4360-4375. [PMID: 37781034 PMCID: PMC10535714 DOI: 10.7150/ijbs.82302] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 08/01/2023] [Indexed: 10/03/2023] Open
Abstract
Delayed intestinal mucosal healing is one of the pathogenic bases for the recurrence of inflammatory bowel disease (IBD), but how the IBD inflammatory environment impedes intestinal mucosa repair remains unclear. Adenosine diphosphate (ADP) is an endogenous ligand of P2Y1R that is highly produced at sites of inflammation. We herein identify a novel role of ADP to directly facilitate inflammation-induced epithelial permeability, delay wound healing, and disrupt tight junction integrity, and we found that P2Y1R, a receptor preferentially activated by ADP, was significantly upregulated in the colonic mucosa of ulcerative colitis (UC) patients and in colonic epithelial cells of colitis mice. Inhibition of P2Y1R significantly increased the epithelial permeability, decreased the wound healing capacity, and impaired the tight junction integrity in TNF-α-challenged Caco-2 cells. In parallel, the same effects in promoting intestinal mucosa repair were observed in DSS-induced colitis in P2Y1R-/- mice. Mechanistic investigation revealed that P2Y1R inhibition facilitated epithelial AMP-activated protein kinase (AMPK) phosphorylation and gut microbiota homeostasis reconstruction. Taken together, these findings highlight that P2Y1R activation plays an important role in impeding intestinal mucosa repair during colitis, and that P2Y1R is an attractive target for the therapy of IBD.
Collapse
Affiliation(s)
- Qiuchan Huan
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- The State Key Laboratory of Functions and Applications of Medicinal Plants and The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Jiao Peng
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yaoyao Chang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Qiansheng Zhang
- Department of Urology, Minimally Invasive Surgery Center, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tianhang Xing
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Danling Jiang
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Wenke Chen
- Department of Gastroenterology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants and The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou, China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Haitao Xiao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
10
|
Lee KY, Sung TS, Koh BH, Ryoo SB, Chun JN, Kim SH, Park KJ, So I. Distribution and Function of Platelet-derived Growth Factor Receptor Alpha-positive Cells and Purinergic Neurotransmission in the Human Colon: Is It Different Between the Right and Left Colon? J Neurogastroenterol Motil 2022; 28:678-692. [PMID: 36250374 PMCID: PMC9577575 DOI: 10.5056/jnm21117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/17/2022] [Indexed: 11/23/2022] Open
Abstract
Background/Aims Platelet-derived growth factor receptor alpha-positive (PDGFRα+) cells function in the purinergic regulation of gastrointestinal motility, and purines are reportedly inhibitory neurotransmitters in the enteric nervous system. We explore the distribution and function of PDGFRα+ cells related to purinergic inhibitory neurotransmission in human right and left colons. Methods Human colonic segments were prepared with mucosa and submucosa intact, and the circular muscle tension and longitudinal muscle tension were recorded. Purinergic neurotransmitters were administered after recording the regular contractions. Immunohistochemistry was performed on the circular muscle layers. Intracellular recording was performed on the colonic muscular layer. SK3, P2RY1, and PDGFR-α mRNA expression was tested by quantitative real-time polymerase chain reaction (qPCR). Results Adenosine triphosphate (ATP) treatment significantly decreased the frequency and area under the curve (AUC) of the segmental contraction in right and left colons. Beta-nicotinamide adenine dinucleotide (β-NAD) decreased the frequency in the right colon and the amplitude, frequency and AUC in the left colon. Apamin significantly increased frequency and AUC in the left colon, and after apamin pretreatment, ATP and β-NAD did not change segmental contractility. Through intracellular recordings, a resting membrane potential decrease occurred after ATP administration; however, the degree of decrease between the right and left colon was not different. PDGFRα+ cells were distributed evenly in the circular muscle layers of right and left colons. SK3, P2RY1, and PDGFRα expression was not different between the right and left colon. Conclusion Purines reduce right and left colon contractility similarly, and purinergic inhibitory neurotransmission can be regulated by PDGFRα+ cells in the human colon.
Collapse
Affiliation(s)
- Kil-Yong Lee
- Department of Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu-si, Gyeonggi-do, Korea
| | - Tae Sik Sung
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea.,Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, USA
| | - Byoung H Koh
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, NV, USA
| | - Seung-Bum Ryoo
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Nyeo Chun
- Department of Physiology and Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Shin-Hye Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Kyu Joo Park
- Department of Surgery, Seoul National University College of Medicine, Seoul, Korea
| | - Insuk So
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Ding F, Guo R, Cui ZY, Hu H, Zhao G. Clinical application and research progress of extracellular slow wave recording in the gastrointestinal tract. World J Gastrointest Surg 2022; 14:544-555. [PMID: 35979419 PMCID: PMC9258241 DOI: 10.4240/wjgs.v14.i6.544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/21/2022] [Accepted: 05/17/2022] [Indexed: 02/06/2023] Open
Abstract
The physiological function of the gastrointestinal (GI) tract is based on the slow wave generated and transmitted by the interstitial cells of Cajal. Extracellular myoelectric recording techniques are often used to record the characteristics and propagation of slow wave and analyze the models of slow wave transmission under physiological and pathological conditions to further explore the mechanism of GI dysfunction. This article reviews the application and research progress of electromyography, bioelectromagnetic technology, and high-resolution mapping in animal and clinical experiments, summarizes the clinical application of GI electrical stimulation therapy, and reviews the electrophysiological research in the biliary system.
Collapse
Affiliation(s)
- Fan Ding
- Center of Gallbladder Disease, East Hospital of Tongji University, Shanghai 200120, China
- Institute of Gallstone Disease, Tongji University School of Medicine, Shanghai 200331, China
| | - Run Guo
- Department of Ultrasonography, East Hospital of Tongji University, Shanghai 200120, China
| | - Zheng-Yu Cui
- Department of Internal Medicine of Traditional Chinese Medicine, East Hospital of Tongji University, Shanghai 200120, China
| | - Hai Hu
- Center of Gallbladder Disease, East Hospital of Tongji University, Shanghai 200120, China
- Institute of Gallstone Disease, Tongji University School of Medicine, Shanghai 200331, China
| | - Gang Zhao
- Center of Gallbladder Disease, East Hospital of Tongji University, Shanghai 200120, China
- Institute of Gallstone Disease, Tongji University School of Medicine, Shanghai 200331, China
| |
Collapse
|
12
|
Moon SB, Hwang SJ, Baker S, Kim M, Sasse K, Koh SD, Sanders KM, Ward SM. Changes in interstitial cells and gastric excitability in a mouse model of sleeve gastrectomy. PLoS One 2022; 17:e0269909. [PMID: 35737727 PMCID: PMC9223402 DOI: 10.1371/journal.pone.0269909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/31/2022] [Indexed: 11/18/2022] Open
Abstract
Obesity is a critical risk factor of several life-threatening diseases and the prevalence in adults has dramatically increased over the past ten years. In the USA the age-adjusted prevalence of obesity in adults was 42.4%, i.e., with a body mass index (BMI, weight (kg)/height (m)2) that exceeds 30 kg/m2. Obese individuals are at the higher risk of obesity-related diseases, co-morbid conditions, lower quality of life, and increased mortality more than those in the normal BMI range i.e., 18.5-24.9 kg/m2. Surgical treatment continues to be the most efficient and scientifically successful treatment for obese patients. Sleeve gastrectomy or vertical sleeve gastrectomy (VSG) is a relatively new gastric procedure to reduce body weight but is now the most popular bariatric operation. To date there have been few studies examining the changes in the cellular components and pacemaker activity that occur in the gastric wall following VSG and whether normal gastric activity recovers following VSG. In the present study we used a murine model to investigate the chronological changes of gastric excitability including electrophysiological, molecular and morphological changes in the gastric musculature following VSG. There is a significant disruption in specialized interstitial cells of Cajal in the gastric antrum following sleeve gastrectomy. This is associated with a loss of gastric pacemaker activity and post-junctional neuroeffector responses. Over a 4-month recovery period there was a gradual return in interstitial cells of Cajal networks, pacemaker activity and neural responses. These data describe for the first time the changes in gastric interstitial cells of Cajal networks, pacemaker activity and neuroeffector responses and the time-dependent recovery of ICC networks and normalization of motor activity and neural responses following VSG.
Collapse
Affiliation(s)
- Suk Bae Moon
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, United States of America
| | - Sung Jin Hwang
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, United States of America
| | - Sal Baker
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, United States of America
| | - Minkyung Kim
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, United States of America
| | - Kent Sasse
- Sasse Surgical Associates, Reno, Nevada, United States of America
| | - Sang Don Koh
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, United States of America
| | - Kenton M. Sanders
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, United States of America
| | - Sean M. Ward
- Department of Physiology & Cell Biology, Reno School of Medicine, University of Nevada, Reno, Nevada, United States of America
| |
Collapse
|
13
|
Kurahashi M, Baker SA, Kito Y, Bartlett A, Hara M, Takeyama H, Hashitani H, Sanders KM. PDGFRα + Interstitial Cells are Effector Cells of PACAP Signaling in Mouse and Human Colon. Cell Mol Gastroenterol Hepatol 2022; 14:357-373. [PMID: 35569815 PMCID: PMC9250024 DOI: 10.1016/j.jcmgh.2022.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 05/01/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Platelet-derived growth factor receptor α (PDGFRα)-positive interstitial cells (PIC) are interposed between enteric nerve fibers and smooth muscle cells (SMCs) in the tunica muscularis of the gastrointestinal tract. PIC have robust expression of small conductance Ca2+ activated K+ channels 3 (SK3 channels) and transduce inhibitory inputs from purinergic and sympathetic nerves in mouse and human colon. We investigated whether PIC also express pituitary adenylate cyclase-activating polypeptide (PACAP) receptors, PAC1 (PAC1R), and are involved in mediating inhibitory regulation of colonic contractions by PACAP in mouse and human colons. METHODS Gene expression analysis, Ca2+ imaging, and contractile experiments were performed on mouse colonic muscles. Ca2+ imaging, intracellular electrical recordings, and contractile experiments were performed on human colonic muscles. RESULTS Adcyap1r1 (encoding PAC1R) is highly expressed in mouse PIC. Interstitial cells of Cajal (ICC) and SMCs expressed far lower levels of Adcyap1r. Vipr1 and Vipr2 were expressed at low levels in PIC, ICC, and SMCs. PACAP elicited Ca2+ transients in mouse PIC and inhibited spontaneous phasic contractions via SK channels. In human colonic muscles, PAC1R agonists elicited Ca2+ transients in PIC, hyperpolarized SMCs through SK channels and inhibited spontaneous phasic contractions. CONCLUSIONS PIC of mouse and human colon utilize PAC1R-SK channel signal pathway to inhibit colonic contractions in response to PACAP. Effects of PACAP are in addition to the previously described purinergic and sympathetic inputs to PIC. Thus, PIC integrate inhibitory inputs from at least 3 neurotransmitters and utilize several types of receptors to activate SK channels and regulate colonic contractile behaviors.
Collapse
Affiliation(s)
- Masaaki Kurahashi
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa, Iowa City, Iowa, USA,Correspondence Address correspondence to: Masaaki Kurahashi, MD, PhD, 200 Hawkins Dr, University of Iowa, Department of Internal Medicine, Division of Gastroenterology and Hepatology, Iowa City, IA 52242. tel: (319) 467-8963.
| | - Salah A. Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Yoshihiko Kito
- Department of Pharmacology, Faculty of Medicine, Saga University, Saga, Japan
| | - Allison Bartlett
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Masayasu Hara
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiromitsu Takeyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hikaru Hashitani
- Department of Cell Physiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| |
Collapse
|
14
|
Koh SD, Drumm BT, Lu H, Kim HJ, Ryoo SB, Kim HU, Lee JY, Rhee PL, Wang Q, Gould TW, Heredia D, Perrino BA, Hwang SJ, Ward SM, Sanders KM. Propulsive colonic contractions are mediated by inhibition-driven poststimulus responses that originate in interstitial cells of Cajal. Proc Natl Acad Sci U S A 2022; 119:e2123020119. [PMID: 35446689 PMCID: PMC9170151 DOI: 10.1073/pnas.2123020119] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/17/2022] [Indexed: 12/23/2022] Open
Abstract
The peristaltic reflex is a fundamental behavior of the gastrointestinal (GI) tract in which mucosal stimulation activates propulsive contractions. The reflex occurs by stimulation of intrinsic primary afferent neurons with cell bodies in the myenteric plexus and projections to the lamina propria, distribution of information by interneurons, and activation of muscle motor neurons. The current concept is that excitatory cholinergic motor neurons are activated proximal to and inhibitory neurons are activated distal to the stimulus site. We found that atropine reduced, but did not block, colonic migrating motor complexes (CMMCs) in mouse, monkey, and human colons, suggesting a mechanism other than one activated by cholinergic neurons is involved in the generation/propagation of CMMCs. CMMCs were activated after a period of nerve stimulation in colons of each species, suggesting that the propulsive contractions of CMMCs may be due to the poststimulus excitation that follows inhibitory neural responses. Blocking nitrergic neurotransmission inhibited poststimulus excitation in muscle strips and blocked CMMCs in intact colons. Our data demonstrate that poststimulus excitation is due to increased Ca2+ transients in colonic interstitial cells of Cajal (ICC) following cessation of nitrergic, cyclic guanosine monophosphate (cGMP)-dependent inhibitory responses. The increase in Ca2+ transients after nitrergic responses activates a Ca2+-activated Cl− conductance, encoded by Ano1, in ICC. Antagonists of ANO1 channels inhibit poststimulus depolarizations in colonic muscles and CMMCs in intact colons. The poststimulus excitatory responses in ICC are linked to cGMP-inhibited cyclic adenosine monophosphate (cAMP) phosphodiesterase 3a and cAMP-dependent effects. These data suggest alternative mechanisms for generation and propagation of CMMCs in the colon.
Collapse
Affiliation(s)
- Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Bernard T. Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Hongli Lu
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Hyun Jin Kim
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Seung-Bum Ryoo
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Heung-Up Kim
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Ji Yeon Lee
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Sungkyunkwan University School of Medicine, Samsung Medical Center, Gangnam-Gu, Seoul, Korea 135-710
| | - Qianqian Wang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Thomas W. Gould
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Dante Heredia
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Brian A. Perrino
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557
| |
Collapse
|
15
|
Sanders KM, Baker SA, Drumm BT, Kurahashi M. Ca 2+ Signaling Is the Basis for Pacemaker Activity and Neurotransduction in Interstitial Cells of the GI Tract. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1383:229-241. [PMID: 36587162 DOI: 10.1007/978-3-031-05843-1_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Years ago gastrointestinal motility was thought to be due to interactions between enteric nerves and smooth muscle cells (SMCs) in the tunica muscularis. Thus, regulatory mechanisms controlling motility were either myogenic or neurogenic. Now we know that populations of interstitial cells, c-Kit+ (interstitial cells of Cajal or ICC), and PDGFRα+ cells (formerly "fibroblast-like" cells) are electrically coupled to SMCs, forming the SIP syncytium. Pacemaker and neurotransduction functions are provided by interstitial cells through Ca2+ release from the endoplasmic reticulum (ER) and activation of Ca2+-activated ion channels in the plasma membrane (PM). ICC express Ca2+-activated Cl- channels encoded by Ano1. When activated, Ano1 channels produce inward current and, therefore, depolarizing or excitatory effects in the SIP syncytium. PDGFRα+ cells express Ca2+-activated K+ channels encoded by Kcnn3. These channels generate outward current when activated and hyperpolarizing or membrane-stabilizing effects in the SIP syncytium. Inputs from enteric and sympathetic neurons regulate Ca2+ transients in ICC and PDGFRα+ cells, and currents activated in these cells conduct to SMCs and regulate contractile behaviors. ICC also serve as pacemakers, generating slow waves that are the electrophysiological basis for gastric peristalsis and intestinal segmentation. Pacemaker types of ICC express voltage-dependent Ca2+ conductances that organize Ca2+ transients, and therefore Ano1 channel openings, into clusters that define the amplitude and duration of slow waves. Ca2+ handling mechanisms are at the heart of interstitial cell function, yet little is known about what happens to Ca2+ dynamics in these cells in GI motility disorders.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA.
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Masaaki Kurahashi
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa, Iowa, Iowa City, USA
| |
Collapse
|
16
|
The role of enteric inhibitory neurons in intestinal motility. Auton Neurosci 2021; 235:102854. [PMID: 34329834 DOI: 10.1016/j.autneu.2021.102854] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 07/11/2021] [Indexed: 12/21/2022]
Abstract
The enteric nervous system controls much of the mixing and propulsion of nutrients along the digestive tract. Enteric neural circuits involve intrinsic sensory neurons, interneurons and motor neurons. While the role of the excitatory motor neurons is well established, the role of the enteric inhibitory motor neurons (IMNs) is less clear. The discovery of inhibitory transmission in the intestine in the 1960's in the laboratory of Geoff Burnstock triggered the search for the unknown neurotransmitter. It has since emerged that most neurons including the IMNs contain and may utilise more than one transmitter substances; for IMNs these include ATP, the neuropeptide VIP/PACAP and nitric oxide. This review distinguishes the enteric neural pathways underlying the 'standing reflexes' from the pathways operating physiologically during propulsive and non-propulsive movements. Morphological evidence in small laboratory animals indicates that the IMNs are located in the myenteric plexus and project aborally to the circular muscle, where they act by relaxing the muscle. There is ongoing 'tonic' activity of these IMNs to keep the intestinal muscle relaxed. Accommodatory responses to content further activate enteric pathways that involve the IMNs as the final neural element. IMNs are activated by mechanical and chemical stimulation induced by luminal contents, which activate intrinsic sensory enteric neurons and the polarised interneuronal ascending excitatory and descending inhibitory reflex pathways. The latter relaxes the muscle ahead of the advancing bolus, thus facilitating propulsion.
Collapse
|
17
|
Traserra S, Barber C, Maclnnes J, Relea L, MacPherson LC, Cunningham MR, Vergara P, Accarino A, Kennedy C, Jimenez M. Different responses of the blockade of the P2Y1 receptor with BPTU in human and porcine intestinal tissues and in cell cultures. Neurogastroenterol Motil 2021; 33:e14101. [PMID: 33619847 DOI: 10.1111/nmo.14101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 01/11/2021] [Accepted: 01/26/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Gastrointestinal smooth muscle relaxation is accomplished by activation of P2Y1 receptors, therefore this receptor plays an important role in regulation of gut motility. Recently, BPTU was developed as a negative allosteric modulator of the P2Y1 receptor. Accordingly, the aim of this study was to assess the effect of BPTU on purinergic neurotransmission in pig and human gastrointestinal tissues. METHODS Ca2+ imaging in tSA201 cells that express the human P2Y1 receptor, organ bath and microelectrodes in tissues were used to evaluate the effects of BPTU on purinergic responses. KEY RESULTS BPTU concentration dependently (0.1 and 1 µmol L-1 ) inhibited the rise in intracellular Ca2+ evoked by ADP in tSA201 cells. In the pig small intestine, 30 µmol L-1 BPTU reduced the fast inhibitory junction potential by 80%. Smooth muscle relaxations induced by electrical field stimulation were reduced both in pig ileum (EC50 = 6 µmol L-1 ) and colon (EC50 = 35 µmol L-1 ), but high concentrations of BPTU (up to 100 µmol L-1 ) had no effect on human colonic muscle. MRS2500 (1 µmol L-1 ) abolished all responses. Finally, 10 µmol L-1 ADPβS inhibited spontaneous motility and this was partially reversed by 30 µmol L-1 BPTU in pig, but not human colonic tissue and abolished by MRS2500 (1 µmol L-1 ). CONCLUSIONS & INFERENCES BPTU blocks purinergic responses elicited via P2Y1 receptors in cell cultures and in pig gastrointestinal tissue. However, the concentrations needed are higher in pig tissue compared to cell cultures and BPTU was ineffective in human colonic tissue.
Collapse
Affiliation(s)
- Sara Traserra
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Claudia Barber
- Digestive System Research Unit, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Jane Maclnnes
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Lucia Relea
- Digestive System Research Unit, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Lewis C MacPherson
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Margaret R Cunningham
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Patri Vergara
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd),, Instituto de Salud Carlos III, Madrid, Spain
| | - Anna Accarino
- Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd),, Instituto de Salud Carlos III, Madrid, Spain.,Digestive System Research Unit, University Hospital Vall d'Hebron, Barcelona, Spain
| | - Charles Kennedy
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Marcel Jimenez
- Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de enfermedades hepáticas y digestivas (CIBERehd),, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
18
|
King BF. P2X3 receptors participate in purinergic inhibition of gastrointestinal smooth muscle. Auton Neurosci 2021; 234:102830. [PMID: 34116466 DOI: 10.1016/j.autneu.2021.102830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/11/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022]
Abstract
The ATP analogue α,β-meATP is a potent relaxant of gastrointestinal smooth muscle, but its molecular target is uncertain inside the gut. α,β-meATP relaxed the carbachol-precontracted guinea-pig taenia coli in a concentration-dependent manner (EC50, 2.0 ± 0.1 μM). A luciferase-based assay confirmed that α,β-meATP solutions were minimally contaminated with ATP. α,β-meATP-evoked relaxations were inhibited by the competitive P2Y1 antagonist MRS2179 (pA2 = 5.36), but also by the competitive P2X3 antagonist, A-317491 (pA2 = 5.51). When MRS2179 and A-317491 were applied together, residual α,β-meATP responses converted from brief to prolonged relaxations. Sodium nitroprusside (a nitric oxide donor) also caused prolonged relaxations. Immunohistochemistry revealed that P2X3 receptors were present in myenteric ganglion cells and their varicose nerve terminals. The amplitude of α,β-meATP responses was not inhibited by TTX (NaV channel blocker) and ωCgTx (N-type CaV channel blocker). However, responses to α,β-meATP were inhibited by TEA (non-selective K+-channel blocker), indicating that relaxations involved opening K+-channels. The findings of this study are consistent with the conclusion that α,β-meATP stimulates Ca2+-permeable P2X3 receptors on varicose nerve terminals to release inhibitory nucleotides: 1) ATP and β-NAD release results in P2Y1-mediated brief relaxations; 2) another released transmitter (possibly NO) results in prolonged relaxations. Prejunctional P2X3 receptors represent a purinergic feed-forward mechanism to augment the action of inhibitory nerves on gut motility. This positive feed-forward mechanism may counter-balance the known negative feedback mechanism caused by adenosine and prejunctional A1 receptors on inhibitory motor nerves.
Collapse
Affiliation(s)
- Brian F King
- University College London (UCL), Research Department of Neuroscience, Pharmacology & Physiology (NPP), Gower Street, London WC1E 6BT, United Kingdom.
| |
Collapse
|
19
|
Sanders KM, Mutafova-Yambolieva VN. Neurotransmitters responsible for purinergic motor neurotransmission and regulation of GI motility. Auton Neurosci 2021; 234:102829. [PMID: 34146957 DOI: 10.1016/j.autneu.2021.102829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/17/2022]
Abstract
Classical concepts of peripheral neurotransmission were insufficient to explain enteric inhibitory neurotransmission. Geoffrey Burnstock and colleagues developed the idea that ATP or a related purine satisfies the criteria for a neurotransmitter and serves as an enteric inhibitory neurotransmitter in GI muscles. Cloning of purinergic receptors and development of specific drugs and transgenic mice have shown that enteric inhibitory responses depend upon P2Y1 receptors in post-junctional cells. The post-junctional cells that transduce purinergic neurotransmitters in the GI tract are PDGFRα+ cells and not smooth muscle cells (SMCs). PDGFRα+ cells express P2Y1 receptors, are activated by enteric inhibitory nerve stimulation and generate Ca2+ oscillations, express small-conductance Ca2+-activated K+ channels (SK3), and generate outward currents when exposed to P2Y1 agonists. These properties are consistent with post-junctional purinergic responses, and similar responses and effectors are not functional in SMCs. Refinements in methodologies to measure purines in tissue superfusates, such as high-performance liquid chromatography (HPLC) coupled with etheno-derivatization of purines and fluorescence detection, revealed that multiple purines are released during stimulation of intrinsic nerves. β-NAD+ and other purines, better satisfy criteria for the purinergic neurotransmitter than ATP. HPLC has also allowed better detection of purine metabolites, and coupled with isolation of specific types of post-junctional cells, has provided new concepts about deactivation of purine neurotransmitters. In spite of steady progress, many unknowns about purinergic neurotransmission remain and require additional investigation to understand this important regulatory mechanism in GI motility.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, School of Medicine, 1664 North Virginia Street, Reno, NV 89557, USA.
| | - Violeta N Mutafova-Yambolieva
- Department of Physiology and Cell Biology, University of Nevada, School of Medicine, 1664 North Virginia Street, Reno, NV 89557, USA
| |
Collapse
|
20
|
Kuwahara Y, Kato I, Inui T, Marunaka Y, Kuwahara A. The effect of Xenin25 on spontaneous circular muscle contractions of rat distal colon in vitro. Physiol Rep 2021; 9:e14752. [PMID: 33600071 PMCID: PMC7891183 DOI: 10.14814/phy2.14752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 09/19/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
Xenin25 has a variety of physiological functions in the Gastrointestinal (GI) tract, including ion transport and motility. However, the motility responses in the colon induced by Xenin25 remain poorly understood. Therefore, the effect of Xenin25 on the spontaneous circular muscle contractions of the rat distal colon was investigated using organ bath chambers and immunohistochemistry. Xenin25 induced the inhibition followed by postinhibitory spontaneous contractions with a higher frequency in the rat distal colon. This inhibitory effect of Xenin25 was significantly suppressed by TTX but not by atropine. The inhibitory time (the duration of inhibition) caused by Xenin25 was shortened by the NTSR1 antagonist SR48692, the NK1R antagonist CP96345, the VPAC2 receptor antagonist PG99-465, the nitric oxide-sensitive guanylate-cyclase inhibitor ODQ, and the Ca2+ -dependent K+ channel blocker apamin. The higher frequency of postinhibitory spontaneous contractions induced by Xenin25 was also attenuated by ODQ and apamin. SP-, NOS-, and VIP-immunoreactive neurons were detected in the myenteric plexus (MP) of the rat distal colon. Small subsets of the SP-positive neurons were also Calbindin positive. Most of the VIP-positive neurons were also NOS positive, and small subsets of the NK1R-positive neurons were also VIP positive. Based on the present results, we propose the following mechanism. Xenin25 activates neuronal NTSR1 on the SP neurons of IPANs, and transmitters from the VIP and apamin-sensitive NO neurons synergistically inhibit the spontaneous circular muscle contractions via NK1R. Subsequently, the postinhibitory spontaneous contractions are induced by the offset of apamin-sensitive NO neuron activation via the interstitial cells of Cajal. In addition, Xenin25 also activates the muscular NTSR1 to induce relaxation. Thus, Xenin25 is considered to be an important modulator of post prandial circular muscle contraction of distal colon since the release of Xenin25 from enteroendocrine cells is stimulated by food intake.
Collapse
Affiliation(s)
- Yuko Kuwahara
- Department of Molecular cell PhysiologyGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Research Unit for Epithelial PhysiologyResearch Center for Drug Discovery and Pharmaceutical Development ScienceResearch Organization of Science and TechnologyRitsumeikan UniversityKusatsuJapan
| | - Ikuo Kato
- Department of Medical BiochemistryKobe Pharmaceutical UniversityKobeJapan
| | | | - Yoshinori Marunaka
- Department of Molecular cell PhysiologyGraduate School of Medical ScienceKyoto Prefectural University of MedicineKyotoJapan
- Research Unit for Epithelial PhysiologyResearch Center for Drug Discovery and Pharmaceutical Development ScienceResearch Organization of Science and TechnologyRitsumeikan UniversityKusatsuJapan
- Research Institute for Clinical PhysiologyKyoto Industrial Health AssociationKyotoJapan
| | - Atsukazu Kuwahara
- Research Unit for Epithelial PhysiologyResearch Center for Drug Discovery and Pharmaceutical Development ScienceResearch Organization of Science and TechnologyRitsumeikan UniversityKusatsuJapan
| |
Collapse
|
21
|
Baker SA, Leigh WA, Del Valle G, De Yturriaga IF, Ward SM, Cobine CA, Drumm BT, Sanders KM. Ca 2+ signaling driving pacemaker activity in submucosal interstitial cells of Cajal in the murine colon. eLife 2021; 10:64099. [PMID: 33399536 PMCID: PMC7806270 DOI: 10.7554/elife.64099] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Interstitial cells of Cajal (ICC) generate pacemaker activity responsible for phasic contractions in colonic segmentation and peristalsis. ICC along the submucosal border (ICC-SM) contribute to mixing and more complex patterns of colonic motility. We show the complex patterns of Ca2+ signaling in ICC-SM and the relationship between ICC-SM Ca2+ transients and activation of smooth muscle cells (SMCs) using optogenetic tools. ICC-SM displayed rhythmic firing of Ca2+transients ~ 15 cpm and paced adjacent SMCs. The majority of spontaneous activity occurred in regular Ca2+ transients clusters (CTCs) that propagated through the network. CTCs were organized and dependent upon Ca2+ entry through voltage-dependent Ca2+ conductances, L- and T-type Ca2+ channels. Removal of Ca2+ from the external solution abolished CTCs. Ca2+ release mechanisms reduced the duration and amplitude of Ca2+ transients but did not block CTCs. These data reveal how colonic pacemaker ICC-SM exhibit complex Ca2+-firing patterns and drive smooth muscle activity and overall colonic contractions.
Collapse
Affiliation(s)
- Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Wesley A Leigh
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Guillermo Del Valle
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Inigo F De Yturriaga
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Caroline A Cobine
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| |
Collapse
|
22
|
King BF. Burnstock and the legacy of the inhibitory junction potential and P2Y1 receptors. Purinergic Signal 2020; 17:25-31. [PMID: 33125617 PMCID: PMC7954919 DOI: 10.1007/s11302-020-09747-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
The synaptic event called the inhibitory junction potential (IJP) was arguably one of the more important discoveries made by Burnstock and arguably one of his finer legacies. The discovery of the IJP fundamentally changed how electromechanical coupling was visualised in gastrointestinal smooth muscle. Its discovery also set in motion the search for novel inhibitory neurotransmitters in the enteric nervous system, eventually leading to proposal that ATP or a related nucleotide was a major inhibitory transmitter. The subsequent development of purinergic signalling gave impetus to expanding the classification of surface receptors for extracellular ATP, not only in the GI tract but beyond, and then led to successive phases of medicinal chemistry as the P2 receptor field developed. Ultimately, the discovery of the IJP led to the successful cloning of the first P2Y receptor (chick P2Y1) and expansion of mammalian ATP receptors into two classes: metabotropic P2Y receptors (encompassing P2Y1, P2Y2, P2Y4, P2Y6, P2Y11–14 receptors) and ionotropic P2X receptors (encompassing homomeric P2X1–P2X7 receptors). Here, the causal relationship between the IJP and P2Y1 is explored, setting out the milestones reached and achievements made by Burnstock and his colleagues.
Collapse
Affiliation(s)
- Brian F King
- Research Department of Neuroscience, Pharmacology & Physiology (NPP), University College London (UCL), Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
23
|
Durnin L, Kurahashi M, Sanders KM, Mutafova-Yambolieva VN. Extracellular metabolism of the enteric inhibitory neurotransmitter β-nicotinamide adenine dinucleotide (β-NAD) in the murine colon. J Physiol 2020; 598:4509-4521. [PMID: 32735345 DOI: 10.1113/jp280051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS β-Nicotinamide adenine dinucleotide (β-NAD) is a key inhibitory neurotransmitter in the colon. The neuroeffector junction in the gut consists of enteric motor neurons and SIP syncytium, including smooth muscle cells (SMCs), interstitial cells of Cajal (ICC), and cells expressing platelet-derived growth factor receptor α (PDGFRα+ cells). Measuring metabolism of 1,N6 -etheno-NAD (eNAD) in colonic tunica muscularis and in SMCs, ICC and PDGFRα+ cells with HPLC-FLD, we report that (1) in tissues, eNAD is degraded to eADP-ribose, eAMP and e-adenosine (eADO) by CD38, ENPP1 and NT5E, (2) with SMCs and PDGFRα+ cells, eNAD is metabolized to eADO by ENPP1 and NT5E, (3) eNAD is not metabolized by ICC, (4) NT5E is expressed chiefly by SMCs and moderately by PDGFRα+ cells, (5) SIP cells are not the primary location of CD38. These data argue that the duration and strength of purinergic neurotransmission can be modulated by targeting multiple enzymes with specialized cellular distribution in the colon. ABSTRACT Prior studies suggest that β-nicotinamide adenine dinucleotide (β-NAD) is an important inhibitory motor neurotransmitter in the enteric nervous system. Metabolism of β-NAD at the neuroeffector junction (NEJ) is likely to be necessary for terminating inhibitory neurotransmission and may also produce bioactive metabolites. The enteric NEJ consists of enteric neurons and postjunctional cells of the SIP syncytium, including smooth muscle cells (SMCs), interstitial cells of Cajal (ICC), and cells expressing platelet-derived growth factor receptor α (PDGFRα+ cells). We examined possible specialized functions of the NEJ in β-NAD metabolism by determining the degradation of 1,N6 -etheno-NAD (eNAD) in colonic tunica muscularis of wild-type, Cd38-/- , Nt5e-/- , Enpp1-/- and Cd38-/- /Nt5e-/- mice and in SIP cells from mice expressing cell-specific fluorescent reporters purified by fluorescence activated cell sorting (FACS). We measured eNAD and its metabolites eADP-ribose (eADPR), eAMP and e-adenosine (eADO) from tissues and sorted SIP cells using liquid chromatography. eNAD exposed to colonic muscularis of wild-type mice produced eADPR, eAMP and eADO. CD38 mediated the conversion of eNAD to eADPR, whereas ENPP1 mediated degradation of eNAD and eADPR to eAMP. NT5E (aka CD73) was the primary enzyme forming eADO from eAMP. PDGFRα+ cells and SMCs were involved in production of eADO from eNAD, and ICC were not involved in extracellular metabolism of eNAD. CD38 mediated the eNAD metabolism in whole tissues, but CD38 did not appear to be functionally expressed by SMCs or ICC. NT5E was expressed in SMCs > PDGFRα+ cells. Our data show that extracellular metabolism of β-NAD in the colon is mediated by multiple enzymes with cell-specific expression.
Collapse
Affiliation(s)
- Leonie Durnin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Masaaki Kurahashi
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Violeta N Mutafova-Yambolieva
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| |
Collapse
|
24
|
Kurahashi M, Kito Y, Hara M, Takeyama H, Sanders KM, Hashitani H. Norepinephrine Has Dual Effects on Human Colonic Contractions Through Distinct Subtypes of Alpha 1 Adrenoceptors. Cell Mol Gastroenterol Hepatol 2020; 10:658-671.e1. [PMID: 32376421 PMCID: PMC7474159 DOI: 10.1016/j.jcmgh.2020.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Colonic musculature contain smooth muscle cells (SMC), interstitial cells of Cajal (ICC), and platelet-derived growth factor receptor α+ cells (PDGFRα+ cells), which are electrically coupled and operate together as the SIP syncytium. PDGFRα+ cells have enriched expression of small conductance Ca2+-activated K+ (SK) channels. Purinergic enteric neural input activates SK channels in PDGFRα+ cells, hyperpolarizes SMC, and inhibits colonic contractions. Recently we discovered that PDGFRα+ cells in mouse colon have enriched expression of α1A adrenoceptors (ARs), which coupled to activation of SK channels and inhibited colonic motility, and α1A ARs were principal targets for sympathetic regulation of colonic motility. Here we investigated whether PDGFRα+ cells in human colon express α1A ARs and share the roles as targets for sympathetic regulation of colonic motility. METHODS Isometric tension recording, intracellular recording, and Ca2+ imaging were performed on muscles of the human colon. Responses to α1 ARs agonists or electric field stimulation with AR antagonists and neuroleptic reagents were studied. RESULTS Exogenous or endogenous norepinephrine released from nerve fibers inhibited colonic contractions through binding to α1A ARs or enhanced colonic contractions by acting on α1D ARs. Inhibitory responses were blocked by apamin, an antagonist of SK channels. Phenylephrine, α1 AR agonists, or norepinephrine increased intracellular [Ca2+] in PDGFRα+ cells, but not in ICC, and hyperpolarized SMCs by binding to α1 ARs expressed by PDGFRα+ cells. CONCLUSIONS Human colonic contractions are inhibited by α1A ARs expressed in PDGFRα+ cells and activated by α1D ARs expressed in SMC.
Collapse
Affiliation(s)
- Masaaki Kurahashi
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada.
| | - Yoshihiko Kito
- Department of Pharmacology, Saga University, Saga, Japan
| | - Masayasu Hara
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiromitsu Takeyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Hikaru Hashitani
- Department of Cell Physiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
25
|
Ma S, Yeom J, Lim YH. Exogenous NAD + Stimulates MUC2 Expression in LS 174T Goblet Cells via the PLC-Delta/PTGES/PKC-Delta/ERK/CREB Signaling Pathway. Biomolecules 2020; 10:E580. [PMID: 32283838 PMCID: PMC7226023 DOI: 10.3390/biom10040580] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND MUC2, a major component of the mucus layer in the intestine, is associated with antimicrobial activity and gut immune system function. Currently, mucin is mainly known for its critical function in defense against toxic molecules and pathogens. In this study, we investigated the stimulatory effects of exogenous nicotinamide adenine dinucleotide (NAD+) on the expression of MUC2 in LS 174T goblet cells. METHODS Genes related to MUC2 synthesis were measured by quantitative real-time PCR (qPCR). To analyze the gene expression profiles of NAD+-treated LS 174T goblet cells, RNA sequencing was performed. MUC2 expression in the cells and secreted MUC2 were measured by immunocytochemistry (ICC) and ELISA, respectively. RESULTS NAD+ significantly stimulated MUC2 expression at mRNA and protein levels and increased the secretion of MUC2. Through RNA sequencing, we found that the expression of genes involved in arachidonic acid metabolism increased in NAD+-treated cells compared with the negative control cells. NAD+ treatment increased phospholipase C (PLC)-δ and prostaglandin E synthase (PTGES) expression, which was inhibited by the appropriate inhibitors. Among the protein kinase C (PKC) isozymes, PKC-δ was involved in the increase in MUC2 expression. In addition, extracellular signal-regulated kinase (ERK)1/2 and cyclic AMP (cAMP) response element-binding protein (CREB) transcript levels were higher in NAD+-treated cells than in the negative control cells, and the enhanced levels of phosphorylated CREB augmented MUC2 expression. CONCLUSIONS Exogenous NAD+ increases MUC2 expression by stimulating the PLC-δ/PTGES/PKC-δ/ERK/CREB signaling pathway.
Collapse
Affiliation(s)
- Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea; (S.M.); (J.Y.)
| | - Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea; (S.M.); (J.Y.)
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul 02841, Korea; (S.M.); (J.Y.)
- Department of Public Health Science (Brain Korea 21 PLUS program), Graduate School, Korea University, Seoul 02841, Korea
- Department of Laboratory Medicine, Korea University Guro Hospital, Seoul 08308, Korea
| |
Collapse
|
26
|
Traserra S, Villarte S, Traini C, Palacin S, Vergara P, Vannucchi MG, Jimenez M. The asymmetric innervation of the circular and longitudinal muscle of the mouse colon differently modulates myogenic slow phasic contractions. Neurogastroenterol Motil 2020; 32:e13778. [PMID: 31845466 DOI: 10.1111/nmo.13778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 11/07/2019] [Accepted: 11/25/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Neuromuscular transmission has been extensively studied in the circular layer of the mouse colon where a co-transmission of purines acting on P2Y1 receptors and NO has been previously described. However, the corresponding mechanisms in the longitudinal layer are less known. METHODS Electrophysiological and myography techniques were used to evaluate spontaneous phasic contractions (SPC) and neural-mediated responses in the proximal, mid, and distal colon devoid of CD1 mice. Immunohistochemistry against c-kit and PDGFRα was performed in each colonic segment. KEY RESULTS SPC were recorded in both muscle layers at a similar frequency being about four contractions per minute (c.p.m.) in the proximal and distal colon compared to the mid colon (2 c.p.m.). In non-adrenergic, non-cholinergic conditions, L-NNA (1 mmol/L) increased contractility in the circular but not in the longitudinal layer. In the longitudinal muscle, both electrophysiological and mechanical neural-mediated inhibitory responses were L-NNA and ODQ (10 µmol/L) sensitive. NaNP (1 µmol/L) caused cessation of SPC and the response was blocked by ODQ. Neither ADPßS (10 µmol/L) nor CYPPA (10 µmol/L), which both targeted the purinergic pathway, altered longitudinal contractions. PDGFRα + cells were located in both muscle layers and were more numerous compared with cKit + cells, which both formed a heterologous cellular network. A decreasing gradient of the PDGFRα labeling was observed along the colon. CONCLUSION An inhibitory neural tone was absent in the longitudinal layer and neuronal inhibitory responses were mainly nitrergic. Despite the presence of PDGFRα + cells, purinergic responses were absent. Post-junctional pathways located in different cell types might be responsible for neurotransmitter transduction.
Collapse
Affiliation(s)
- Sara Traserra
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Sonia Villarte
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Chiara Traini
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Sara Palacin
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Patri Vergara
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Maria Giuliana Vannucchi
- Department of Experimental and Clinical Medicine, Research Unit of Histology and Embryology, University of Florence, Florence, Italy
| | - Marcel Jimenez
- Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, Barcelona, Spain
| |
Collapse
|
27
|
Kurahashi M, Kito Y, Baker SA, Jennings LK, Dowers JGR, Koh SD, Sanders KM. A novel postsynaptic signal pathway of sympathetic neural regulation of murine colonic motility. FASEB J 2020; 34:5563-5577. [PMID: 32086857 DOI: 10.1096/fj.201903134r] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/04/2020] [Accepted: 02/13/2020] [Indexed: 01/14/2023]
Abstract
Transcriptome data revealed α1 adrenoceptors (ARs) expression in platelet-derived growth factor receptor α+ cells (PDGFRα+ cells) in murine colonic musculature. The role of PDGFRα+ cells in sympathetic neural regulation of murine colonic motility was investigated. Norepinephrine (NE), via α1A ARs, activated a small conductance Ca2+ -activated K+ (SK) conductance, evoked outward currents and hyperpolarized PDGFRα+ cells (the α1A AR-SK channel signal pathway). α1 AR agonists increased intracellular Ca2+ transients in PDGFRα+ cells and inhibited spontaneous phasic contractions (SPCs) of colonic muscle through activation of a SK conductance. Sympathetic nerve stimulation inhibited both contractions of distal colon and propulsive contractions represented by the colonic migrating motor complexes (CMMCs) via the α1A AR-SK channel signal pathway. Postsynaptic signaling through α1A ARs in PDGFRα+ cells is a novel mechanism that conveys part of stress responses in the colon. PDGFRα+ cells appear to be a primary effector of sympathetic neural regulation of murine colonic motility.
Collapse
Affiliation(s)
- Masaaki Kurahashi
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Yoshihiko Kito
- Department of Pharmacology, Saga University, Saga, Japan
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Libby K Jennings
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - James G R Dowers
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
28
|
Rissiek B, Stabernack J, Cordes M, Duan Y, Behr S, Menzel S, Magnus T, Koch-Nolte F. Astrocytes and Microglia Are Resistant to NAD +-Mediated Cell Death Along the ARTC2/P2X7 Axis. Front Mol Neurosci 2020; 12:330. [PMID: 32009900 PMCID: PMC6971201 DOI: 10.3389/fnmol.2019.00330] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/23/2019] [Indexed: 12/31/2022] Open
Abstract
ADP-ribosylation of the P2X7k splice variant on mouse T cells by Ecto-ADP-ribosyltransferase ARTC2.2 in response to its substrate extracellular nicotinamide adenine dinucleotide (NAD+) triggers cell death. Since NAD+ is released as a danger signal during tissue damage, this NAD+-induced cell death (NICD) may impact the survival of other cell populations co-expressing P2X7 and of one of the ARTC2 isoforms (ARTC2.1, ARTC2.2). NICD of brain-resident, non-T cell populations has only been rudimentarily investigated. In this study, we evaluated the susceptibility of two glia cell populations, astrocytes and microglia, towards NICD. We found that astrocytes and microglia strongly upregulate cell surface levels of ARTC2.1 and ADP-ribosylation of cell surface proteins in response to treatment with lipopolysaccharide (LPS) and the mitogen-activated protein kinase kinase (MEK) 1 and 2 inhibitor U0126, but do not respond to extracellular NAD+ with P2X7 activation and induction of cell death. Furthermore, we found that astrocytes and microglia preferentially express the ADP-ribosylation-insensitive P2X7a splice variant, likely accounting for the resistance of these cells to NICD.
Collapse
Affiliation(s)
- Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Joschi Stabernack
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Maike Cordes
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Yinghui Duan
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Sarah Behr
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology at University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology at University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
29
|
Abstract
Purinergic signaling involves extracellular purines and pyrimidines acting upon specific cell surface purinoceptors classified into the P1, P2X, and P2Y families for nucleosides and nucleotides. This widespread signaling mechanism is active in all major tissues and influences a range of functions in health and disease. Orthologs to all but one of the human purinoceptors have been found in mouse, making this laboratory animal a useful model to study their function. Indeed, analyses of purinoceptors via knock-in or knockout approaches to produce gain or loss of function phenotypes have revealed several important therapeutic targets. None of the homozygous purinoceptor knockouts proved to be developmentally lethal, which suggest that either these receptors are not involved in key developmental processes or that the large number of receptors in each family allowed for functional compensation. Different models for the same purinoceptor often show compatible phenotypes but there have been examples of significant discrepancies. These revealed unexpected differences in the structure of human and mouse genes and emphasized the importance of the genetic background of different mouse strains. In this chapter, we provide an overview of the current knowledge and new trends in the modifications of purinoceptor genes in vivo. We discuss the resulting phenotypes, their applications and relative merits and limitations of mouse models available to study purinoceptor subtypes.
Collapse
Affiliation(s)
- Robin M H Rumney
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Dariusz C Górecki
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK.
- Military Institute of Hygiene and Epidemiology, Warsaw, Poland.
| |
Collapse
|
30
|
Spear ET, Mawe GM. Enteric neuroplasticity and dysmotility in inflammatory disease: key players and possible therapeutic targets. Am J Physiol Gastrointest Liver Physiol 2019; 317:G853-G861. [PMID: 31604034 PMCID: PMC6962496 DOI: 10.1152/ajpgi.00206.2019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal functions, including motility and secretion, are locally controlled by enteric neural networks housed within the wall of the gut. The fidelity of these functions depends on the precision of intercellular signaling among cellular elements, including enteric neurons, epithelial cells, immune cells, and glia, all of which are vulnerable to disruptive influences during inflammatory events. This review article describes current knowledge regarding inflammation-induced neuroplasticity along key elements of enteric neural circuits, what is known about the causes of these changes, and possible therapeutic targets for protecting and/or repairing the integrity of intrinsic enteric neurotransmission. Changes that have been detected in response to inflammation include increased epithelial serotonin availability, hyperexcitability of intrinsic primary afferent neurons, facilitation of synaptic activity among enteric neurons, and attenuated purinergic neuromuscular transmission. Dysfunctional propulsive motility has been detected in models of colitis, where causes include the changes described above, and in models of multiple sclerosis and other autoimmune conditions, where autoantibodies are thought to mediate dysmotility. Other cells implicated in inflammation-induced neuroplasticity include muscularis macrophages and enteric glia. Targeted treatments that are discussed include 5-hydroxytryptamine receptor 4 agonists, cyclooxygenase inhibitors, antioxidants, B cell depletion therapy, and activation of anti-inflammatory pathways.
Collapse
Affiliation(s)
- Estelle T. Spear
- 1Division of Gastroenterology and Hepatology, Department of Medicine, School of Medicine, Stanford University, Stanford, California
| | - Gary M. Mawe
- 2Department of Neurological Sciences, The University of Vermont, Burlington, Vermont
| |
Collapse
|
31
|
Diadenosine tetraphosphate activates P2Y 1 receptors that cause smooth muscle relaxation in the mouse colon. Eur J Pharmacol 2019; 855:160-166. [PMID: 31063775 DOI: 10.1016/j.ejphar.2019.05.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 05/03/2019] [Accepted: 05/03/2019] [Indexed: 01/01/2023]
Abstract
P2Y1 receptors play an essential role in inhibitory neuromuscular transmission in the gastrointestinal tract. The signalling pathway involves the opening of small conductance calcium activated potassium-channels (Kca2 family) that results in smooth muscle hyperpolarization and relaxation. Inorganic polyphosphates and dinucleotidic polyphosphates are putative neurotransmitters that potentially act on P2Y1 receptors. A pharmacological approach using both orthosteric (MRS2500) and allosteric (BPTU) blockers of the P2Y1 receptor and openers (CyPPA) and blockers (apamin) of Kca2 channels was used to pharmacologically characterise the effect of these neurotransmitters. Organ bath and microelectrodes were used to evaluate the effect of P1,P4-Di (adenosine-5') tetraphosphate ammonium salt (Ap4A), inorganic polyphosphates (PolyP) and CyPPA on spontaneous contractions and membrane potential of mouse colonic smooth muscle cells. PolyP neither modified contractions nor membrane potential. In contrast, Ap4A caused a concentration-dependent inhibition of spontaneous contractions reaching a maximum effect at 100 μM Ap4A response was antagonised by MRS2500 (1 μM), BPTU (3 μM) and apamin (1 μM). CyPPA (10 μM) inhibited spontaneous contractions and this response was antagonised by apamin but it was not affected by MRS2500 or BPTU. Both CyPPA and Ap4A caused smooth muscle hyperpolarization that was blocked by apamin and MRS2500 respectively. We conclude that Ap4A but not PolyP activates P2Y1 receptors causing smooth muscle hyperpolarization and relaxation. Ap4A signalling causes activation of Kca2 channels through activation of P2Y1 receptors. In contrast, CyPPA acts directly on Kca2 channels. Further studies are needed to evaluate if dinucleotidic polyphosphates are released from inhibitory motor neurons.
Collapse
|
32
|
Kim HJ, La JH, Kim HM, Yang IS, Sung TS. Anti-diarrheal effect of Scutellaria baicalensis is associated with suppression of smooth muscle in the rat colon. Exp Ther Med 2019; 17:4748-4756. [PMID: 31105793 DOI: 10.3892/etm.2019.7469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Scutellaria baicalensis (S. baicalensis) has been used to manage diarrhea, and its anti-inflammatory effects are responsible for anti-diarrheal effects. However, there are no data concerning its direct effect on colonic motility. Therefore, the effects of the major components of S. baicalensis (baicalin, baicalein and wogonin) on colonic motility were investigated. A segment of the distal colon of rats was placed in Krebs solution to monitor spontaneous giant contractions (GCs). Changes in GCs were recorded after applying baicalin, baicalein or wogonin. After pretreatment with Nω-nitro-L-arginine methyl ester hydrochloride (L-NAME), 1H-(1,2,4)-oxadiazolo (4,2-a) quinoxalin-1-one (ODQ), tetradotoxin, w-conotoxin, apamin, and iberiotoxin, changes in GCs by wogonin were recorded and analyzed. The segment of the distal colon showed spontaneous GCs at a mean amplitude of 3.7±0.3 g with a frequency of 0.8±0.1/min. Baicalin, baicalein, and wogonin reduced both the amplitude and the frequency of GCs in a dose-dependent manner. Wogonin had the most potent inhibitory effect on GCs (IC50 was 14.6 µM in amplitude and 14.2 µM in frequency). Wogonin-induced GC reduction was not significantly affected by the inhibition of nitric oxide/cGMP pathways with L-NAME and ODQ. Blocking the enteric neurotransmission with tetradotoxin and ω-conotoxin was ineffective on the wogonin-induced reduction of GCs. Ca2+-activated K+ (KCa) channel blockers (apamin and iberiotoxin) significantly attenuated the inhibitory effects of wogonin on GCs (P<0.01). Wogonin was effective in inhibiting colonic motility, probably through the opening of KCa channels located in the smooth muscle apparatus. These findings suggest that wogonin may be a candidate drug for the management of dysmotility-related diarrhea.
Collapse
Affiliation(s)
- Hyun Ju Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jun-Ho La
- Department of Neuroscience, Cell Biology and Anatomy, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Hee Man Kim
- Department of Internal Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea.,Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557-0357, USA
| | - Il-Suk Yang
- Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Tae Sik Sung
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557-0357, USA
| |
Collapse
|
33
|
Tonic inhibition of murine proximal colon is due to nitrergic suppression of Ca 2+ signaling in interstitial cells of Cajal. Sci Rep 2019; 9:4402. [PMID: 30867452 PMCID: PMC6416298 DOI: 10.1038/s41598-019-39729-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 01/30/2019] [Indexed: 12/18/2022] Open
Abstract
Spontaneous excitability and contractions of colonic smooth muscle cells (SMCs) are normally suppressed by inputs from inhibitory motor neurons, a behavior known as tonic inhibition. The post-junctional cell(s) mediating tonic inhibition have not been elucidated. We investigated the post-junctional cells mediating tonic inhibition in the proximal colon and whether tonic inhibition results from suppression of the activity of Ano1 channels, which are expressed exclusively in interstitial cells of Cajal (ICC). We found that tetrodotoxin (TTX), an inhibitor of nitric oxide (NO) synthesis, L-NNA, and an inhibitor of soluble guanylyl cyclase, ODQ, greatly enhanced colonic contractions. Ano1 antagonists, benzbromarone and Ani9 inhibited the effects of TTX, L-NNA and ODQ. Ano1 channels are activated by Ca2+ release from the endoplasmic reticulum (ER) in ICC, and blocking Ca2+ release with a SERCA inhibitor (thapsigargin) or a store-operated Ca2+ entry blocker (GSK 7975 A) reversed the effects of TTX, L-NNA and ODQ. Ca2+ imaging revealed that TTX, L-NNA and ODQ increased Ca2+ transient firing in colonic ICC. Our results suggest that tonic inhibition in the proximal colon occurs through suppression of Ca2+ release events in ICC. Suppression of Ca2+ release in ICC limits the open probability of Ano1 channels, reducing the excitability of electrically-coupled SMCs.
Collapse
|
34
|
Pustovit KB, Potekhina VM, Ivanova AD, Petrov AM, Abramochkin DV, Kuzmin VS. Extracellular ATP and β-NAD alter electrical properties and cholinergic effects in the rat heart in age-specific manner. Purinergic Signal 2019; 15:107-117. [PMID: 30756226 DOI: 10.1007/s11302-019-09645-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 01/04/2019] [Indexed: 12/20/2022] Open
Abstract
Extracellular ATP and nicotinamide adenine dinucleotide (β-NAD) demonstrate properties of neurotransmitters and neuromodulators in peripheral and central nervous system. It has been shown previously that ATP and β-NAD affect cardiac functioning in adult mammals. Nevertheless, the modulation of cardiac activity by purine compounds in the early postnatal development is still not elucidated. Also, the potential influence of ATP and β-NAD on cholinergic neurotransmission in the heart has not been investigated previously. Age-dependence of electrophysiological effects produced by extracellular ATP and β-NAD was studied in the rat myocardium using sharp microelectrode technique. ATP and β-NAD could affect ventricular and supraventricular myocardium independent from autonomic influences. Both purines induced reduction of action potentials (APs) duration in tissue preparations of atrial, ventricular myocardium, and myocardial sleeves of pulmonary veins from early postnatal rats similarly to myocardium of adult animals. Both purine compounds demonstrated weak age-dependence of the effect. We have estimated the ability of ATP and β-NAD to alter cholinergic effects in the heart. Both purines suppressed inhibitory effects produced by stimulation of intracardiac parasympathetic nerve in right atria from adult animals, but not in preparations from neonates. Also, ATP and β-NAD suppressed rest and evoked release of acetylcholine (ACh) in adult animals. β-NAD suppressed effects of parasympathetic stimulation and ACh release stronger than ATP. In conclusion, ATP and β-NAD control the heart at the postsynaptic and presynaptic levels via affecting the cardiac myocytes APs and ACh release. Postsynaptic and presynaptic effects of purines may be antagonistic and the latter demonstrates age-dependence.
Collapse
Affiliation(s)
- Ksenia B Pustovit
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, Russia, 119991.,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovitjanova 1, Moscow, Russia, 117997
| | - Viktoria M Potekhina
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, Russia, 119991
| | - Alexandra D Ivanova
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, Russia, 119991
| | - Alexey M Petrov
- Institute of Neuroscience, Kazan State Medial University, Butlerova st. 49, Kazan, Russia, 420012.,Laboratory of Biophysics of Synaptic Processes, Kazan Institute of Biochemistry and Biophysics, Federal Research Center "Kazan Scientific Center of RAS", P. O. Box 30, Lobachevsky Str., 2/31, Kazan, Russia, 420111
| | - Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, Russia, 119991.,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovitjanova 1, Moscow, Russia, 117997.,Laboratory of Cardiac Physiology, Institute of Physiology, Komi Science Center, Ural Division, Russian Academy of Sciences, Pervomayskaya 50, Syktyvkar, Russia, 167982
| | - Vlad S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, Russia, 119991. .,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovitjanova 1, Moscow, Russia, 117997.
| |
Collapse
|
35
|
Gallego D, Malagelada C, Accarino A, Gori A, Malagelada JR, Azpiroz F, De Giorgio R, Jimenez M. Functional neuromuscular impairment in severe intestinal dysmotility. Neurogastroenterol Motil 2018; 30:e13458. [PMID: 30178519 DOI: 10.1111/nmo.13458] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/18/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Chronic intestinal pseudo-obstruction (CIPO) and enteric dysmotility (ED) are severe intestinal motility disorders usually associated with underlying neuromuscular abnormalities. OBJECTIVE To evaluate the in vitro neuromuscular function of patients with severe intestinal motility disorders. METHODS Full-thickness intestinal biopsies (16 jejunum and 3 ileum) obtained from patients with CIPO (n = 10) and ED (n = 9) were studied using muscle bath and microelectrode techniques. Control samples (n = 6 ileum and n = 6 jejunum) were used to establish the range of normality. KEY RESULTS Fourteen parameters were defined to assess muscle contractility and nerve-muscle interaction: five to evaluate smooth muscle and interstitial cells of Cajal (ICC) and nine to evaluate inhibitory neuromuscular transmission. For each sample, a parameter was scored 0 if the value was inside the normal range or a value of 1 if it was outside. Patients' samples (CIPO/ED) had more abnormal parameters than controls (P < 0.001 for both jejunum and ileum). Functional abnormalities were found to be heterogeneous. The most prevalent abnormality was a decreased purinergic neuromuscular transmission, which was detected in 43.8% of jejunal samples. CONCLUSIONS AND INFERENCES Abnormalities of neuromuscular intestinal function are detected in vitro in severe intestinal dysmotility. However, consistent with the heterogeneity of the disease pathophysiology, functional impairment cannot be attributed to a single mechanism. Specifically, defects of purinergic neuromuscular transmission may have an important role in motility disorders of the gastrointestinal tract.
Collapse
Affiliation(s)
- Diana Gallego
- Department of Cell Biology, Physiology and Immunology and Neurosciences Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| | - Carolina Malagelada
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain.,Digestive System Research Unit, University Hospital Vall d'Hebron, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Accarino
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain.,Digestive System Research Unit, University Hospital Vall d'Hebron, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alessandra Gori
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Juan Ramón Malagelada
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain.,Digestive System Research Unit, University Hospital Vall d'Hebron, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Fernando Azpiroz
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain.,Digestive System Research Unit, University Hospital Vall d'Hebron, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Marcel Jimenez
- Department of Cell Biology, Physiology and Immunology and Neurosciences Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
| |
Collapse
|
36
|
Sung TS, Lu H, Sung J, Yeom JH, Perrino BA, Koh SD. The functional role of protease-activated receptors on contractile responses by activation of Ca 2+ sensitization pathways in simian colonic muscles. Am J Physiol Gastrointest Liver Physiol 2018; 315:G921-G931. [PMID: 30260688 PMCID: PMC6336947 DOI: 10.1152/ajpgi.00255.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
It has been known that activation of protease-activated receptors (PARs) affects gastrointestinal motility. In this study, we tested the effects of PAR agonists on electrical and contractile responses and Ca2+ sensitization pathways in simian colonic muscles. The Simian colonic muscle was initially hyperpolarized by PAR agonists. After the transient hyperpolarization, simian colonic muscle repolarized to the control resting membrane potential (RMP) without a delayed depolarization. Apamin significantly reduced the initial hyperpolarization, suggesting that activation of small conductance Ca2+-activated K+ (SK) channels is involved in the initial hyperpolarization. In contractile experiments, PAR agonists caused an initial relaxation followed by an increase in contractions. These delayed contractile responses were not matched with the electrical responses that showed no after depolarization of the RMP. To investigate the possible involvement of Rho-associated protein kinase 2 (ROCK) pathways in the PAR effects, muscle strips were treated with ROCK inhibitors, which significantly reduced the PAR agonist-induced contractions. Furthermore, PAR agonists increased MYPT1 phosphorylation, and ROCK inhibitors completely blocked MYPT1 phosphorylation. PAR agonists alone had no effect on CPI-17 phosphorylation. In the presence of apamin, PAR agonists significantly increased CPI-17 phosphorylation, which was blocked by protein kinase C (PKC) inhibitors suggesting that Ca2+ influx is increased by apamin and is activating PKC. In conclusion, these studies show that PAR activators induce biphasic responses in simian colonic muscles. The initial inhibitory responses by PAR agonists are mainly mediated by activation of SK channels and delayed contractile responses are mainly mediated by the CPI-17 and ROCK Ca2+ sensitization pathways in simian colonic muscles. NEW & NOTEWORTHY In the present study, we found that the contractile responses of simian colonic muscles to protease-activated receptor (PAR) agonists are different from the previously reported contractile responses of murine colonic muscles. Ca2+ sensitization pathways mediate the contractile responses of simian colonic muscles to PAR agonists without affecting the membrane potential. These findings emphasize novel mechanisms of PAR agonist-induced contractions possibly related to colonic dysmotility in inflammatory bowel disease.
Collapse
Affiliation(s)
- Tae Sik Sung
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Hongli Lu
- 2Department of Physiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Juno Sung
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Jong Hoon Yeom
- 3Department of Anesthesiology and Pain Medicine, Hanyang University, Seoul, Republic of Korea
| | - Brian A. Perrino
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
37
|
Friebe A, Voußen B, Groneberg D. NO-GC in cells 'off the beaten track'. Nitric Oxide 2018; 77:12-18. [DOI: 10.1016/j.niox.2018.03.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/14/2018] [Accepted: 02/23/2018] [Indexed: 02/08/2023]
|
38
|
Baker SA, Drumm BT, Cobine CA, Keef KD, Sanders KM. Inhibitory Neural Regulation of the Ca 2+ Transients in Intramuscular Interstitial Cells of Cajal in the Small Intestine. Front Physiol 2018; 9:328. [PMID: 29686622 PMCID: PMC5900014 DOI: 10.3389/fphys.2018.00328] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/15/2018] [Indexed: 01/03/2023] Open
Abstract
Gastrointestinal motility is coordinated by enteric neurons. Both inhibitory and excitatory motor neurons innervate the syncytium consisting of smooth muscle cells (SMCs) interstitial cells of Cajal (ICC) and PDGFRα+ cells (SIP syncytium). Confocal imaging of mouse small intestines from animals expressing GCaMP3 in ICC were used to investigate inhibitory neural regulation of ICC in the deep muscular plexus (ICC-DMP). We hypothesized that Ca2+ signaling in ICC-DMP can be modulated by inhibitory enteric neural input. ICC-DMP lie in close proximity to the varicosities of motor neurons and generate ongoing Ca2+ transients that underlie activation of Ca2+-dependent Cl- channels and regulate the excitability of SMCs in the SIP syncytium. Electrical field stimulation (EFS) caused inhibition of Ca2+ for the first 2-3 s of stimulation, and then Ca2+ transients escaped from inhibition. The NO donor (DEA-NONOate) inhibited Ca2+ transients and Nω-Nitro-L-arginine (L-NNA) or a guanylate cyclase inhibitor (ODQ) blocked inhibition induced by EFS. Purinergic neurotransmission did not affect Ca2+ transients in ICC-DMP. Purinergic neurotransmission elicits hyperpolarization of the SIP syncytium by activation of K+ channels in PDGFRα+ cells. Generalized hyperpolarization of SIP cells by pinacidil (KATP agonist) or MRS2365 (P2Y1 agonist) also had no effect on Ca2+ transients in ICC-DMP. Peptidergic transmitter receptors (VIP and PACAP) are expressed in ICC and can modulate ICC-DMP Ca2+ transients. In summary Ca2+ transients in ICC-DMP are blocked by enteric inhibitory neurotransmission. ICC-DMP lack a voltage-dependent mechanism for regulating Ca2+ release, and this protects Ca2+ handling in ICC-DMP from membrane potential changes in other SIP cells.
Collapse
Affiliation(s)
| | | | | | | | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno, NV, United States
| |
Collapse
|
39
|
Song NN, Lu HL, Lu C, Tong L, Huang SQ, Huang X, Chen J, Kim YC, Xu WX. Diabetes-induced colonic slow transit mediated by the up-regulation of PDGFRα + cells/SK3 in streptozotocin-induced diabetic mice. Neurogastroenterol Motil 2018; 30. [PMID: 29521017 DOI: 10.1111/nmo.13326] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 02/06/2018] [Indexed: 12/18/2022]
Abstract
BACKGROUND A major complication related to gastrointestinal (GI) symptoms in diabetic patients is chronic constipation. Constipation has serious negative impacts on quality of life; however, without a comprehensive understanding of the disease, currently available treatments cannot provide a cure. Platelet-derived growth factor receptor alpha-positive cells (PDGFRα+ cells), which form the SIP syncytium with interstitial cells of Cajal and smooth muscle cells, play important roles in GI motility. In the present study, the contributions of PDGFRα+ cells to diabetes-induced colonic slow transit were investigated in streptozotocin (STZ)-induced diabetic mice. METHODS Western blotting, quantitative PCR, contractile experiments, and intracellular recording were used in the present study. KEY RESULTS The results demonstrated that the colon length was increased in STZ-treated mice. The colonic transit of artificial fecal pellets in vitro was significantly delayed in STZ-treated mice. The mRNA and protein expression of PDGFRα, small-conductance Ca2+ -activated K channels (SK3), and P2Y1 receptors were increased in the colons of STZ-treated mice. In contractile experiments, the colonic smooth muscles were more sensitive to the SK3 agonist and antagonist (CyPPA and apamin) and the P2Y1 agonist and antagonist (MRS2365 and MRS2500) in STZ-treated mice. Intracellular recordings showed the responses of membrane potentials in colonic smooth muscle cells to CyPPA, apamin, MRS2365, and MRS2500 were more sensitive in STZ-treated mice. The electric field stimulation-induced P2Y1/SK3-dependent fast inhibitory junctional potentials (fIJPs) of colonic smooth muscles were more significantly hyperpolarized in STZ-treated mice. CONCLUSIONS AND INFERENCES These results suggest that the purinergic neurotransmitters/P2Y1/SK3 signaling pathway is up-regulated in the diabetic colons, thereby mediating diabetes-induced colonic slow transit.
Collapse
Affiliation(s)
- N-N Song
- Department of Anatomy & Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, Affiliated to Shanghai, JiaoTong University School of Medicine, Shanghai, China
| | - H-L Lu
- Department of Anatomy & Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - C Lu
- Department of Anatomy & Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - L Tong
- Department of Anatomy & Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - S-Q Huang
- Department of Anatomy & Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - X Huang
- Department of Anatomy & Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - J Chen
- Department of Pediatric Surgery, Xin Hua Hospital, Affiliated to Shanghai, JiaoTong University School of Medicine, Shanghai, China
| | - Y-C Kim
- Department of Physiology, Chungbuk National University College of Medicine, Cheongju, Chungbuk, Korea
| | - W-X Xu
- Department of Anatomy & Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Pediatric Surgery, Xin Hua Hospital, Affiliated to Shanghai, JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
40
|
Park IK, Kim JH, Park CG, Kim MY, Parajuli SP, Hong CS, Choi S, Jun JY. Effects of ATP on Pacemaker Activity of Interstitial Cells of Cajal from the Mouse Small Intestine. Chonnam Med J 2018; 54:63-71. [PMID: 29399568 PMCID: PMC5794481 DOI: 10.4068/cmj.2018.54.1.63] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 01/23/2023] Open
Abstract
Purinergic receptors play an important role in regulating gastrointestinal (GI) motility. Interstitial cells of Cajal (ICCs) are pacemaker cells that regulate GI smooth muscle activity. We studied the functional roles of external adenosine 5′-triphosphate (ATP) on pacemaker activity in cultured ICCs from mouse small intestines by using the whole-cell patch clamp technique and intracellular Ca2+ ([Ca2+]i) imaging. External ATP dose-dependently depolarized the resting membrane and produced tonic inward pacemaker currents, and these effects were antagonized by suramin, a purinergic P2 receptor antagonist. ATP-induced effects on pacemaker currents were suppressed by an external Na+-free solution and inhibited by the nonselective cation channel blockers, flufenamic acid and niflumic acid. The removal of external Ca2+ or treatment with thapsigargin (inhibitor of Ca2+ uptake into endoplasmic reticulum) inhibited the ATP-induced effects on pacemaker currents. Spontaneous [Ca2+]i oscillations were enhanced by external ATP. These results suggest that external ATP modulates pacemaker activity by activating nonselective cation channels via external Ca2+ influx and [Ca2+]i release from the endoplasmic reticulum. Thus, it seems that activating the purinergic P2 receptor may modulate GI motility by acting on ICCs in the small intestine.
Collapse
Affiliation(s)
- Il Koo Park
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Jin Ho Kim
- Department of Neurology, College of Medicine, Chosun University, Gwangju, Korea
| | - Chan Guk Park
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Man Yoo Kim
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | | | - Chan Sik Hong
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Seok Choi
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Jae Yeoul Jun
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| |
Collapse
|
41
|
Durnin L, Lees A, Manzoor S, Sasse KC, Sanders KM, Mutafova-Yambolieva VN. Loss of nitric oxide-mediated inhibition of purine neurotransmitter release in the colon in the absence of interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 2017; 313:G419-G433. [PMID: 28705804 PMCID: PMC5792210 DOI: 10.1152/ajpgi.00045.2017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/26/2017] [Accepted: 07/11/2017] [Indexed: 01/31/2023]
Abstract
Regulation of colonic motility depends on the integrity of enteric inhibitory neurotransmission mediated by nitric oxide (NO), purine neurotransmitters, and neuropeptides. Intramuscular interstitial cells of Cajal (ICC-IM) and platelet-derived growth factor receptor-α-positive (PDGFRα+) cells are involved in generating responses to NO and purine neurotransmitters, respectively. Previous studies have suggested a decreased nitrergic and increased purinergic neurotransmission in KitW/KitW-v (W/Wv ) mice that display lesions in ICC-IM along the gastrointestinal tract. However, contributions of NO to these phenotypes have not been evaluated. We used small-chamber superfusion assays and HPLC to measure the spontaneous and electrical field stimulation (EFS)-evoked release of nicotinamide adenine dinucleotide (NAD+)/ADP-ribose, uridine adenosine tetraphosphate (Up4A), adenosine 5'-triphosphate (ATP), and metabolites from the tunica muscularis of human, monkey, and murine colons and circular muscle of monkey colon, and we tested drugs that modulate NO levels or blocked NO receptors. NO inhibited EFS-evoked release of purines in the colon via presynaptic neuromodulation. Colons from W/Wv, Nos1-/- , and Prkg1-/- mice displayed augmented neural release of purines that was likely due to altered nitrergic neuromodulation. Colons from W/Wv mice demonstrated decreased nitrergic and increased purinergic relaxations in response to nerve stimulation. W/Wv mouse colons demonstrated reduced Nos1 expression and reduced NO release. Our results suggest that enhanced purinergic neurotransmission may compensate for the loss of nitrergic neurotransmission in muscles with partial loss of ICC. The interactions between nitrergic and purinergic neurotransmission in the colon provide novel insight into the role of neurotransmitters and effector cells in the neural regulation of gastrointestinal motility.NEW & NOTEWORTHY This is the first study investigating the role of nitric oxide (NO) and intramuscular interstitial cells of Cajal (ICC-IM) in modulating neural release of purines in colon. We found that NO inhibited release of purines in human, monkey, and murine colons and that colons from KitW/KitW-v (W/Wv ) mice, which present with partial loss of ICC-IM, demonstrated augmented neural release of purines. Interactions between nitrergic and purinergic neurotransmission may affect motility in disease conditions with ICC-IM deficiencies.
Collapse
Affiliation(s)
- Leonie Durnin
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Andrea Lees
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and
| | - Sheerien Manzoor
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and
| | | | - Kenton M. Sanders
- 1Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada; and
| | | |
Collapse
|
42
|
Sanders KM, Kito Y, Hwang SJ, Ward SM. Regulation of Gastrointestinal Smooth Muscle Function by Interstitial Cells. Physiology (Bethesda) 2017; 31:316-26. [PMID: 27488743 DOI: 10.1152/physiol.00006.2016] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interstitial cells of mesenchymal origin form gap junctions with smooth muscle cells in visceral smooth muscles and provide important regulatory functions. In gastrointestinal (GI) muscles, there are two distinct classes of interstitial cells, c-Kit(+) interstitial cells of Cajal and PDGFRα(+) cells, that regulate motility patterns. Loss of these cells may contribute to symptoms in GI motility disorders.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada; and
| | - Yoshihiko Kito
- Department of Pharmacology, Faculty of Medicine, Saga University, Nabeshima, Japan
| | - Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada; and
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada; and
| |
Collapse
|
43
|
Smith TK, Koh SD. A model of the enteric neural circuitry underlying the generation of rhythmic motor patterns in the colon: the role of serotonin. Am J Physiol Gastrointest Liver Physiol 2017; 312:G1-G14. [PMID: 27789457 PMCID: PMC5283906 DOI: 10.1152/ajpgi.00337.2016] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/19/2016] [Indexed: 01/31/2023]
Abstract
We discuss the role of multiple cell types involved in rhythmic motor patterns in the large intestine that include tonic inhibition of the muscle layers interrupted by rhythmic colonic migrating motor complexes (CMMCs) and secretomotor activity. We propose a model that assumes these motor patterns are dependent on myenteric descending 5-hydroxytryptamine (5-HT, serotonin) interneurons. Asynchronous firing in 5-HT neurons excite inhibitory motor neurons (IMNs) to generate tonic inhibition occurring between CMMCs. IMNs release mainly nitric oxide (NO) to inhibit the muscle, intrinsic primary afferent neurons (IPANs), glial cells, and pacemaker myenteric pacemaker interstitial cells of Cajal (ICC-MY). Mucosal release of 5-HT from enterochromaffin (EC) cells excites the mucosal endings of IPANs that synapse with 5-HT descending interneurons and perhaps ascending interneurons, thereby coupling EC cell 5-HT to myenteric 5-HT neurons, synchronizing their activity. Synchronized 5-HT neurons generate a slow excitatory postsynaptic potential in IPANs via 5-HT7 receptors and excite glial cells and ascending excitatory nerve pathways that are normally inhibited by NO. Excited glial cells release prostaglandins to inhibit IMNs (disinhibition) to allow full excitation of ICC-MY and muscle by excitatory motor neurons (EMNs). EMNs release ACh and tachykinins to excite pacemaker ICC-MY and muscle, leading to the simultaneous contraction of both the longitudinal and circular muscle layers. Myenteric 5-HT neurons also project to the submucous plexus to couple motility with secretion, especially during a CMMC. Glial cells are necessary for switching between different colonic motor behaviors. This model emphasizes the importance of myenteric 5-HT neurons and the likely consequence of their coupling and uncoupling to mucosal 5-HT by IPANs during colonic motor behaviors.
Collapse
Affiliation(s)
- Terence Keith Smith
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
44
|
Shaylor LA, Hwang SJ, Sanders KM, Ward SM. Convergence of inhibitory neural inputs regulate motor activity in the murine and monkey stomach. Am J Physiol Gastrointest Liver Physiol 2016; 311:G838-G851. [PMID: 27634009 PMCID: PMC5130542 DOI: 10.1152/ajpgi.00062.2016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 09/12/2016] [Indexed: 01/31/2023]
Abstract
Inhibitory motor neurons regulate several gastric motility patterns including receptive relaxation, gastric peristaltic motor patterns, and pyloric sphincter opening. Nitric oxide (NO) and purines have been identified as likely candidates that mediate inhibitory neural responses. However, the contribution from each neurotransmitter has received little attention in the distal stomach. The aims of this study were to identify the roles played by NO and purines in inhibitory motor responses in the antrums of mice and monkeys. By using wild-type mice and mutants with genetically deleted neural nitric oxide synthase (Nos1-/-) and P2Y1 receptors (P2ry1-/-) we examined the roles of NO and purines in postjunctional inhibitory responses in the distal stomach and compared these responses to those in primate stomach. Activation of inhibitory motor nerves using electrical field stimulation (EFS) produced frequency-dependent inhibitory junction potentials (IJPs) that produced muscle relaxations in both species. Stimulation of inhibitory nerves during slow waves terminated pacemaker events and associated contractions. In Nos1-/- mice IJPs and relaxations persisted whereas in P2ry1-/- mice IJPs were absent but relaxations persisted. In the gastric antrum of the non-human primate model Macaca fascicularis, similar NO and purine neural components contributed to inhibition of gastric motor activity. These data support a role of convergent inhibitory neural responses in the regulation of gastric motor activity across diverse species.
Collapse
Affiliation(s)
- Lara A. Shaylor
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sean M. Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
45
|
Durnin L, Moreland N, Lees A, Mutafova-Yambolieva VN. A commonly used ecto-ATPase inhibitor, ARL-67156, blocks degradation of ADP more than the degradation of ATP in murine colon. Neurogastroenterol Motil 2016; 28:1370-81. [PMID: 27060478 PMCID: PMC5002237 DOI: 10.1111/nmo.12836] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 03/14/2016] [Indexed: 01/24/2023]
Abstract
BACKGROUND Adenosine 5'-triphosphate (ATP) is released extracellularly as a neurotransmitter and an autocrine or paracrine mediator in numerous systems, including the gastrointestinal tract. It is rapidly degraded to active and inactive metabolites by membrane-bound enzymes. Investigators frequently use inhibitors of ATP hydrolysis such as ARL-67156 and POM-1 to suppress the catabolism of ATP and prolong its effects in pharmacological studies. Our aim was to investigate directly the effects of ARL-67156 and POM-1 on the degradation of ATP and adenosine 5'-diphosphate (ADP) in mouse colonic muscles. METHODS The degradation of ATP and ADP was evaluated by superfusing tissues with 1,N(6) -etheno-ATP (eATP) and 1,N(6) -etheno-ADP (eADP) as substrates and monitoring the decrease in substrate and increase in products (i.e., eADP, eAMP, and e-adenosine) by high-performance liquid chromatography techniques with fluorescence detection. Relaxation responses to etheno-derivatized and non-derivatized ATP and ADP were examined in isometric tension experiments. KEY RESULTS ARL-67156 inhibits the degradation of ADP but not of ATP, whereas POM-1 inhibits the degradation of ATP but not of ADP in murine colonic muscles. Consequently, ARL-67156 enhances relaxation responses to both ATP and ADP, whereas POM-1 reduces relaxation to ATP and does not affect relaxation to ADP. CONCLUSIONS & INFERENCES Studies that use ARL-67156 to inhibit ATP degradation in smooth muscle likely evaluate responses to accumulated ADP rather than ATP. POM-1 appears to be a more selective inhibitor of ATP degradation in the mouse colon. The choice of pharmacological tools in studies on extracellular ATP signaling may affect the interpretation of experimental data in functional studies.
Collapse
Affiliation(s)
- L. Durnin
- Department of Physiology and Cell Biology; University of Nevada School of Medicine; Reno NV USA
| | - N. Moreland
- Department of Physiology and Cell Biology; University of Nevada School of Medicine; Reno NV USA
| | - A. Lees
- Department of Physiology and Cell Biology; University of Nevada School of Medicine; Reno NV USA
| | | |
Collapse
|
46
|
Mañé N, Jiménez-Sábado V, Jiménez M. BPTU, an allosteric antagonist of P2Y1 receptor, blocks nerve mediated inhibitory neuromuscular responses in the gastrointestinal tract of rodents. Neuropharmacology 2016; 110:376-385. [PMID: 27496690 DOI: 10.1016/j.neuropharm.2016.07.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 06/28/2016] [Accepted: 07/26/2016] [Indexed: 12/28/2022]
Abstract
P2Y1 receptors mediate nerve mediated purinergic inhibitory junction potentials (IJP) and relaxations in the gastrointestinal (GI) tract in a wide range of species including rodents and humans. A new P2Y1 antagonist, with a non-nucleotide structure, BPTU, has recently been described using X-ray crystallography as the first allosteric G-protein-coupled receptor antagonist located entirely outside of the helical bundle. In this study, we tested its effect on purinergic responses in the gastrointestinal tract of rodents using electrophysiological and myographic techniques. BPTU concentration dependently inhibited purinergic inhibitory junction potentials and inhibition of spontaneous motility induced by electrical field stimulation in the colon of rats (EC50 = 0.3 μM) and mice (EC50 = 0.06 μM). Mechanical inhibitory responses were also concentration-dependently blocked in the stomach of both species. Compared to MRS2500, BPTU displays a lower potency. In the rat colon nicotine induced relaxation was also blocked by BPTU. BPTU also blocked the cessation of spontaneous contractility elicited by ADPβS and the P2Y1 agonist MRS2365. We conclude that BPTU is a novel antagonist with different structural and functional properties than nucleotidic antagonists that is able to block the P2Y1 receptor located at the neuromuscular junction of the GI tract.
Collapse
Affiliation(s)
- Noemí Mañé
- Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Verónica Jiménez-Sábado
- Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Marcel Jiménez
- Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, (CIBERehd), Instituto de Salud Carlos III, Barcelona, Spain.
| |
Collapse
|
47
|
Durnin L, Hayoz S, Corrigan RD, Yanez A, Koh SD, Mutafova-Yambolieva VN. Urothelial purine release during filling of murine and primate bladders. Am J Physiol Renal Physiol 2016; 311:F708-F716. [PMID: 27465992 DOI: 10.1152/ajprenal.00387.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 07/26/2016] [Indexed: 01/15/2023] Open
Abstract
During urinary bladder filling the bladder urothelium releases chemical mediators that in turn transmit information to the nervous and muscular systems to regulate sensory sensation and detrusor muscle activity. Defects in release of urothelial mediators may cause bladder dysfunctions that are characterized with aberrant bladder sensation during bladder filling. Previous studies have demonstrated release of ATP from the bladder urothelium during bladder filling, and ATP remains the most studied purine mediator that is released from the urothelium. However, the micturition cycle is likely regulated by multiple purine mediators, since various purine receptors are found present in many cell types in the bladder wall, including urothelial cells, afferent nerves, interstitial cells in lamina propria, and detrusor smooth muscle cells. Information about the release of other biologically active purines during bladder filling is still lacking. Decentralized bladders from C57BL/6 mice and Cynomolgus monkeys (Macaca fascicularis) were filled with physiological solution at different rates. Intraluminal fluid was analyzed by high-performance liquid chromatography with fluorescence detection for simultaneous evaluation of ATP, ADP, AMP, adenosine, nicotinamide adenine dinucleotide (NAD+), ADP-ribose, and cADP-ribose content. We also measured ex vivo bladder filling pressures and performed cystometry in conscious unrestrained mice at different filling rates. ATP, ADP, AMP, NAD+, ADPR, cADPR, and adenosine were detected released intravesically at different ratios during bladder filling. Purine release increased with increased volumes and rates of filling. Our results support the concept that multiple urothelium-derived purines likely contribute to the complex regulation of bladder sensation during bladder filling.
Collapse
Affiliation(s)
- Leonie Durnin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sebastien Hayoz
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Robert D Corrigan
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Andrew Yanez
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | | |
Collapse
|
48
|
Kuzmin VS, Pustovit KB, Abramochkin DV. Effects of exogenous nicotinamide adenine dinucleotide (NAD+) in the rat heart are mediated by P2 purine receptors. J Biomed Sci 2016; 23:50. [PMID: 27350532 PMCID: PMC4924331 DOI: 10.1186/s12929-016-0267-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 06/17/2016] [Indexed: 12/13/2022] Open
Abstract
Background Recently, NAD+ has been considered as an essential factor, participating in nerve control of physiological functions and intercellular communication. NAD+ also has been supposed as endogenous activator of P1 and P2 purinoreceptors. Effects of extracellular NAD+ remain poorly investigated in cardiac tissue. This study aims to investigate the effects of extracellular NAD+ in different types of supraventricular and ventricular working myocardium from rat and their potential mechanisms. Methods The standard technique of sharp microelectrode action potential recording in cardiac multicellular preparations was used to study the effects of NAD+. Results Extracellular NAD+ induced significant changes in bioelectrical activity of left auricle (LA), right auricle (RA), pulmonary veins (PV) and right ventricular wall (RV) myocardial preparations. 10–100 μM NAD+ produced two opposite effects in LA and RA – quickly developing and transient prolongation of action potentials (AP) and delayed sustained AP shortening, which follows the initial positive effect. In PV and RV only AP shortening was observed in response to NAD+ application. In PV preparations AP shortening induced by NAD+ may be considered as a potential proarrhythmic effect. Revealed cardiotropic effects of NAD+ are likely to be mediated by P2 purine receptors, since P1 blocker DPCPX failed to affect them and P2 antagonist suramin abolished NAD + −induced alterations of electrical activity. P2X receptors may be responsible for NAD + −induced short-lasting AP prolongation, while P2Y receptors mediate persistent AP shortening. The latter effect is partially removed by PLC inhibitor U73122 showing the potential involvement of phosphoinositide signaling pathway in mediation of NAD+ cardiotropic effects. Conclusions Extracellular NAD+ is supposed to be a novel regulator of cardiac electrical activity. P2 receptors represent the main target of NAD+ at least in the rat heart.
Collapse
Affiliation(s)
- Vladislav S Kuzmin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, 119991, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovitjanova 1, Moscow, 117997, Russia
| | - Ksenia B Pustovit
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, 119991, Russia.,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovitjanova 1, Moscow, 117997, Russia
| | - Denis V Abramochkin
- Department of Human and Animal Physiology, Lomonosov Moscow State University, Leninskie gory 1, building 12, Moscow, 119991, Russia. .,Department of Physiology, Pirogov Russian National Research Medical University, Ostrovitjanova 1, Moscow, 117997, Russia.
| |
Collapse
|
49
|
P2Y receptor-mediated transient relaxation of rat longitudinal ileum preparations involves phospholipase C activation, intracellular Ca(2+) release and SK channel activation. Acta Pharmacol Sin 2016; 37:617-28. [PMID: 27018177 DOI: 10.1038/aps.2015.137] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 12/10/2015] [Indexed: 12/13/2022]
Abstract
AIM Purinergic signaling plays a major role in the enteric nervous system, where it governs gut motility through a number of P2X and P2Y receptors. The aim of this study was to investigate the P2Y receptor-mediated motility in rat longitudinal ileum preparations. METHODS Ileum smooth muscle strips were prepared from rats, and fixed in an organ bath. Isometric contraction and relaxation responses of the muscle strips were measured with force transducers. Drugs were applied by adding of stock solutions to the organ bath to yield the individual final concentrations. RESULTS Application of the non-hydrolyzable P2 receptor agonists α,β-Me-ATP or 2-Me-S-ADP (10, 100 μmol/L) dose-dependently elicited a transient relaxation response followed by a sustained contraction. The relaxation response was largely blocked by SK channel blockers apamin (500 nmol/L) and UCL1684 (10 μmol/L), PLC inhibitor U73122 (100 μmol/L), IP3 receptor blocker 2-APB (100 μmol/L) or sarcoendoplasmic Ca(2+) ATPase inhibitor thapsigargin (1 μmol/L), but not affected by atropine, NO synthase blocker L-NAME or tetrodotoxin. Furthermore, α,β-Me-ATP-induced relaxation was suppressed by P2Y1 receptor antagonist MRS2179 (50 μmol/L) or P2Y13 receptor antagonist MRS2211 (100 μmol/L), and was abolished by co-application of the two antagonists, whereas 2-Me-S-ADP-induced relaxation was abolished by P2Y6 receptor antagonist MRS2578 (50 μmol/L). In addition, P2Y1 receptor antagonist MRS2500 (1 μmol/L) not only abolished α,β-Me-ATP-induced relaxation, but also suppressed 2-Me-S-ADP-induced relaxation. CONCLUSION P2Y receptor agonist-induced transient relaxation of rat ileum smooth muscle strips is mediated predominantly by P2Y1 receptor, but also by P2Y6 and P2Y13 receptors, and involves PLC, IP3, Ca(2+) release and SK channel activation, but is independent of acetylcholine and NO release.
Collapse
|
50
|
Reed DE, Zhang Y, Beyak MJ, Lourenssen S, Blennerhassett MG, Paterson WG, Vanner SJ. Stress increases descending inhibition in mouse and human colon. Neurogastroenterol Motil 2016; 28:569-80. [PMID: 26744175 DOI: 10.1111/nmo.12755] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 11/16/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND A relationship between stress and the symptoms of irritable bowel syndrome (IBS) has been well established but the cellular mechanisms are poorly understood. Therefore, we investigated effects of stress and stress hormones on colonic descending inhibition and transit in mouse models and human tissues. METHODS Stress was applied using water avoidance stress (WAS) in the animal model or mimicked using stress hormones, adrenaline (5 nM), and corticosterone (1 μM). Intracellular recordings were obtained from colonic circular smooth muscle cells in isolated smooth muscle/myenteric plexus preparations and the inhibitory junction potential (IJP) was elicited by nerve stimulation or balloon distension oral to the site of recording. KEY RESULTS Water avoidance stress increased the number of fecal pellets compared to control (p < 0.05). WAS also caused a significant increase in IJP amplitude following balloon distension. Stress hormones also increased the IJP amplitude following nerve stimulation and balloon distension (p < 0.05) in control mice but had no effect in colons from stressed mice. No differences were observed with application of ATP between stress and control tissues, suggesting the actions of stress hormones were presynaptic. Stress hormones had a large effect in the nerve stimulated IJP in human colon (increased >50%). Immunohistochemical studies identified alpha and beta adrenergic receptor immunoreactivity on myenteric neurons in human colon. CONCLUSIONS & INFERENCES These studies suggest that WAS and stress hormones can signal via myenteric neurons to increase inhibitory neuromuscular transmission. This could lead to greater descending relaxation, decreased transit time, and subsequent diarrhea.
Collapse
Affiliation(s)
- D E Reed
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - Y Zhang
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - M J Beyak
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - S Lourenssen
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | | | - W G Paterson
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - S J Vanner
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| |
Collapse
|