1
|
Liu J, Zhang S, Emadi S, Guo T, Chen L, Feng B. Morphological, molecular, and functional characterization of mouse glutamatergic myenteric neurons. Am J Physiol Gastrointest Liver Physiol 2024; 326:G279-G290. [PMID: 38193160 PMCID: PMC11211033 DOI: 10.1152/ajpgi.00200.2023] [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: 09/18/2023] [Revised: 12/26/2023] [Accepted: 01/02/2024] [Indexed: 01/10/2024]
Abstract
The enteric nervous system (ENS) functions largely independently of the central nervous system (CNS). Glutamate, the dominant neurotransmitter in the CNS and sensory afferents, is not a primary neurotransmitter in the ENS. Only a fraction (∼2%) of myenteric neurons in the mouse distal colon and rectum (colorectum) are positive for vesicular glutamate transporter type 2 (VGLUT2), the structure and function of which remain undetermined. Here, we systematically characterized VGLUT2-positive enteric neurons (VGLUT2-ENs) through sparse labeling with adeno-associated virus, single-cell mRNA sequencing (scRNA-seq), and GCaMP6f calcium imaging. Our results reveal that the majority of VGLUT2-ENs (29 of 31, 93.5%) exhibited Dogiel type I morphology with a single aborally projecting axon; most axons (26 of 29, 89.7%) are between 4 and 10 mm long, each traversing 19 to 34 myenteric ganglia. These anatomical features exclude the VGLUT2-ENs from being intrinsic primary afferent or motor neurons. The scRNA-seq conducted on 52 VGLUT2-ENs suggests different expression profiles from conventional descending interneurons. Ex vivo GCaMP6f recordings from flattened colorectum indicate that almost all VGLUT2-EN (181 of 215, 84.2%) are indirectly activated by colorectal stretch via nicotinic cholinergic neural transmission. In conclusion, VGLUT2-ENs are a functionally unique group of enteric neurons with single aborally projecting long axons that traverse multiple myenteric ganglia and are activated indirectly by colorectal mechanical stretch. This knowledge will provide a solid foundation for subsequent studies on the potential interactions of VGLUT2-EN with extrinsic colorectal afferents via glutamatergic neurotransmission.NEW & NOTEWORTHY We reveal that VGLUT2-positive enteric neurons (EN), although constituting a small fraction of total EN, are homogeneously expressed in the myenteric ganglia, with a slight concentration at the intermediate region between the colon and rectum. Through anatomic, molecular, and functional analyses, we demonstrated that VGLUT2-ENs are activated indirectly by noxious circumferential colorectal stretch via nicotinic cholinergic transmission, suggesting their participation in mechanical visceral nociception.
Collapse
Affiliation(s)
- Jia Liu
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Shaopeng Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Sharareh Emadi
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Tiantian Guo
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Longtu Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut, United States
| |
Collapse
|
2
|
Liu J, Zhang S, Emadi S, Guo T, Chen L, Feng B. Morphological, molecular, and functional characterization of mouse glutamatergic myenteric neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.558146. [PMID: 37781576 PMCID: PMC10541117 DOI: 10.1101/2023.09.18.558146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The enteric nervous system (ENS) functions largely independently of the central nervous system (CNS). Correspondingly, glutamate, the dominant neurotransmitter in the CNS and sensory afferents, is not a primary neurotransmitter in the ENS. Only a fraction (approximately 2%) of myenteric neurons in the mouse distal colon and rectum (colorectum) are positive for vesicular glutamate transporter type 2 (VGLUT2), the structure and function of which remain undetermined. Here, we systematically characterized VGLUT2-positive enteric neurons (VGLUT2-ENs) through sparse labeling with adeno-associated virus, single-cell mRNA sequencing (scRNA-seq), and GCaMP6f calcium imaging. Our results reveal that the majority of VGLUT2-ENs (29 out of 31, 93.5%) exhibited Dogiel type I morphology with a single aborally projecting axon; most axons (26 out of 29, 89.7%) are between 4 and 10 mm long, each traversing 19 to 34 myenteric ganglia. These anatomical features exclude the VGLUT2-ENs from being intrinsic primary afferent or motor neurons. The scRNA-seq conducted on 52 VGLUT2-ENs suggests different expression profiles from conventional descending interneurons. Ex vivo GCaMP6f recordings from flattened colorectum indicate that almost all VGLUT2-EN (181 out of 215, 84.2%) are indirectly activated by colorectal stretch via nicotinic cholinergic neural transmission. In conclusion, VGLUT2-ENs are a functionally unique group of enteric neurons with single aborally projecting long axons that traverse multiple myenteric ganglia and are activated indirectly by colorectal mechanical stretch. This knowledge will provide a solid foundation for subsequent studies on the potential interactions of VGLUT2-EN with extrinsic colorectal afferents via glutamatergic neurotransmission. New & Noteworthy We reveal that VGLUT2-positive enteric neurons (EN), although constituting a small fraction of total EN, are homogeneously expressed in the myenteric ganglia, with a slight concentration at the intermediate region between the colon and rectum. This concentration coincides with the entry zone of extrinsic afferents into the colorectum. Given that VGLUT2-ENs are indirectly activated by colorectal mechanical stretch, they are likely to participate in visceral nociception through glutamatergic neural transmission with extrinsic afferents.
Collapse
|
3
|
Sharkey KA, Mawe GM. The enteric nervous system. Physiol Rev 2023; 103:1487-1564. [PMID: 36521049 PMCID: PMC9970663 DOI: 10.1152/physrev.00018.2022] [Citation(s) in RCA: 118] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Of all the organ systems in the body, the gastrointestinal tract is the most complicated in terms of the numbers of structures involved, each with different functions, and the numbers and types of signaling molecules utilized. The digestion of food and absorption of nutrients, electrolytes, and water occurs in a hostile luminal environment that contains a large and diverse microbiota. At the core of regulatory control of the digestive and defensive functions of the gastrointestinal tract is the enteric nervous system (ENS), a complex system of neurons and glia in the gut wall. In this review, we discuss 1) the intrinsic neural control of gut functions involved in digestion and 2) how the ENS interacts with the immune system, gut microbiota, and epithelium to maintain mucosal defense and barrier function. We highlight developments that have revolutionized our understanding of the physiology and pathophysiology of enteric neural control. These include a new understanding of the molecular architecture of the ENS, the organization and function of enteric motor circuits, and the roles of enteric glia. We explore the transduction of luminal stimuli by enteroendocrine cells, the regulation of intestinal barrier function by enteric neurons and glia, local immune control by the ENS, and the role of the gut microbiota in regulating the structure and function of the ENS. Multifunctional enteric neurons work together with enteric glial cells, macrophages, interstitial cells, and enteroendocrine cells integrating an array of signals to initiate outputs that are precisely regulated in space and time to control digestion and intestinal homeostasis.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Gary M Mawe
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont
| |
Collapse
|
4
|
Chen W, Liao L, Huang Z, Lu Y, Lin Y, Pei Y, Yi S, Huang C, Cao H, Tan B. Patchouli alcohol improved diarrhea-predominant irritable bowel syndrome by regulating excitatory neurotransmission in the myenteric plexus of rats. Front Pharmacol 2022; 13:943119. [PMID: 36452228 PMCID: PMC9703083 DOI: 10.3389/fphar.2022.943119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 10/31/2022] [Indexed: 09/07/2023] Open
Abstract
Background and Purpose: Irritable bowel syndrome (IBS) is usually associated with chronic gastrointestinal disorders. Its most common subtype is accompanied with diarrhea (IBS-D). The enteric nervous system (ENS) modulates major gastrointestinal motility and functions whose aberration may induce IBS-D. The enteric neurons are susceptible to long-term neurotransmitter level alterations. The patchouli alcohol (PA), extracted from Pogostemonis Herba, has been reported to regulate neurotransmitter release in the ENS, while its effectiveness against IBS-D and the underlying mechanism remain unknown. Experimental Approach: In this study, we established an IBS-D model in rats through chronic restraint stress. We administered the rats with 5, 10, and 20 mg/kg of PA for intestinal and visceral examinations. The longitudinal muscle myenteric plexus (LMMP) neurons were further immunohistochemically stained for quantitative, morphological, and neurotransmitters analyses. Key Results: We found that PA decreased visceral sensitivity, diarrhea symptoms and intestinal transit in the IBS-D rats. Meanwhile, 10 and 20 mg/kg of PA significantly reduced the proportion of excitatory LMMP neurons in the distal colon, decreased the number of acetylcholine (Ach)- and substance P (SP)-positive neurons in the distal colon and restored the levels of Ach and SP in the IBS-D rats. Conclusion and Implications: These findings indicated that PA modulated LMMP excitatory neuron activities, improved intestinal motility and alleviated IBS-induced diarrheal symptoms, suggesting the potential therapeutic efficacy of PA against IBS-D.
Collapse
Affiliation(s)
- Wanyu Chen
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lu Liao
- Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Guangzhou, China
| | - Zitong Huang
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yulin Lu
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yukang Lin
- College of Integrated Chinese and Western Medicines, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Pei
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shulin Yi
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chen Huang
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongying Cao
- School of Chinese Materia Medica, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Tan
- Research Centre of Basic Intergrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
5
|
Abstract
Chemotherapy-induced gastrointestinal dysfunction is a common occurrence associated with many different classes of chemotherapeutic agents. Gastrointestinal toxicity includes mucositis, diarrhea, and constipation, and can often be a dose-limiting complication, induce cessation of treatment and could be life threatening. The gastrointestinal epithelium is rich in rapidly dividing cells and hence is a prime target for chemotherapeutic drugs. The incidence of gastrointestinal toxicity, including diarrhea and mucositis, is extremely high for a wide array of chemotherapeutic and radiation regimens. In fact, 60%-100% of patients on high-dose chemotherapy suffer from gastrointestinal side effects. Unfortunately, treatment options are limited, and therapy is often restricted to palliative care. Therefore, there is a great unmet therapeutic need for preventing and treating chemotherapy-induced gastrointestinal toxicities in the clinic. In this review, we discuss our current understanding of the mechanisms underlying chemotherapy-induced diarrhea and mucositis, and emerging mechanisms involving the enteric nervous system, smooth muscle cells and enteric immune cells. Recent evidence has also implicated gut dysbiosis in the pathogenesis of not only chemotherapy-induced mucositis and diarrhea, but also chemotherapy-induced peripheral neuropathy. Oxidative stress induced by chemotherapeutic agents results in post-translational modification of ion channels altering neuronal excitability. Thus, investigating how chemotherapy-induced changes in the gut- microbiome axis may lead to gut-related toxicities will be critical in the discovery of new drug targets for mitigating adverse gastrointestinal effects associated with chemotherapy treatment.
Collapse
Affiliation(s)
- Hamid I Akbarali
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States.
| | - Karan H Muchhala
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Donald K Jessup
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Stanley Cheatham
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
6
|
Feng J, Hibberd TJ, Luo J, Yang P, Xie Z, Travis L, Spencer NJ, Hu H. Modification of Neurogenic Colonic Motor Behaviours by Chemogenetic Ablation of Calretinin Neurons. Front Cell Neurosci 2022; 16:799717. [PMID: 35317196 PMCID: PMC8934436 DOI: 10.3389/fncel.2022.799717] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 01/31/2022] [Indexed: 12/31/2022] Open
Abstract
How the enteric nervous system determines the pacing and propagation direction of neurogenic contractions along the colon remains largely unknown. We used a chemogenetic strategy to ablate enteric neurons expressing calretinin (CAL). Mice expressing human diphtheria toxin receptor (DTR) in CAL neurons were generated by crossing CAL-ires-Cre mice with Cre-dependent ROSA26-DTR mice. Immunohistochemical analysis revealed treatment with diphtheria toxin incurred a 42% reduction in counts of Hu-expressing colonic myenteric neurons (P = 0.036), and 57% loss of CAL neurons (comprising ∼25% of all Hu neurons; P = 0.004) compared to control. As proportions of Hu-expressing neurons, CAL neurons that contained nitric oxide synthase (NOS) were relatively spared (control: 15 ± 2%, CAL-DTR: 13 ± 1%; P = 0.145), while calretinin neurons lacking NOS were significantly reduced (control: 26 ± 2%, CAL-DTR: 18 ± 5%; P = 0.010). Colonic length and pellet sizes were significantly reduced without overt inflammation or changes in ganglionic density. Interestingly, colonic motor complexes (CMCs) persisted with increased frequency (mid-colon interval 111 ± 19 vs. 189 ± 24 s, CAL-DTR vs. control, respectively, P < 0.001), decreased contraction size (mid-colon AUC 26 ± 24 vs. 59 ± 13 gram/seconds, CAL-DTR vs. control, respectively, P < 0.001), and lacked preferential anterograde migration (P < 0.001). The functional effects of modest calretinin neuron ablation, particularly increased neurogenic motor activity frequencies, differ from models that incur general enteric neuron loss, and suggest calretinin neurons may contribute to pacing, force, and polarity of CMCs in the large bowel.
Collapse
Affiliation(s)
- Jing Feng
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Tim J. Hibberd
- College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Jialie Luo
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Pu Yang
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Zili Xie
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Lee Travis
- College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
| | - Nick J. Spencer
- College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Adelaide, SA, Australia
- *Correspondence: Nick J. Spencer,
| | - Hongzhen Hu
- Center for the Study of Itch and Sensory Disorders, Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, United States
- Hongzhen Hu,
| |
Collapse
|
7
|
Identifying Types of Neurons in the Human Colonic Enteric Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1383:243-249. [PMID: 36587163 DOI: 10.1007/978-3-031-05843-1_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Distinguishing and characterising the different classes of neurons that make up a neural circuit has been a long-term goal for many neuroscientists. The enteric nervous system is a large but moderately simple part of the nervous system. Enteric neurons in laboratory animals have been extensively characterised morphologically, electrophysiologically, by projections and immunohistochemically. However, studies of human enteric nervous system are less advanced despite the potential availability of tissue from elective surgery (with appropriate ethics permits). Recent studies using single cell sequencing have confirmed and extended the classification of enteric neurons in mice and human, but it is not clear whether an encompassing classification has been achieved. We present preliminary data on a means to distinguish classes of myenteric neurons in specimens of human colon combining immunohistochemical, morphological, projection and size data on single cells. A method to apply multiple layers of antisera to specimens was developed, allowing up to 12 markers to be characterised in individual neurons. Applied to multi-axonal Dogiel type II neurons, this approach demonstrated that they constitute fewer than 5% of myenteric neurons, are nearly all immunoreactive for choline acetyltransferase and tachykinins. Many express the calcium-binding proteins calbindin and calretinin and they are larger than average myenteric cells. This methodology provides a complementary approach to single-cell mRNA profiling to provide a comprehensive account of the types of myenteric neurons in the human colon.
Collapse
|
8
|
Abstract
Major advances in our understanding of the functional heterogeneity of enteric neurons are driven by the application of newly developed, innovative methods. In contrast to this progress, both animal and human enteric neurons are usually divided into only two morphological subpopulations, “Dogiel type II” neurons (with several long processes) and “Dogiel type I” neurons (with several short processes). This implies no more than the distinction of intrinsic primary afferent from all other enteric neurons. The well-known chemical and functional diversity of enteric neurons is not reflected by this restrictive dichotomy of morphological data. Recent structural investigations of human enteric neurons were performed by different groups which mainly used two methodical approaches, namely detecting the architecture of their processes and target-specific tracing of their axonal courses. Both methods were combined with multiple immunohistochemistry in order to decipher neurochemical codes. This review integrates these morphological and immunohistological data and presents a classification of human enteric neurons which we believe is not yet complete but provides an essential foundation for the further development of human gastrointestinal neuropathology.
Collapse
Affiliation(s)
- Axel Brehmer
- Institute of Anatomy and Cell Biology, Friedrich-Alexander Universität Erlangen-Nürnberg, Krankenhausstr. 9, 91054, Erlangen, Germany.
| |
Collapse
|
9
|
Nitrergic and Substance P Immunoreactive Neurons in the Enteric Nervous System of the Bottlenose Dolphin ( Tursiops truncatus) Intestine. Animals (Basel) 2021; 11:ani11041057. [PMID: 33918065 PMCID: PMC8069003 DOI: 10.3390/ani11041057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The gastrointestinal tract of the bottlenose dolphin (Tursiops truncatus) differs structurally and functionally from that of terrestrial mammals. In particular, the intestine does not show any macroscopic subdivision and lacks a caecum. In addition, the histological aspect of the intestine is relatively constant, without marked differences between the anterior and posterior parts. Although the intestine of these cetaceans presents differences in comparison with terrestrial mammals, little information is currently available on their enteric nervous system. The aim of the present study was to investigate the morphological and quantitative aspects of neurons immunoreactive (IR) for the neuronal nitric oxide synthase (nNOS) and Substance P (SP) in the intestine of bottlenose dolphins (Tursiops truncatus). In these dolphin specimens, a smaller number of nNOS-IR neurons in the submucosal plexus and a larger number of SP-IR neurons in the myenteric plexus were observed compared to other mammals. Interestingly, no co-localization between nNOS- and SP-IR neurons was detected in either of the plexuses, suggesting the existence of two completely distinct functional classes of neurons in the intestine of the bottlenose dolphin. Abstract Compared with other mammals, the digestive system of cetaceans presents some remarkable anatomical and physiological differences. However, the neurochemical features of the enteric nervous system (ENS) in these animals have only been described in part. The present study gives a description of the nitrergic and selected peptidergic systems in the myenteric plexus (MP) and submucosal plexus (SMP) of the intestine of the bottlenose dolphin (Tursiops truncatus). The distribution and morphology of neurons immunoreactive (IR) for the neuronal nitric oxide synthase (nNOS) and Substance P (SP) were immunohistochemically studied in formalin-fixed specimens from the healthy intestine of three animals, and the data were compared with those described in the literature on other mammals (human and non-human). In bottlenose dolphins, the percentages of nitrergic neurons (expressed as median and interquartile range—IQR) were 28% (IQR = 19–29) in the MP and 1% (IQR = 0–2) in the SMP, while the percentages of SP-IR neurons were 31% (IQR = 22–37) in the MP and 41% (IQR = 24–63) in the SMP. Although morphological features of nNOS- and SP-IR neurons were similar to those reported in other mammals, we found some noticeable differences in the percentages of enteric neurons. In fact, we detected a lower proportion of nNOS-IR neurons in the SMP and a higher proportion of SP-IR neurons in the MP compared to other mammals. To the best of the authors’ knowledge, this study represents the first description and quantification of nNOS-IR neurons and the first quantification of SP-IR neurons in the intestine of a cetacean species. As nNOS and SP are important mediators of intestinal functions and the nitrergic population is an important target for many neuroenteropathies, data obtained from a healthy intestine provide a necessary basis to further investigate and understand possible functional differences and motor intestinal dysfunctions/alterations in these special mammals.
Collapse
|
10
|
Wright CM, Schneider S, Smith-Edwards KM, Mafra F, Leembruggen AJL, Gonzalez MV, Kothakapa DR, Anderson JB, Maguire BA, Gao T, Missall TA, Howard MJ, Bornstein JC, Davis BM, Heuckeroth RO. scRNA-Seq Reveals New Enteric Nervous System Roles for GDNF, NRTN, and TBX3. Cell Mol Gastroenterol Hepatol 2021; 11:1548-1592.e1. [PMID: 33444816 PMCID: PMC8099699 DOI: 10.1016/j.jcmgh.2020.12.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/24/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Bowel function requires coordinated activity of diverse enteric neuron subtypes. Our aim was to define gene expression in these neuron subtypes to facilitate development of novel therapeutic approaches to treat devastating enteric neuropathies, and to learn more about enteric nervous system function. METHODS To identify subtype-specific genes, we performed single-nucleus RNA-seq on adult mouse and human colon myenteric plexus, and single-cell RNA-seq on E17.5 mouse ENS cells from whole bowel. We used immunohistochemistry, select mutant mice, and calcium imaging to validate and extend results. RESULTS RNA-seq on 635 adult mouse colon myenteric neurons and 707 E17.5 neurons from whole bowel defined seven adult neuron subtypes, eight E17.5 neuron subtypes and hundreds of differentially expressed genes. Manually dissected human colon myenteric plexus yielded RNA-seq data from 48 neurons, 3798 glia, 5568 smooth muscle, 377 interstitial cells of Cajal, and 2153 macrophages. Immunohistochemistry demonstrated differential expression for BNC2, PBX3, SATB1, RBFOX1, TBX2, and TBX3 in enteric neuron subtypes. Conditional Tbx3 loss reduced NOS1-expressing myenteric neurons. Differential Gfra1 and Gfra2 expression coupled with calcium imaging revealed that GDNF and neurturin acutely and differentially regulate activity of ∼50% of myenteric neurons with distinct effects on smooth muscle contractions. CONCLUSION Single cell analyses defined genes differentially expressed in myenteric neuron subtypes and new roles for TBX3, GDNF and NRTN. These data facilitate molecular diagnostic studies and novel therapeutics for bowel motility disorders.
Collapse
Affiliation(s)
- Christina M Wright
- Department of Pediatrics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sabine Schneider
- Department of Pediatrics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kristen M Smith-Edwards
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Fernanda Mafra
- Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | | | - Michael V Gonzalez
- Center for Applied Genomics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Deepika R Kothakapa
- Department of Pediatrics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jessica B Anderson
- Department of Pediatrics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Beth A Maguire
- Department of Pediatrics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tao Gao
- Department of Pediatrics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Tricia A Missall
- Department of Dermatology, University of Florida, Gainesville, Florida
| | - Marthe J Howard
- Department of Neurosciences, University of Toledo Health Sciences Campus, Toledo, Ohio
| | - Joel C Bornstein
- Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Brian M Davis
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Pittsburgh Center for Pain Research, University of Pittsburgh, Pittsburgh, Pennsylvania; Center for Neuroscience at the University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Robert O Heuckeroth
- Department of Pediatrics, Abramson Research Center, Children's Hospital of Philadelphia Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
11
|
Humenick A, Chen BN, Wattchow DA, Zagorodnyuk VP, Dinning PG, Spencer NJ, Costa M, Brookes SJH. Characterization of putative interneurons in the myenteric plexus of human colon. Neurogastroenterol Motil 2021; 33:e13964. [PMID: 32839997 PMCID: PMC7772282 DOI: 10.1111/nmo.13964] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/07/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND The enteric nervous system contains multiple classes of neurons, distinguishable by morphology, immunohistochemical markers, and projections; however, specific combinations differ between species. Here, types of enteric neurons in human colon were characterized immunohistochemically, using retrograde tracing combined with multiple labeling immunohistochemistry, focussing on non-motor neurons. METHODS The fluorescent carbocyanine tracer, DiI, was applied to the myenteric plexus in ex vivo preparations, filling neurons projecting within the plexus. Limits of projection lengths of motor neurons were established, allowing them to be excluded from the analysis. Long ascending and descending interneurons were then distinguished by labeling for discriminating immunohistochemical markers: calbindin, calretinin, enkephalin, 5-hydroxytryptamine, nitric oxide synthase, and substance P. These results were combined with a previous published study in which nitric oxide synthase and choline acetyltransferase immunoreactivities were established. KEY RESULTS Long ascending neurons (with projections longer than 8 mm, which excludes more than 95% motor neurons) formed four types, in descending order of abundance, defined by immunoreactivity for: (a) ChAT+/ENK+, (b) ChAT+/ENK+/SP+, (c) ChAT+/Calb+, and (d) ChAT+/ENK+/Calb+. Long descending neurons, up to 70 mm long also formed at least four types, distinguished by immunoreactivity for (a) NOS + cells (without ChAT), (b) ChAT+/NOS+, (c) ChAT+/Calret+, and (d) ChAT+/5HT + cells (with or without NOS). CONCLUSIONS AND INFERENCES Long interneurons, which do not innervate muscularis externa, are likely to coordinate neural activity over distances of many centimeters along the colon. Characterizing their neurochemical coding provides a basis for understanding their roles, investigating their connectivity, and building a comprehensive account of human colonic enteric neurons.
Collapse
Affiliation(s)
- Adam Humenick
- Human Physiology, Medical Bioscience, College of Medicine and Public Health, Flinders University, South Australia 5042
| | - Bao Nan Chen
- Human Physiology, Medical Bioscience, College of Medicine and Public Health, Flinders University, South Australia 5042
| | - David A Wattchow
- Department of Surgery, Flinders Medical Centre, Human, South Australia 5042
| | | | - Phil G Dinning
- Department of Surgery, Flinders Medical Centre, Human, South Australia 5042
| | - Nick J Spencer
- Human Physiology, Medical Bioscience, College of Medicine and Public Health, Flinders University, South Australia 5042
| | - Marcello Costa
- Human Physiology, Medical Bioscience, College of Medicine and Public Health, Flinders University, South Australia 5042
| | - Simon JH Brookes
- Human Physiology, Medical Bioscience, College of Medicine and Public Health, Flinders University, South Australia 5042
| |
Collapse
|
12
|
Morarach K, Mikhailova A, Knoflach V, Memic F, Kumar R, Li W, Ernfors P, Marklund U. Diversification of molecularly defined myenteric neuron classes revealed by single-cell RNA sequencing. Nat Neurosci 2020; 24:34-46. [PMID: 33288908 PMCID: PMC7610403 DOI: 10.1038/s41593-020-00736-x] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/07/2020] [Indexed: 12/21/2022]
Abstract
Autonomous regulation of the intestine requires the combined activity of functionally distinct neurons of the enteric nervous system (ENS). However, the variety of enteric neuron types and how they emerge during development remain largely unknown. Here, we define a molecular taxonomy of twelve enteric neuron classes within the myenteric plexus of the mouse small intestine using single cell RNA-sequencing. We present cell-cell communication features, histochemical markers for motor, sensory, and interneurons together with transgenic tools for class-specific targeting. Transcriptome analysis of embryonic ENS uncovers a novel principle of neuronal diversification, where two neuron classes arise through a binary neurogenic branching, and all other identities emerge through subsequent post-mitotic differentiation. We identify generic and class-specific transcriptional regulators and functionally connect Pbx3 to a post-mitotic fate transition. Our results offer a conceptual and molecular resource for dissecting ENS circuits, and predicting key regulators for directed differentiation of distinct enteric neuron classes.
Collapse
Affiliation(s)
- Khomgrit Morarach
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Anastassia Mikhailova
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Viktoria Knoflach
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fatima Memic
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rakesh Kumar
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.,Zoology Department, Ravenshaw University, Cuttack, India
| | - Wei Li
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ulrika Marklund
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Sodium-glucose co-transporter (SGLT) inhibitor restores lost axonal varicosities of the myenteric plexus in a mouse model of high-fat diet-induced obesity. Sci Rep 2020; 10:12372. [PMID: 32704004 PMCID: PMC7378553 DOI: 10.1038/s41598-020-69256-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 07/08/2020] [Indexed: 01/19/2023] Open
Abstract
Diabetes impairs enteric nervous system functions; however, ultrastructural changes underlying the pathophysiology of the myenteric plexus and the effects of sodium-glucose co-transporter (SGLT) inhibitors are poorly understood. This study aimed to investigate three-dimensional ultrastructural changes in axonal varicosities in the myenteric plexus and the effect thereon of the SGLT inhibitor phlorizin in mice fed a high-fat diet (HFD). Three-dimensional ultrastructural analysis using serial block-face imaging revealed that non-treated HFD-fed mice had fewer axonal varicosities and synaptic vesicles in the myenteric plexus than did normal diet-fed control mice. Furthermore, mitochondrial volume was increased and lysosome number decreased in the axons of non-treated HFD-fed mice when compared to those of control mice. Phlorizin treatment restored the axonal varicosities and organelles in HFD-fed mice. Although HFD did not affect the immunolocalisation of PGP9.5, it reduced synaptophysin immunostaining in the myenteric plexus, which was restored by phlorizin treatment. These results suggest that impairment of the axonal varicosities and their synaptic vesicles underlies the damage to the enteric neurons caused by HFD feeding. SGLT inhibitor treatment could restore axonal varicosities and organelles, which may lead to improved gastrointestinal functions in HFD-induced obesity as well as diabetes.
Collapse
|
14
|
Hung LY, Parathan P, Boonma P, Wu Q, Wang Y, Haag A, Luna RA, Bornstein JC, Savidge TC, Foong JPP. Antibiotic exposure postweaning disrupts the neurochemistry and function of enteric neurons mediating colonic motor activity. Am J Physiol Gastrointest Liver Physiol 2020; 318:G1042-G1053. [PMID: 32390463 PMCID: PMC7311661 DOI: 10.1152/ajpgi.00088.2020] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The period during and immediately after weaning is an important developmental window when marked shifts in gut microbiota can regulate the maturation of the enteric nervous system (ENS). Because microbiota-derived signals that modulate ENS development are poorly understood, we examined the physiological impact of the broad spectrum of antibiotic, vancomycin-administered postweaning on colonic motility, neurochemistry of enteric neurons, and neuronal excitability. The functional impact of vancomycin on enteric neurons was investigated by Ca2+ imaging in Wnt1-Cre;R26R-GCaMP3 reporter mice to characterize alterations in the submucosal and the myenteric plexus, which contains the neuronal circuitry controlling gut motility. 16S rDNA sequencing of fecal specimens after oral vancomycin demonstrated significant deviations in microbiota abundance, diversity, and community composition. Vancomycin significantly increased the relative family rank abundance of Akkermansiaceae, Lactobacillaceae, and Enterobacteriaceae at the expense of Lachnospiraceae and Bacteroidaceae. In sharp contrast to neonatal vancomycin exposure, microbiota compositional shifts in weaned animals were associated with slower colonic migrating motor complexes (CMMCs) without mucosal serotonin biosynthesis being altered. The slowing of CMMCs is linked to disruptions in the neurochemistry of the underlying enteric circuitry. This included significant reductions in cholinergic and calbindin+ myenteric neurons, neuronal nitric oxide synthase+ submucosal neurons, neurofilament M+ enteric neurons, and increased proportions of cholinergic submucosal neurons. The antibiotic treatment also increased transmission and responsiveness in myenteric and submucosal neurons that may enhance inhibitory motor pathways, leading to slower CMMCs. Differential vancomycin responses during neonatal and weaning periods in mice highlight the developmental-specific impact of antibiotics on colonic enteric circuitry and motility.
Collapse
Affiliation(s)
- Lin Y. Hung
- 1Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Pavitha Parathan
- 1Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Prapaporn Boonma
- 2Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,3Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas,4Faculty of Medicine, King Mongkut’s Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Qinglong Wu
- 2Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,3Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas
| | - Yi Wang
- 1Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Anthony Haag
- 2Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,3Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas
| | - Ruth Ann Luna
- 2Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,3Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas
| | - Joel C. Bornstein
- 1Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| | - Tor C. Savidge
- 2Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,3Texas Children’s Microbiome Center, Texas Children’s Hospital, Houston, Texas
| | - Jaime P. P. Foong
- 1Department of Physiology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
15
|
Smolilo DJ, Hibberd TJ, Costa M, Wattchow DA, De Fontgalland D, Spencer NJ. Intrinsic sensory neurons provide direct input to motor neurons and interneurons in mouse distal colon via varicose baskets. J Comp Neurol 2020; 528:2033-2043. [PMID: 32003462 DOI: 10.1002/cne.24872] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/22/2020] [Accepted: 01/26/2020] [Indexed: 12/20/2022]
Abstract
Connections from intrinsic primary afferent neurons (IPANs), to ascending motor and interneurons have been described in guinea pig colon. These mono- and polysynaptic circuits may underlie polarized motor reflexes evoked by local gut stimulation. There is a need to translate findings in guinea pig to mouse, a species increasingly used in enteric neuroscience. Here, mouse distal colon was immunolabeled for CGRP, a marker of putative IPANs. This revealed a combination of large, intensely immunofluorescent axons in myenteric plexus and circular muscle, and thinner varicose axons with less immunofluorescence. The latter formed dense, basket-like varicosity clusters (CGRP+ baskets) that enveloped myenteric nerve cell bodies. Immunolabeling after 4-5 days in organ culture caused loss of large CGRP+ axons, but not varicose CGRP+ fibers and CGRP+ baskets. Baskets were characterized further by triple labeling with CGRP, nitric oxide synthase (NOS) and calretinin (CALR) antibodies. Approximately half (48%) of nerve cell bodies inside CGRP+ baskets lacked both NOS and CALR, while two overlapping populations containing NOS and/or CALR comprised the remainder. Quantitative analysis revealed CGRP+ varicosities were most abundant in baskets, followed by CALR+ varicosities, with a high degree of colocalization between the two markers. Few NOS+ varicosities occurred in baskets. Significantly higher proportions of CALR+ and CGRP+ varicosities colocalized in baskets than in circular muscle. In conclusion, CGRP+ baskets in mouse colon are formed by intrinsic enteric neurons with a neurochemical profile consistent with IPANs and have direct connections to both excitatory and inhibitory neurons.
Collapse
Affiliation(s)
- David J Smolilo
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia, Australia
| | - Timothy J Hibberd
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia, Australia
| | - Marcello Costa
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia, Australia
| | - David A Wattchow
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia, Australia
| | - Dayan De Fontgalland
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia, Australia
| | - Nick J Spencer
- Visceral Neurophysiology Laboratory, College of Medicine and Public Health, Centre for Neuroscience, Flinders University, Bedford Park, South Australia, Australia
| |
Collapse
|
16
|
Costa M, Hibberd TJ, Keightley LJ, Wiklendt L, Arkwright JW, Dinning PG, Brookes SJH, Spencer NJ. Neural motor complexes propagate continuously along the full length of mouse small intestine and colon. Am J Physiol Gastrointest Liver Physiol 2020; 318:G99-G108. [PMID: 31709829 DOI: 10.1152/ajpgi.00185.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cyclical propagating waves of muscle contraction have been recorded in isolated small intestine or colon, referred to here as motor complexes (MCs). Small intestinal and colonic MCs are neurogenic, occur at similar frequencies, and propagate orally or aborally. Whether they can be coordinated between the different gut regions is unclear. Motor behavior of whole length mouse intestines, from duodenum to terminal rectum, was recorded by intraluminal multisensor catheter. Small intestinal MCs were recorded in 27/30 preparations, and colonic MCs were recorded in all preparations (n = 30) with similar frequencies (0.54 ± 0.03 and 0.58 ± 0.02 counts/min, respectively). MCs propagated across the ileo-colonic junction in 10/30 preparations, forming "full intestine" MCs. The cholinesterase inhibitor physostigmine increased the probability of a full intestine MC but had no significant effect on frequency, speed, or direction. Nitric oxide synthesis blockade by Nω-nitro-l-arginine, after physostigmine, increased MC frequency in small intestine only. Hyoscine-resistant MCs were recorded in the colon but not small intestine (n = 5). All MCs were abolished by hexamethonium (n = 18) or tetrodotoxin (n = 2). The enteric neural mechanism required for motor complexes is present along the full length of both the small and large intestine. In some cases, colonic MCs can be initiated in the distal colon and propagate through the ileo-colonic junction, all the way to duodenum. In conclusion, the ileo-colonic junction provides functional neural continuity for propagating motor activity that originates in the small or large intestine.NEW & NOTEWORTHY Intraluminal manometric recordings revealed motor complexes can propagate antegradely or retrogradely across the ileo-colonic junction, spanning the entire small and large intestines. The fundamental enteric neural mechanism(s) underlying cyclic motor complexes exists throughout the length of the small and large intestine.
Collapse
Affiliation(s)
- Marcello Costa
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Timothy James Hibberd
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Lauren J Keightley
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Lukasz Wiklendt
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - John W Arkwright
- Computer Science, Engineering and Mathematics, Flinders University, Adelaide, South Australia, Australia
| | - Philip G Dinning
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia.,Department of Gastroenterology and Surgery, Flinders Medical Centre, Flinders University, Adelaide, South Australia, Australia
| | - Simon J H Brookes
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Nick J Spencer
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
17
|
Swaminathan M, Fung C, Finkelstein DI, Bornstein JC, Foong JPP. α-Synuclein Regulates Development and Function of Cholinergic Enteric Neurons in the Mouse Colon. Neuroscience 2019; 423:76-85. [PMID: 31705886 DOI: 10.1016/j.neuroscience.2019.10.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 10/15/2019] [Accepted: 10/16/2019] [Indexed: 02/07/2023]
Abstract
Alpha-Synuclein (α-Syn) is expressed in the central nervous system and the nervous system of the gut (enteric nervous system, ENS), and is well known to be the major constituent of Lewy bodies which are the hallmark of Parkinson's disease. Gastrointestinal disorders frequently manifest several years before motor deficits develop in Parkinson's patients. Despite extensive research on pathological rodent models, the physiological role of α-Syn in the normal ENS is unclear hampering analysis of its neuropathology. We compared the ENS in colons of α-Syn-knockout (α-Syn KO) and wild-type mice using immunohistochemistry and calcium-imaging of responses to synaptic input. We found that α-Syn is predominantly expressed in cholinergic varicosities, which contain vesicular acetylcholine transporter. α-Syn KO mice had higher enteric neuron density and a larger proportion of cholinergic neurons, notably those containing calretinin, demonstrating a role for α-Syn in regulating development of these neurons. Moreover, α-Syn deletion enhanced the amplitude of synaptically activated [Ca2+]i transients that are primarily mediated by acetylcholine activating nicotinic receptors suggesting that α-Syn modulates the availability of acetylcholine in enteric nerve terminals.
Collapse
Affiliation(s)
- M Swaminathan
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - C Fung
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - D I Finkelstein
- The Florey Institute of Neuroscience and Mental Health, Kenneth Myer Building, The University of Melbourne, Parkville, VIC, Australia
| | - J C Bornstein
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - J P P Foong
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
18
|
Swaminathan M, Hill-Yardin EL, Bornstein JC, Foong JPP. Endogenous Glutamate Excites Myenteric Calbindin Neurons by Activating Group I Metabotropic Glutamate Receptors in the Mouse Colon. Front Neurosci 2019; 13:426. [PMID: 31118881 PMCID: PMC6504831 DOI: 10.3389/fnins.2019.00426] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/15/2019] [Indexed: 12/20/2022] Open
Abstract
Glutamate is a classic excitatory neurotransmitter in the central nervous system (CNS), but despite several studies reporting the expression of glutamate together with its various receptors and transporters within the enteric nervous system (ENS), its role in the gut remains elusive. In this study, we characterized the expression of the vesicular glutamate transporter, vGluT2, and examined the function of glutamate in the myenteric plexus of the distal colon by employing calcium (Ca2+)-imaging on Wnt1-Cre; R26R-GCaMP3 mice which express a genetically encoded fluorescent Ca2+ indicator in all enteric neurons and glia. Most vGluT2 labeled varicosities contained the synaptic vesicle release protein, synaptophysin, but not vesicular acetylcholine transporter, vAChT, which labels vesicles containing acetylcholine, the primary excitatory neurotransmitter in the ENS. The somata of all calbindin (calb) immunoreactive neurons examined received close contacts from vGluT2 varicosities, which were more numerous than those contacting nitrergic neurons. Exogenous application of L-glutamic acid (L-Glu) and N-methyl-D-aspartate (NMDA) transiently increased the intracellular Ca2+ concentration [Ca2+]i in about 25% of myenteric neurons. Most L-Glu responsive neurons were calb immunoreactive. Blockade of NMDA receptors with APV significantly reduced the number of neurons responsive to L-Glu and NMDA, thus showing functional expression of NMDA receptors on enteric neurons. However, APV resistant responses to L-Glu and NMDA suggest that other glutamate receptors were present. APV did not affect [Ca2+]i transients evoked by electrical stimulation of interganglionic nerve fiber tracts, which suggests that NMDA receptors are not involved in synaptic transmission. The group I metabotropic glutamate receptor (mGluR) antagonist, PHCCC, significantly reduced the amplitude of [Ca2+]i transients evoked by a 20 pulse (20 Hz) train of electrical stimuli in L-Glu responsive neurons. This stimulus is known to induce slow synaptic depolarizations. Further, some neurons that had PHCCC sensitive [Ca2+]i transients were calb immunoreactive and received vGluT2 varicosities. Overall, we conclude that electrically evoked release of endogenous glutamate mediates slow synaptic transmission via activation of group I mGluRs expressed by myenteric neurons, particularly those immunoreactive for calb.
Collapse
Affiliation(s)
- Mathusi Swaminathan
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Elisa L Hill-Yardin
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia.,School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Joel C Bornstein
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| | - Jaime P P Foong
- Department of Physiology, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
19
|
Hibberd TJ, Feng J, Luo J, Yang P, Samineni VK, Gereau RW, Kelley N, Hu H, Spencer NJ. Optogenetic Induction of Colonic Motility in Mice. Gastroenterology 2018; 155:514-528.e6. [PMID: 29782847 PMCID: PMC6715392 DOI: 10.1053/j.gastro.2018.05.029] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 04/12/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND & AIMS Strategies are needed to increase gastrointestinal transit without systemic pharmacologic agents. We investigated whether optogenetics, focal application of light to control enteric nervous system excitability, could be used to evoke propagating contractions and increase colonic transit in mice. METHODS We generated transgenic mice with Cre-mediated expression of light-sensitive channelrhodopsin-2 (ChR2) in calretinin neurons (CAL-ChR2 Cre+ mice); Cre- littermates served as controls. Colonic myenteric neurons were analyzed by immunohistochemistry, patch-clamp, and calcium imaging studies. Motility was assessed by mechanical, electrophysiological, and video recording in vitro and by fecal output in vivo. RESULTS In isolated colons, focal light stimulation of calretinin enteric neurons evoked classic polarized motor reflexes (50/58 stimulations), followed by premature anterograde propagating contractions (39/58 stimulations). Light stimulation could evoke motility from sites along the entire colon. These effects were prevented by neural blockade with tetrodotoxin (n = 2), and did not occur in control mice (n = 5). Light stimulation of proximal colon increased the proportion of natural fecal pellets expelled over 15 minutes in vitro (75% ± 17% vs 32% ± 8% for controls) (P < .05). In vivo, activation of wireless light-emitting diodes implanted onto the colon wall significantly increased hourly fecal pellet output in conscious, freely moving mice (4.2 ± 0.4 vs 1.3 ± 0.3 in controls) (P < .001). CONCLUSIONS In studies of mice, we found that focal activation of a subset of enteric neurons can increase motility of the entire colon in vitro, and fecal output in vivo. Optogenetic control of enteric neurons might therefore be used to modify gut motility.
Collapse
Affiliation(s)
- Timothy J Hibberd
- College of Medicine and Public Health & Centre for Neuroscience, Flinders University, Australia
| | - Jing Feng
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| | - Jialie Luo
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| | - Pu Yang
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St. Louis, MO, USA
| | - Vijay K Samineni
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Robert W Gereau
- Washington University Pain Center, Department of Anesthesiology, Washington University School of Medicine, St Louis, MO, USA
| | - Nigel Kelley
- SA Biomedical Engineering, SA Health, Government of South Australia, Australia
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch, Washington University School of Medicine, St Louis, Missouri.
| | - Nick J Spencer
- College of Medicine and Public Health and Centre for Neuroscience, Flinders University, Adelaide, Australia.
| |
Collapse
|
20
|
Hibberd TJ, Travis L, Wiklendt L, Costa M, Brookes SJH, Hu H, Keating DJ, Spencer NJ. Synaptic activation of putative sensory neurons by hexamethonium-sensitive nerve pathways in mouse colon. Am J Physiol Gastrointest Liver Physiol 2018; 314:G53-G64. [PMID: 28935683 DOI: 10.1152/ajpgi.00234.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastrointestinal tract contains its own independent population of sensory neurons within the gut wall. These sensory neurons have been referred to as intrinsic primary afferent neurons (IPANs) and can be identified by immunoreactivity to calcitonin gene-related peptide (CGRP) in mice. A common feature of IPANs is a paucity of fast synaptic inputs observed during sharp microelectrode recordings. Whether this is observed using different recording techniques is of particular interest for understanding the physiology of these neurons and neural circuit modeling. Here, we imaged spontaneous and evoked activation of myenteric neurons in isolated whole preparations of mouse colon and correlated recordings with CGRP and nitric oxide synthase (NOS) immunoreactivity, post hoc. Calcium indicator fluo 4 was used for this purpose. Calcium responses were recorded in nerve cell bodies located 5-10 mm oral to transmural electrical nerve stimuli. A total of 618 recorded neurons were classified for CGRP or NOS immunoreactivity. Aboral electrical stimulation evoked short-latency calcium transients in the majority of myenteric neurons, including ~90% of CGRP-immunoreactive Dogiel type II neurons. Activation of Dogiel type II neurons had a time course consistent with fast synaptic transmission and was always abolished by hexamethonium (300 μM) and by low-calcium Krebs solution. The nicotinic receptor agonist 1,1-dimethyl-4-phenylpiperazinium iodide (during synaptic blockade) directly activated Dogiel type II neurons. The present study suggests that murine colonic Dogiel type II neurons receive prominent fast excitatory synaptic inputs from hexamethonium-sensitive neural pathways. NEW & NOTEWORTHY Myenteric neurons in isolated mouse colon were recorded using calcium imaging and then neurochemically defined. Short-latency calcium transients were detected in >90% of calcitonin gene-related peptide-immunoreactive neurons to electrical stimulation of hexamethonium-sensitive pathways. Putative sensory Dogiel type II calcitonin gene-related peptide-immunoreactive myenteric neurons may receive widespread fast synaptic inputs in mouse colon.
Collapse
Affiliation(s)
- Timothy J Hibberd
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University , Adelaide South Australia
| | - Lee Travis
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University , Adelaide South Australia
| | - Lukasz Wiklendt
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University , Adelaide South Australia
| | - Marcello Costa
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University , Adelaide South Australia
| | - Simon J H Brookes
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University , Adelaide South Australia
| | - Hongzhen Hu
- Department of Anesthesiology, Washington University , Saint Louis, Missouri
| | - Damien J Keating
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University , Adelaide South Australia
| | - Nick J Spencer
- Discipline of Human Physiology and Centre for Neuroscience, Flinders University , Adelaide South Australia
| |
Collapse
|
21
|
Dong Y, Wang Z, Qin Z, Cao J, Chen Y. Role of serotonin in the intestinal mucosal epithelium barrier in weaning mice undergoing stress-induced diarrhea. J Mol Histol 2017; 49:85-97. [PMID: 29260435 DOI: 10.1007/s10735-017-9749-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/05/2017] [Indexed: 12/17/2022]
Abstract
Stress-induced diarrhea is a frequent and challenging threat to humans and domestic animals. Serotonin (5-HT) has been shown to be involved in the pathological process of stress-induced diarrhea. However, the role of 5-HT in stress-induced diarrhea remains unclear. A stress-induced diarrhea model was established in 21-day-old ICR weaning mice through an intragastric administration of 0.25 mL of 0.4 g/mL folium sennae and restraint of the hind legs with adhesive tape for 4 h to determine whether 5-HT regulates the mucosal barrier to cause diarrhea. Mice with decreased levels of 5-HT were pretreated with an intraperitoneal injection of 300 mg/kg p-chlorophenylalanine (PCPA), a 5-HT synthesis inhibitor. After 5 days of treatment, the stress level, body weight and intestinal mucosal morphology indexes were measured. Compared to the controls, the mice with stress-induced diarrhea displayed a stress reaction, with increased corticosterone levels, as well as increased 5-HT-positive cells. However, the mice with stress-induced diarrhea exhibited decreased body weights, villus height to crypt depth ratios (V/C), and Occludin and Claudin1 expression. The PCPA injection reversed these effects in mice with different degrees of stress-induced diarrhea. Based on these findings, inhibition of 5-HT synthesis relieved the stress response and improved the health of the intestinal tract, including both the intestinal absorption capacity, as determined by the villus height and crypt depth, and the mucosal barrier function, as determined by the tight junction proteins of epithelial cell.
Collapse
Affiliation(s)
- Yulan Dong
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China
| | - Zixu Wang
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China
| | - Zhuoming Qin
- Institute of Poultry, Shandong Academy of Agricultural Sciences, Jinan, 250100, People's Republic of China
| | - Jing Cao
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China
| | - Yaoxing Chen
- Laboratory of Veterinary Anatomy, College of Animal Medicine, China Agricultural University, Haidian, Beijing, 100193, People's Republic of China.
| |
Collapse
|
22
|
Fung C, Boesmans W, Cirillo C, Foong JPP, Bornstein JC, Vanden Berghe P. VPAC Receptor Subtypes Tune Purinergic Neuron-to-Glia Communication in the Murine Submucosal Plexus. Front Cell Neurosci 2017; 11:118. [PMID: 28487635 PMCID: PMC5403822 DOI: 10.3389/fncel.2017.00118] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 04/10/2017] [Indexed: 12/20/2022] Open
Abstract
The enteric nervous system (ENS) situated within the gastrointestinal tract comprises an intricate network of neurons and glia which together regulate intestinal function. The exact neuro-glial circuitry and the signaling molecules involved are yet to be fully elucidated. Vasoactive intestinal peptide (VIP) is one of the main neurotransmitters in the gut, and is important for regulating intestinal secretion and motility. However, the role of VIP and its VPAC receptors within the enteric circuitry is not well understood. We investigated this in the submucosal plexus of mouse jejunum using calcium (Ca2+)-imaging. Local VIP application induced Ca2+-transients primarily in neurons and these were inhibited by VPAC1- and VPAC2-antagonists (PG 99-269 and PG 99-465 respectively). These VIP-evoked neural Ca2+-transients were also inhibited by tetrodotoxin (TTX), indicating that they were secondary to action potential generation. Surprisingly, VIP induced Ca2+-transients in glia in the presence of the VPAC2 antagonist. Further, selective VPAC1 receptor activation with the agonist ([K15, R16, L27]VIP(1-7)/GRF(8-27)) predominantly evoked glial responses. However, VPAC1-immunoreactivity did not colocalize with the glial marker glial fibrillary acidic protein (GFAP). Rather, VPAC1 expression was found on cholinergic submucosal neurons and nerve fibers. This suggests that glial responses observed were secondary to neuronal activation. Trains of electrical stimuli were applied to fiber tracts to induce endogenous VIP release. Delayed glial responses were evoked when the VPAC2 antagonist was present. These findings support the presence of an intrinsic VIP/VPAC-initiated neuron-to-glia signaling pathway. VPAC1 agonist-evoked glial responses were inhibited by purinergic antagonists (PPADS and MRS2179), thus demonstrating the involvement of P2Y1 receptors. Collectively, we showed that neurally-released VIP can activate neurons expressing VPAC1 and/or VPAC2 receptors to modulate purine-release onto glia. Selective VPAC1 activation evokes a glial response, whereas VPAC2 receptors may act to inhibit this response. Thus, we identified a component of an enteric neuron-glia circuit that is fine-tuned by endogenous VIP acting through VPAC1- and VPAC2-mediated pathways.
Collapse
Affiliation(s)
- Candice Fung
- Department of Physiology, The University of MelbourneParkville, VIC, Australia.,Laboratory for Enteric Neuroscience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU LeuvenLeuven, Belgium
| | - Werend Boesmans
- Laboratory for Enteric Neuroscience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU LeuvenLeuven, Belgium
| | - Carla Cirillo
- Laboratory for Enteric Neuroscience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU LeuvenLeuven, Belgium
| | - Jaime P P Foong
- Department of Physiology, The University of MelbourneParkville, VIC, Australia
| | - Joel C Bornstein
- Department of Physiology, The University of MelbourneParkville, VIC, Australia
| | - Pieter Vanden Berghe
- Laboratory for Enteric Neuroscience (LENS), Translational Research Center for Gastrointestinal Disorders (TARGID), KU LeuvenLeuven, Belgium
| |
Collapse
|
23
|
Visualizing the enteric nervous system using genetically engineered double reporter mice: Comparison with immunofluorescence. PLoS One 2017; 12:e0171239. [PMID: 28158225 PMCID: PMC5291392 DOI: 10.1371/journal.pone.0171239] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 01/17/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND AIMS The enteric nervous system (ENS) plays a crucial role in the control of gastrointestinal motility, secretion and absorption functions. Immunohistochemistry has been widely used to visualize neurons of the ENS for more than two decades. Genetically engineered mice that report specific proteins can also be used to visualize neurons of the ENS. The goal of our study was to develop a mouse that expresses fluorescent neuronal nitric oxide synthase (nNOS) and choline acetyltransferase (ChAT), the two proteins expressed in 95% of the ENS neurons. We compared ENS neurons visualized in the reporter mouse with the wild type mouse stained using classical immunostaining techniques. METHODS Mice hemizygous for ChAT-ChR2-YFP BAC transgene with expression of the mhChR2:YFP fusion protein directed by ChAT promoter/enhancer regions on the BAC transgene were purchased commercially. The Cre/LoxP technique of somatic recombination was used to construct mice with nNOS positive neurons. The two mice were crossbred and tissues were harvested and examined using fluorescent microscopy. Immunostaining was performed in the wild type mice, using antibodies to nNOS, ChAT, Hu and PGP 9.5. RESULTS Greater than 95% of the ENS neurons were positive for either nNOS or ChAT or both. The nNOS and ChAT neurons and their processes in the ENS were well visualized in all the regions of the GI tract, i.e., esophagus, small intestine and colon. The number of nNOS and ChAT neurons was approximately same in the reporter mouse and immunostaining method in the wild type mouse. The nNOS fluorescence in the reporter mouse was seen in both cytoplasm as well as nucleus but in the immunostained specimens it was seen only in the cytoplasm. CONCLUSION We propose that the genetically engineered double reporter mouse for ChAT and nNOS proteins is a powerful tool to study of the effects of various diseases on the ENS without the need for immunostaining.
Collapse
|
24
|
Ascl1 Is Required for the Development of Specific Neuronal Subtypes in the Enteric Nervous System. J Neurosci 2016; 36:4339-50. [PMID: 27076429 DOI: 10.1523/jneurosci.0202-16.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 02/25/2016] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED The enteric nervous system (ENS) is organized into neural circuits within the gastrointestinal wall where it controls the peristaltic movements, secretion, and blood flow. Although proper gut function relies on the complex neuronal composition of the ENS, little is known about the transcriptional networks that regulate the diversification into different classes of enteric neurons and glia during development. Here we redefine the role of Ascl1 (Mash1), one of the few regulatory transcription factors described during ENS development. We show that enteric glia and all enteric neuronal subtypes appear to be derived from Ascl1-expressing progenitor cells. In the gut of Ascl1(-/-) mutant mice, neurogenesis is delayed and reduced, and posterior gliogenesis impaired. The ratio of neurons expressing Calbindin, TH, and VIP is selectively decreased while, for instance, 5-HT(+) neurons, which previously were believed to be Ascl1-dependent, are formed in normal numbers. Essentially the same differentiation defects are observed in Ascl1(KINgn2) transgenic mutants, where the proneural activity of Ngn2 replaces Ascl1, demonstrating that Ascl1 is required for the acquisition of specific enteric neuronal subtype features independent of its role in neurogenesis. In this study, we provide novel insights into the expression and function of Ascl1 in the differentiation process of specific neuronal subtypes during ENS development. SIGNIFICANCE STATEMENT The molecular mechanisms underlying the generation of different neuronal subtypes during development of the enteric nervous system are poorly understood despite its pivotal function in gut motility and involvement in gastrointestinal pathology. This report identifies novel roles for the transcription factor Ascl1 in enteric gliogenesis and neurogenesis. Moreover, independent of its proneurogenic activity, Ascl1 is required for the normal expression of specific enteric neuronal subtype characteristics. Distinct enteric neuronal subtypes are formed in a temporally defined order, and we observe that the early-born 5-HT(+) neurons are generated in Ascl1(-/-) mutants, despite the delayed neurogenesis. Enteric nervous system progenitor cells may therefore possess strong intrinsic control over their specification at the initial waves of neurogenesis.
Collapse
|
25
|
Neuronal Differentiation in Schwann Cell Lineage Underlies Postnatal Neurogenesis in the Enteric Nervous System. J Neurosci 2015; 35:9879-88. [PMID: 26156989 DOI: 10.1523/jneurosci.1239-15.2015] [Citation(s) in RCA: 167] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Elucidation of the cellular identity of neuronal precursors provides mechanistic insights into the development and pathophysiology of the nervous system. In the enteric nervous system (ENS), neurogenesis persists from midgestation to the postnatal period. Cellular mechanism underlying the long-term neurogenesis in the ENS has remained unclear. Using genetic fate mapping in mice, we show here that a subset of Schwann cell precursors (SCPs), which invades the gut alongside the extrinsic nerves, adopts a neuronal fate in the postnatal period and contributes to the ENS. We found SCP-derived neurogenesis in the submucosal region of the small intestine in the absence of vagal neural crest-derived ENS precursors. Under physiological conditions, SCPs comprised up to 20% of enteric neurons in the large intestine and gave rise mainly to restricted neuronal subtypes, calretinin-expressing neurons. Genetic ablation of Ret, the signaling receptor for glial cell line-derived neurotrophic factor, in SCPs caused colonic oligoganglionosis, indicating that SCP-derived neurogenesis is essential to ENS integrity. Identification of Schwann cells as a physiological neurogenic source provides novel insight into the development and disorders of neural crest-derived tissues. SIGNIFICANCE STATEMENT Elucidating the cellular identity of neuronal precursors provides novel insights into development and function of the nervous system. The enteric nervous system (ENS) is innervated richly by extrinsic nerve fibers, but little is known about the significance of extrinsic innervation to the structural integrity of the ENS. This report reveals that a subset of Schwann cell precursors (SCPs), which invades the gut alongside the extrinsic nerves, adopts a neuronal fate and differentiates into specific neuronal subtypes. SCP-specific ablation of the Ret gene leads to colonic oligoganglionosis, demonstrating a crucial role of SCP-derived neurogenesis in ENS development. Cross-lineage differentiation capacity in SCPs suggests their potential involvement in the development and pathology of a wide variety of neural crest-derived cell types.
Collapse
|
26
|
Stenkamp-Strahm CM, Nyavor YEA, Kappmeyer AJ, Horton S, Gericke M, Balemba OB. Prolonged high fat diet ingestion, obesity, and type 2 diabetes symptoms correlate with phenotypic plasticity in myenteric neurons and nerve damage in the mouse duodenum. Cell Tissue Res 2015; 361:411-26. [PMID: 25722087 DOI: 10.1007/s00441-015-2132-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Accepted: 01/20/2015] [Indexed: 12/18/2022]
Abstract
Symptoms of diabetic gastrointestinal dysmotility indicate neuropathy of the enteric nervous system. Long-standing diabetic enteric neuropathy has not been fully characterized, however. We used prolonged high fat diet ingestion (20 weeks) in a mouse model to mimic human obese and type 2 diabetic conditions, and analyzed changes seen in neurons of the duodenal myenteric plexus. Ganglionic and neuronal size, number of neurons per ganglionic area, density indices of neuronal phenotypes (immunoreactive nerve cell bodies and varicosities per ganglion or tissue area) and nerve injury were measured. Findings were compared with results previously seen in mice fed the same diet for 8 weeks. Compared to mice fed standard chow, those on a prolonged high fat diet had smaller ganglionic and cell soma areas. Myenteric VIP- and ChAT-immunoreactive density indices were also reduced. Myenteric nerve fibers were markedly swollen and cytoskeletal protein networks were disrupted. The number of nNOS nerve cell bodies per ganglia was increased, contrary to the reduction previously seen after 8 weeks, but the density index of nNOS varicosities was reduced. Mice fed high fat and standard chow diets experienced an age-related reduction in total neurons, with bias towards neurons of sensory phenotype. Meanwhile, ageing was associated with an increase in excitatory neuronal markers. Collectively, these results support a notion that nerve damage underlies diabetic symptoms of dysmotility, and reveals adaptive ENS responses to the prolonged ingestion of a high fat diet. This highlights a need to mechanistically study long-term diet-induced nerve damage and age-related impacts on the ENS.
Collapse
Affiliation(s)
- Chloe M Stenkamp-Strahm
- Department of Biological Sciences, University of Idaho, 875 Perimeter Drive 3051, LSS 252, Moscow, ID, 83844-3051, USA
| | | | | | | | | | | |
Collapse
|
27
|
Avetisyan M, Schill EM, Heuckeroth RO. Building a second brain in the bowel. J Clin Invest 2015; 125:899-907. [PMID: 25664848 DOI: 10.1172/jci76307] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The enteric nervous system (ENS) is sometimes called the "second brain" because of the diversity of neuronal cell types and complex, integrated circuits that permit the ENS to autonomously regulate many processes in the bowel. Mechanisms supporting ENS development are intricate, with numerous proteins, small molecules, and nutrients that affect ENS morphogenesis and mature function. Damage to the ENS or developmental defects cause vomiting, abdominal pain, constipation, growth failure, and early death. Here, we review molecular mechanisms and cellular processes that govern ENS development, identify areas in which more investigation is needed, and discuss the clinical implications of new basic research.
Collapse
|
28
|
Smith TK, Park KJ, Hennig GW. Colonic migrating motor complexes, high amplitude propagating contractions, neural reflexes and the importance of neuronal and mucosal serotonin. J Neurogastroenterol Motil 2014; 20:423-46. [PMID: 25273115 PMCID: PMC4204412 DOI: 10.5056/jnm14092] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 09/05/2014] [Accepted: 09/08/2014] [Indexed: 12/15/2022] Open
Abstract
The colonic migrating motor complex (CMMC) is a critical neurally mediated rhythmic propulsive contraction observed in the large intestine of many mammals. It seems to be equivalent to the high amplitude propagating contractions (HAPCs) in humans. This review focuses on the probable neural mechanisms involved in producing the CMMC or HAPC, their likely dependence on mucosal and neuronal serotonin and pacemaker insterstitial cells of Cajal networks and how intrinsic neural reflexes affect them. Discussed is the possibility that myenteric 5-hydroxytryptamine (5-HT) neurons are not only involved in tonic inhibition of the colon, but are also involved in generating the CMMC and modulation of the entire enteric nervous system, including coupling motility to secretion and blood flow. Mucosal 5-HT appears to be important for the initiation and effective propagation of CMMCs, although this mechanism is a longstanding controversy since the 1950s, which we will address. We argue that the slow apparent propagation of the CMMC/HAPC down the colon is unlikely to result from a slowly conducting wave front of neural activity, but more likely because of an interaction between ascending excitatory and descending (serotonergic) inhibitory neural pathways interacting both within the myenteric plexus and at the level of the muscle. That is, CMMC/HAPC propagation appears to be similar to esophageal peristalsis. The suppression of inhibitory (neuronal nitric oxide synthase) motor neurons and mucosal 5-HT release by an upregulation of prostaglandins has important implications in a number of gastrointestinal disorders, especially slow transit constipation.
Collapse
Affiliation(s)
- Terence K Smith
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Kyu Joo Park
- Department of Surgery, School of Medicine, Seoul National University, Seoul Korea
| | - Grant W Hennig
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| |
Collapse
|
29
|
Bergner AJ, Stamp LA, Gonsalvez DG, Allison MB, Olson DP, Myers MG, Anderson CR, Young HM. Birthdating of myenteric neuron subtypes in the small intestine of the mouse. J Comp Neurol 2014; 522:514-27. [PMID: 23861145 DOI: 10.1002/cne.23423] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 06/26/2013] [Accepted: 07/03/2013] [Indexed: 12/31/2022]
Abstract
There are many different types of enteric neurons. Previous studies have identified the time at which some enteric neuron subtypes are born (exit the cell cycle) in the mouse, but the birthdates of some major enteric neuron subtypes are still incompletely characterized or unknown. We combined 5-ethynynl-2'-deoxyuridine (EdU) labeling with antibody markers that identify myenteric neuron subtypes to determine when neuron subtypes are born in the mouse small intestine. We found that different neurochemical classes of enteric neuron differed in their birthdates; serotonin neurons were born first with peak cell cycle exit at E11.5, followed by neurofilament-M neurons, calcitonin gene-related peptide neurons (peak cell cycle exit for both at embryonic day [E]12.5-E13.5), tyrosine hydroxylase neurons (E15.5), nitric oxide synthase 1 (NOS1) neurons (E15.5), and calretinin neurons (postnatal day [P]0). The vast majority of myenteric neurons had exited the cell cycle by P10. We did not observe any EdU+/NOS1+ myenteric neurons in the small intestine of adult mice following EdU injection at E10.5 or E11.5, which was unexpected, as previous studies have shown that NOS1 neurons are present in E11.5 mice. Studies using the proliferation marker Ki67 revealed that very few NOS1 neurons in the E11.5 and E12.5 gut were proliferating. However, Cre-lox-based genetic fate-mapping revealed a small subpopulation of myenteric neurons that appears to express NOS1 only transiently. Together, our results confirm a relationship between enteric neuron subtype and birthdate, and suggest that some enteric neurons exhibit neurochemical phenotypes during development that are different from their mature phenotype.
Collapse
Affiliation(s)
- Annette J Bergner
- Department of Anatomy & Neuroscience, University of Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Localisation and activation of the neurokinin 1 receptor in the enteric nervous system of the mouse distal colon. Cell Tissue Res 2014; 356:319-32. [PMID: 24728885 DOI: 10.1007/s00441-014-1822-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 01/20/2014] [Indexed: 12/31/2022]
Abstract
The substance P neurokinin 1 receptor (NK1R) regulates motility, secretion, inflammation and pain in the intestine. The distribution of the NK1R is a key determinant of the functional effects of substance P in the gut. Information regarding the distribution of NK1R in subtypes of mouse enteric neurons is lacking and is the focus of the present study. NK1R immunoreactivity (NK1R-IR) is examined in whole-mount preparations of the mouse distal colon by indirect immunofluorescence and confocal microscopy. The distribution of NK1R-IR within key functional neuronal subclasses was determined by using established neurochemical markers. NK1R-IR was expressed by a subpopulation of myenteric and submucosal neurons; it was mainly detected in large multipolar myenteric neurons and was colocalized with calcitonin gene-related peptide, neurofilament M, choline acetyltransferase and calretinin. The remaining NK1R-immunoreactive neurons were positive for nitric oxide synthase. NK1R was expressed by most of the submucosal neurons and was exclusively co-expressed with vasoactive intestinal peptide, with no overlap with choline acetyltransferase. Treatment with substance P resulted in the concentration-dependent internalisation of NK1R from the cell surface into endosome-like structures. Myenteric NK1R was mainly expressed by intrinsic primary afferent neurons, with minor expression by descending interneurons and inhibitory motor neurons. Submucosal NK1R was restricted to non-cholinergic secretomotor neurons. These findings highlight key differences in the neuronal distribution of NK1R-IR between the mouse, rat and guinea-pig, with important implications for the functional role of NK1R in regulating intestinal motility and secretion.
Collapse
|
31
|
Okamoto T, Barton MJ, Hennig GW, Birch GC, Grainger N, Corrigan RD, Koh SD, Sanders KM, Smith TK. Extensive projections of myenteric serotonergic neurons suggest they comprise the central processing unit in the colon. Neurogastroenterol Motil 2014; 26:556-70. [PMID: 24460867 DOI: 10.1111/nmo.12302] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/12/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND 5-Hydroxytryptamine (5-HT, serotonin) is an important regulator of colonic motility and secretion; yet the role of serotonergic neurons in the colon is controversial. METHODS We used immunohistochemical techniques to examine their projections throughout the enteric nervous system and interstitial cells of Cajal (ICC) networks in the murine proximal to mid colon. KEY RESULTS Serotonergic neurons, which were mainly calbindin positive, occurred only in myenteric ganglia (1 per 3 ganglia). They were larger than nNOS neurons but similar in size to Dogiel Type II (AH) neurons. 5-HT neurons, appeared to make numerous varicose contacts with each other, most nNOS neurons, Dogiel Type II/AH neurons and glial cells. 5-HT, calbindin and nNOS nerve fibers also formed a thin perimuscular nerve plexus that was associated with ganglia, which contained both nNOS positive and negative neurons, which lay directly upon the submucosal pacemaker ICC network. Neurons in perimuscular ganglia were surrounded by 5-HT varicosities. Submucous ganglia contained nNOS positive and negative neurons, and calbindin positive neurons, which also appeared richly supplied by serotonergic nerve varicosities. Serotonergic nerve fibers ran along submucosal arterioles, but not veins. Varicosities of serotonergic nerve fibers were closely associated with pacemaker ICC networks and with intramuscular ICC (ICC-IM). 5-HT2B receptors were found on a subpopulation of non-5-HT containing myenteric neurons and their varicosities, pacemaker ICC-MY and ICC-IM. CONCLUSIONS & INFERENCES Myenteric serotonergic neurons, whose axons exhibit considerable divergence, regulate the entire enteric nervous system and are important in coordinating motility with secretion. They are not just interneurons, as regularly assumed, but possibly also motor neurons to ICC and blood vessels, and some may even be sensory neurons.
Collapse
Affiliation(s)
- T Okamoto
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang Y, Bitner D, Pontes Filho AA, Li F, Liu S, Wang H, Yang F, Adhikari S, Gordon J, Srinivasan S, Hu W. Expression and function of NIK- and IKK2-binding protein (NIBP) in mouse enteric nervous system. Neurogastroenterol Motil 2014; 26:77-97. [PMID: 24011459 PMCID: PMC3962790 DOI: 10.1111/nmo.12234] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 08/15/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND NIK- and IKK2-binding protein (NIBP)/TRAPPC9 is expressed in brain neurons, and human NIBP mutations are associated with neurodevelopmental disorders. The cellular distribution and function of NIBP in the enteric nervous system (ENS) remain unknown. METHODS Western blot and reverse transcription-polymerase chain reaction analysis were used respectively to identify the protein and mRNA expression of NIBP and other neuronal markers. Multi-labeled immunofluorescent microscopy and confocal image analysis were used to examine the cellular distribution of NIBP-like immunoreactivity (IR) in whole mount intestine. Enteric neuronal cell line (ENC) was infected with lentivirus carrying NIBP or its shRNA expression vectors and treated with vehicle or tumor necrosis factor (TNF)α. KEY RESULTS NIBP is expressed at both mRNA and protein levels in different regions and layers of the mouse intestine. NIBP-like-IR was co-localized with various neuronal markers, but not with glial, smooth muscular, or interstitial cells of Cajal markers. A small population of NIBP-expressing cells and fibers in extra-ganglionic and intra-ganglionic area were negative for pan-neuronal markers HuD or Peripherin. Relatively high NIBP-like-IR was found in 35-44% of myenteric neurons and 9-10% of submucosal neurons. Approximately 98%, 87%, and 43% of these relatively high NIBP-expressing neurons were positive for choline acetyltransferase, neuronal nitric oxide synthase and Calretinin, respectively. NIBP shRNA knockdown in ENC inhibited TNFα-induced NFκB activation and neuronal differentiation, whereas NIBP overexpression promoted it. CONCLUSIONS & INFERENCES NIBP is extensively expressed in the ENS with relatively high level in a subpopulation of enteric neurons. Various NIBP expression levels in different neurons may represent dynamic trafficking or posttranslational modification of NIBP in some functionally active neurons and ultimately regulate ENS plasticity.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Daniel Bitner
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Adalto Alfredo Pontes Filho
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Fang Li
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Shu Liu
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Hong Wang
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Fan Yang
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Sam Adhikari
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Jennifer Gordon
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Emory University, 615 Michael St., Atlanta, GA 30322 and Atlanta VAMC, Decatur, GA, 30331
| | - Wenhui Hu
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
33
|
Fu M, Landreville S, Agapova OA, Wiley LA, Shoykhet M, Harbour JW, Heuckeroth RO. Retinoblastoma protein prevents enteric nervous system defects and intestinal pseudo-obstruction. J Clin Invest 2013; 123:5152-64. [PMID: 24177421 PMCID: PMC3859411 DOI: 10.1172/jci67653] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 08/15/2013] [Indexed: 12/14/2022] Open
Abstract
The retinoblastoma 1 (RB1) tumor suppressor is a critical regulator of cell cycle progression and development. To investigate the role of RB1 in neural crest-derived melanocytes, we bred mice with a floxed Rb1 allele with mice expressing Cre from the tyrosinase (Tyr) promoter. TyrCre+;Rb1fl/fl mice exhibited no melanocyte defects but died unexpectedly early with intestinal obstruction, striking defects in the enteric nervous system (ENS), and abnormal intestinal motility. Cre-induced DNA recombination occurred in all enteric glia and most small bowel myenteric neurons, yet phenotypic effects of Rb1 loss were cell-type specific. Enteric glia were twice as abundant in mutant mice compared with those in control animals, while myenteric neuron number was normal. Most myenteric neurons also appeared normal in size, but NO-producing myenteric neurons developed very large nuclei as a result of DNA replication without cell division (i.e., endoreplication). Parallel studies in vitro found that exogenous NO and Rb1 shRNA increased ENS precursor DNA replication and nuclear size. The large, irregularly shaped nuclei in NO-producing neurons were remarkably similar to those in progeria, an early-onset aging disorder that has been linked to RB1 dysfunction. These findings reveal a role for RB1 in the ENS.
Collapse
Affiliation(s)
- Ming Fu
- Department of Pediatrics and
Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.
Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
Department of Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Solange Landreville
- Department of Pediatrics and
Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.
Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
Department of Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Olga A. Agapova
- Department of Pediatrics and
Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.
Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
Department of Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Luke A. Wiley
- Department of Pediatrics and
Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.
Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
Department of Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Michael Shoykhet
- Department of Pediatrics and
Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.
Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
Department of Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - J. William Harbour
- Department of Pediatrics and
Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.
Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
Department of Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Robert O. Heuckeroth
- Department of Pediatrics and
Department of Ophthalmology and Visual Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA.
Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida, USA.
Department of Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
34
|
Heredia DJ, Gershon MD, Koh SD, Corrigan RD, Okamoto T, Smith TK. Important role of mucosal serotonin in colonic propulsion and peristaltic reflexes: in vitro analyses in mice lacking tryptophan hydroxylase 1. J Physiol 2013; 591:5939-57. [PMID: 24127620 DOI: 10.1113/jphysiol.2013.256230] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Although there is general agreement that mucosal 5-hydroxytryptamine (5-HT) can initiate peristaltic reflexes in the colon, recent studies have differed as to whether or not the role of mucosal 5-HT is critical. We therefore tested the hypothesis that the secretion of 5-HT from mucosal enterochromaffin (EC) cells is essential for the manifestation of murine colonic peristaltic reflexes. To do so, we analysed the mechanisms underlying faecal pellet propulsion in isolated colons of mice lacking tryptophan hydroxylase 1 (Tph1(-/-) mice), which is the rate-limiting enzyme in the biosynthesis of mucosal but not neuronal 5-HT. We used video analysis of faecal pellet propulsion, tension transducers to record colonic migrating motor complexes (CMMCs) and intracellular microelectrodes to record circular muscle activity occurring spontaneously or following intraluminal distension. When compared with control (Tph1(+/+)) mice, Tph1(-/-) animals exhibited: (1) an elongated colon; (2) larger faecal pellets; (3) orthograde propulsion followed by retropulsion (not observed in Tph1(+/+) colon); (4) slower in vitro propulsion of larger faecal pellets (28% of Tph1(+/+)); (5) CMMCs that infrequently propagated in an oral to anal direction because of impaired descending inhibition; (6) reduced CMMCs and inhibitory responses to intraluminal balloon distension; (7) an absence of reflex activity in response to mucosal stimulation. In addition, (8) thin pellets that propagated along the control colon failed to do so in Tph1(-/-) colon; and (9) the 5-HT3 receptor antagonist ondansetron, which reduced CMMCs and blocked their propagation in Tph1(+/+) mice, failed to alter CMMCs in Tph1(-/-) animals. Our observations suggest that mucosal 5-HT is essential for reflexes driven by mucosal stimulation and is also important for normal propagation of CMMCs and propulsion of pellets in the isolated colon.
Collapse
Affiliation(s)
- Dante J Heredia
- T. K. Smith: Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Newgreen DF, Dufour S, Howard MJ, Landman KA. Simple rules for a "simple" nervous system? Molecular and biomathematical approaches to enteric nervous system formation and malformation. Dev Biol 2013; 382:305-19. [PMID: 23838398 PMCID: PMC4694584 DOI: 10.1016/j.ydbio.2013.06.029] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 06/28/2013] [Accepted: 06/28/2013] [Indexed: 11/17/2022]
Abstract
We review morphogenesis of the enteric nervous system from migratory neural crest cells, and defects of this process such as Hirschsprung disease, centering on cell motility and assembly, and cell adhesion and extracellular matrix molecules, along with cell proliferation and growth factors. We then review continuum and agent-based (cellular automata) models with rules of cell movement and logistical proliferation. Both movement and proliferation at the individual cell level are modeled with stochastic components from which stereotyped outcomes emerge at the population level. These models reproduced the wave-like colonization of the intestine by enteric neural crest cells, and several new properties emerged, such as colonization by frontal expansion, which were later confirmed biologically. These models predict a surprising level of clonal heterogeneity both in terms of number and distribution of daughter cells. Biologically, migrating cells form stable chains made up of unstable cells, but this is not seen in the initial model. We outline additional rules for cell differentiation into neurons, axon extension, cell-axon and cell-cell adhesions, chemotaxis and repulsion which can reproduce chain migration. After the migration stage, the cells re-arrange as a network of ganglia. Changes in cell adhesion molecules parallel this, and we describe additional rules based on Steinberg's Differential Adhesion Hypothesis, reflecting changing levels of adhesion in neural crest cells and neurons. This was able to reproduce enteric ganglionation in a model. Mouse mutants with disturbances of enteric nervous system morphogenesis are discussed, and these suggest future refinement of the models. The modeling suggests a relatively simple set of cell behavioral rules could account for complex patterns of morphogenesis. The model has allowed the proposal that Hirschsprung disease is mostly an enteric neural crest cell proliferation defect, not a defect of cell migration. In addition, the model suggests an explanations for zonal and skip segment variants of Hirschsprung disease, and also gives a novel stochastic explanation for the observed discordancy of Hirschsprung disease in identical twins.
Collapse
Affiliation(s)
- Donald F Newgreen
- The Murdoch Children's Research Institute, Royal Children's Hospital, Parkville, VIC 3052, Australia.
| | | | | | | |
Collapse
|
36
|
Obermayr F, Stamp LA, Anderson CR, Young HM. Genetic fate-mapping of tyrosine hydroxylase-expressing cells in the enteric nervous system. Neurogastroenterol Motil 2013; 25:e283-91. [PMID: 23438425 DOI: 10.1111/nmo.12105] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Accepted: 01/31/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND During development of the enteric nervous system, a subpopulation of enteric neuron precursors transiently expresses catecholaminergic properties. The progeny of these transiently catecholaminergic (TC) cells have not been fully characterized. METHODS We combined in vivo Cre-lox-based genetic fate-mapping with phenotypic analysis to fate-map enteric neuron subtypes arising from tyrosine hydroxylase (TH)-expressing cells. KEY RESULTS Less than 3% of the total (Hu(+) ) neurons in the myenteric plexus of the small intestine of adult mice are generated from transiently TH-expressing cells. Around 50% of the neurons generated from transiently TH-expressing cells are calbindin neurons, but their progeny also include calretinin, neurofilament-M, and serotonin neurons. However, only 30% of the serotonin neurons and small subpopulations (<10%) of the calbindin, calretinin, and neurofilament-M neurons are generated from TH-expressing cells; only 0.2% of nitric oxide synthase neurons arise from TH-expressing cells. CONCLUSIONS & INFERENCES Transiently, catecholaminergic cells give rise to subpopulations of multiple enteric neuron subtypes, but the majority of each of the neuron subtypes arises from non-TC cells.
Collapse
Affiliation(s)
- F Obermayr
- Department of Anatomy & Neuroscience, University of Melbourne, Parkville, Vic., Australia
| | | | | | | |
Collapse
|
37
|
Okamoto T, Bayguinov PO, Broadhead MJ, Smith TK. Ca(2+) transients in submucous neurons during the colonic migrating motor complex in the isolated murine large intestine. Neurogastroenterol Motil 2012; 24:769-78, e354. [PMID: 22632417 DOI: 10.1111/j.1365-2982.2012.01934.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND The colonic migrating motor complex (CMMC) is a spontaneous, rhythmic, and neurally mediated motor pattern generated by myenteric neurons, which can propel fecal pellets in mice. Our aim was to determine whether submucous neurons were also activated during the CMMC. METHODS :The isolated murine colon was opened and sections of mucosa were removed to expose the submucous ganglia, which were then loaded with Fluo-4. KEY RESULTS Colonic migrating motor complexes, which occurred spontaneously or by mechanically stimulating the mucosa, were identified by displacement of the tissue (duration = 23.3 s). Between CMMCs, spontaneous Ca(2+) transients (frequency = 0.9 Hz) were observed in 55% (n = 8) of submucous neurons. During the CMMC, 98% (seven ganglia, n = 7) of submucous neurons within the same ganglion exhibited rapid Ca(2+) transients (1.6 Hz) superimposed on a sustained rise in Ca(2+) (duration ∼23 s) that occurred 1.7 s following the mucosal stimulus; whereas other neurons exhibited a similar, but delayed response that occurred either at 7 or 13 s following the stimulus. The activity in submucous neurons was correlated with activity in adjacent nerve varicosities. Ondansetron (1 mm; 5-HT(3) antagonist) significantly reduced the frequency and duration of the Ca(2+) transient responses. CONCLUSIONS & INFERENCES Activity in the submucous neurons appears to be secondary to that in the myenteric plexus and appears to be generated largely by activity in myenteric descending (serotonergic) interneurons. During the CMMC, there is likely to be an increase in secretion to lubricate and facilitate fecal pellet propulsion.
Collapse
Affiliation(s)
- T Okamoto
- Department of Cell Biology and Physiology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | | | | |
Collapse
|
38
|
Mourad FH, Saadé NE. Neural regulation of intestinal nutrient absorption. Prog Neurobiol 2011; 95:149-62. [PMID: 21854830 DOI: 10.1016/j.pneurobio.2011.07.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 04/28/2011] [Accepted: 07/20/2011] [Indexed: 12/17/2022]
Abstract
The nervous system and the gastrointestinal (GI) tract share several common features including reciprocal interconnections and several neurotransmitters and peptides known as gut peptides, neuropeptides or hormones. The processes of digestion, secretion of digestive enzymes and then absorption are regulated by the neuro-endocrine system. Luminal glucose enhances its own absorption through a neuronal reflex that involves capsaicin sensitive primary afferent (CSPA) fibres. Absorbed glucose stimulates insulin release that activates hepatoenteric neural pathways leading to an increase in the expression of glucose transporters. Adrenergic innervation increases glucose absorption through α1 and β receptors and decreases absorption through activation of α2 receptors. The vagus nerve plays an important role in the regulation of diurnal variation in transporter expression and in anticipation to food intake. Vagal CSPAs exert tonic inhibitory effects on amino acid absorption. It also plays an important role in the mediation of the inhibitory effect of intestinal amino acids on their own absorption at the level of proximal or distal segment. However, chronic extrinsic denervation leads to a decrease in intestinal amino acid absorption. Conversely, adrenergic agonists as well as activation of CSPA fibres enhance peptides uptake through the peptide transporter PEPT1. Finally, intestinal innervation plays a minimal role in the absorption of fat digestion products. Intestinal absorption of nutrients is a basic vital mechanism that depends essentially on the function of intestinal mucosa. However, intrinsic and extrinsic neural mechanisms that rely on several redundant loops are involved in immediate and long-term control of the outcome of intestinal function.
Collapse
Affiliation(s)
- Fadi H Mourad
- Department of Internal Medicine, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| | | |
Collapse
|
39
|
Deiteren A, De Winter BY, Nullens S, Pelckmans PA, De Man JG. Role of tachykinin receptors in the modulation of colonic peristaltic activity in mice. Eur J Pharmacol 2011; 667:339-47. [PMID: 21645508 DOI: 10.1016/j.ejphar.2011.05.044] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2010] [Revised: 05/16/2011] [Accepted: 05/22/2011] [Indexed: 11/20/2022]
Abstract
Tachykinins are important mediators of neuroneuronal and neuromuscular transmission in the gastrointestinal tract, however their contribution to colonic peristalsis in mice remains unclear. Therefore, our aim was to characterise the functional role of tachykinins in mediating peristalsis by evaluating the effect of selective tachykinin NK(1), NK(2) and NK(3) receptor agonists and antagonists on in vitro colonic peristaltic activity in mice. Using a modified Trendelenburg set-up, gradual distension of proximal and distal colonic segments evoked rhythmic, aborally migrating contractions. Peristaltic activity was assessed by quantifying the amplitude and interval of the corresponding pressure waves. Stimulation of NK(1) receptors showed regional differences as both the pressure amplitude and interval were enhanced in the distal colon without affecting peristalsis proximally. Blockade of NK(1) receptors reduced the peristaltic pressure amplitude in the proximal and distal colon while the interval was not significantly altered. NK(2) receptor stimulation resulted in a modest enhancement of the amplitude in proximal and distal segments and a slightly prolonged interval distally. Blockade of NK(2) receptors reduced the peristaltic pressure amplitude and interval in the distal colon. NK(3) receptor stimulation significantly augmented the amplitude in both segments and prolonged the interval distally. However, NK(3) receptor blockade had no effect on peristaltic activity. In conclusion, tachykinins contribute to colonic peristalsis in mice by acting mainly on NK(1) and NK(2) receptors and their effects show a proximal-to-distal gradient. NK(3) receptors might play a role in conditions of excess tachykinin release but appear not to be involved under the conditions of the present study.
Collapse
Affiliation(s)
- Annemie Deiteren
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | | | | | | | | |
Collapse
|
40
|
Neural precursor death is central to the pathogenesis of intestinal aganglionosis in Ret hypomorphic mice. J Neurosci 2010; 30:5211-8. [PMID: 20392943 DOI: 10.1523/jneurosci.6244-09.2010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The RET tyrosine kinase is required for the migration, proliferation, and survival of the enteric neural crest-derived cells (ENCCs) that form the enteric nervous system (ENS). Hypomorphic RET alleles cause intestinal aganglionosis [Hirschsprung disease (HSCR)], in which delayed migration and successive nonapoptotic ENCC death are considered to be major contributory factors. The significance of ENCC death in intestinal aganglionosis, however, has remained unclear. We show that elevated expression of Bcl-xL inhibits ENCC death in both Ret-null and hypomorphic states. However, the rescued Ret-null mice showed ENS malfunction with reduced nitric oxide synthase expression in colonic neurons, revealing the requirement of RET for neuronal differentiation. In contrast, the inhibition of cell death allows morphologically and functionally normal ENS formation in Ret hypomorphic mice. These results indicate that ENCC death is a principal cause of intestinal aganglionosis in a Ret hypomorphic state, and suggest that the inhibition of cell death is a route to the prevention of HSCR.
Collapse
|
41
|
Tan LL, Bornstein JC, Anderson CR. The neurochemistry and innervation patterns of extrinsic sensory and sympathetic nerves in the myenteric plexus of the C57Bl6 mouse jejunum. Neuroscience 2009; 166:564-79. [PMID: 20034545 DOI: 10.1016/j.neuroscience.2009.12.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 12/11/2009] [Accepted: 12/12/2009] [Indexed: 12/31/2022]
Abstract
In vitro anterograde tracing of axons in mesenteric nerve trunks using biotinamide in combination with immunohistochemical labelling was used to characterize the extrinsic nerve projections in the myenteric plexus of the mouse jejunum. Anterogradely-labelled spinal sensory fibres innervating the enteric nervous system were identified by their immunoreactivity for calcitonin gene-related peptide (CGRP), while sympathetic noradrenergic fibres were detected with tyrosine hydroxylase (TH), using confocal microscopy. The presence of these markers has been previously described in the spinal sensory and sympathetic fibres. Labelled extrinsic nerve fibres in the myenteric plexus were identified apposing enteric neurons that were immunoreactive for either calretinin (CalR), calbindin (CalB) or nitric oxide synthase (NOS). Of the total anterogradely labelled axons in the myenteric plexus, 20% were CGRP-immunoreactive. Labelled CGRP-immunoreactive varicosities were closely apposed to CalR-immunoreactive myenteric cells, many of which were Dogiel type I (40%; interneurons) or type II (20%; intrinsic sensory) neurons. Labelled CGRP-immunoreactive varicosities were also observed in close appositions to CalB-immunoreactive myenteric cell bodies, of which a small subset had type II morphology (18%; intrinsic sensory neurons). A further 43% of all biotinamide-filled fibres were immunoreactive for TH and these fibres were apposed to CalR-immunoreactive cell bodies (small-sized; excitatory motor neurons) and NOS-immunoreactive cell bodies (either type I or small neurons; inhibitory motor neurons and interneurons) in the myenteric plexus. The results provide a neurochemical and neuroanatomical basis for connections between dorsal root afferent neurons and myenteric neurons and suggest an anatomical substrate for the well-known modulation of enteric circuits from sympathetic nerves. No anterogradely-labelled fibres were stained for NOS-immunoreactivity, despite more than 60% of dorsal root ganglion (DRG) neurons retrogradely labelled from the jejunum showing NOS-immunoreactivity. This was due to a substantial, time-dependent, and apparently selective, loss of NOS from extrinsic axons under in vitro conditions. Lastly, a small population of non-immunoreactive biotinamide-filled fibres (<1%) gave rise to dense terminal structures around individual myenteric cell bodies lacking CalR, CalB or NOS. These specialized endings may represent vagal fibres or a subset of spinal sensory neurons that do not contain CGRP.
Collapse
Affiliation(s)
- L L Tan
- Department of Physiology, University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | |
Collapse
|
42
|
Chiocchetti R, Bombardi C, Mongardi-Fantaguzzi C, Venturelli E, Russo D, Spadari A, Montoneri C, Romagnoli N, Grandis A. Intrinsic innervation of the horse ileum. Res Vet Sci 2009; 87:177-85. [DOI: 10.1016/j.rvsc.2009.03.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2008] [Revised: 12/10/2008] [Accepted: 03/13/2009] [Indexed: 10/20/2022]
|
43
|
Abstract
The mature enteric nervous system (ENS) is composed of many different neuron subtypes and enteric glia, which all arise from the neural crest. How this diversity is generated from neural crest-derived cells is a central question in neurogastroenterology, as defects in these processes are likely to underlie some paediatric motility disorders. Here we review the developmental appearance (the earliest age at which expression of specific markers can be localized) and birthdates (the age at which precursors exit the cell cycle) of different enteric neuron subtypes, and their projections to some targets. We then focus on what is known about the mechanisms underlying the generation of enteric neuron diversity and axon pathfinding. Finally, we review the development of the ENS in humans and the etiologies of a number of paediatric motility disorders.
Collapse
Affiliation(s)
- Marlene M Hao
- Department of Anatomy & Cell Biology, University of MelbourneParkville, Victoria, Australia
| | - Heather M Young
- Department of Anatomy & Cell Biology, University of MelbourneParkville, Victoria, Australia
| |
Collapse
|
44
|
Identification of neuron types in the submucosal ganglia of the mouse ileum. Cell Tissue Res 2009; 336:179-89. [PMID: 19326148 DOI: 10.1007/s00441-009-0773-2] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Accepted: 01/28/2009] [Indexed: 12/20/2022]
Abstract
The continuing and even expanding use of genetically modified mice to investigate the normal physiology and development of the enteric nervous system and for the study of pathophysiology in mouse models emphasises the need to identify all the neuron types and their functional roles in mice. An investigation that chemically and morphologically defined all the major neuron types with cell bodies in myenteric ganglia of the mouse small intestine was recently completed. The present study was aimed at the submucosal ganglia, with the purpose of similarly identifying the major neuron types with cell bodies in these ganglia. We found that the submucosal neurons could be divided into three major groups: neurons with vasoactive intestinal peptide (VIP) immunoreactivity (51% of neurons), neurons with choline acetyltransferase (ChAT) immunoreactivity (41% of neurons) and neurons that expressed neither of these markers. Most VIP neurons contained neuropeptide Y (NPY) and about 40% were immunoreactive for tyrosine hydroxylase (TH); 22% of all submucosal neurons were TH/VIP. VIP-immunoreactive nerve terminals in the mucosa were weakly immunoreactive for TH but separate populations of TH- and VIP-immunoreactive axons innervated the arterioles in the submucosa. Of the ChAT neurons, about half were immunoreactive for both somatostatin and calcitonin gene-related peptide (CGRP). Calretinin immunoreactivity occurred in over 90% of neurons, including the VIP neurons. The submucosal ganglia and submucosal arterioles were innervated by sympathetic noradrenergic neurons that were immunoreactive for TH and NPY; no VIP and few calretinin fibres innervated submucosal neurons. We conclude that the submucosal ganglia contain cell bodies of VIP/NPY/TH/calretinin non-cholinergic secretomotor neurons, VIP/NPY/calretinin vasodilator neurons, ChAT/CGRP/somatostatin/calretinin cholinergic secretomotor neurons and small populations of cholinergic and non-cholinergic neurons whose targets have yet to be identified. No evidence for the presence of type-II putative intrinsic primary afferent neurons was found.
Collapse
|
45
|
Sibaev A, Yüce B, Kemmer M, Van Nassauw L, Broedl U, Allescher HD, Göke B, Timmermans JP, Storr M. Cannabinoid-1 (CB1) receptors regulate colonic propulsion by acting at motor neurons within the ascending motor pathways in mouse colon. Am J Physiol Gastrointest Liver Physiol 2009; 296:G119-28. [PMID: 19033531 DOI: 10.1152/ajpgi.90274.2008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Cannabinoid-1 (CB(1)) receptors on myenteric neurons are involved in the regulation of intestinal motility. Our aim was to investigate CB(1) receptor involvement in ascending neurotransmission in mouse colon and to characterize the involved structures by functional and morphological means. Presence of the CB(1) receptor was investigated by RT-PCR, and immunohistochemistry was used for colabeling studies. Myenteric reflex responses were initiated by electrical stimulation (ES) at different distances, and junction potentials (JP) were recorded from circular smooth muscle cells by intracellular recording in an unpartitioned and a partitioned recording chamber. In vivo colonic propulsion was tested in wild-type and CB(1)(-/-) mice. Immunostaining with the cytoskeletal marker peripherin showed CB(1) immunoreactivity both on Dogiel type I and type II neurons. Further neurochemical characterization revealed CB(1) on choline acetyltransferase-, calretinin-, and 5-HT-immunopositive myenteric neurons, but nitrergic neurons appeared immunonegative for CB(1) immunostaining. Solitary spindle-shaped CB(1)-immunoreactive cells in between smooth muscle cells lacked specific markers for interstitial cells of Cajal or glial cells. ES elicited neuronally mediated excitatory JP (EJP) and inhibitory JP. Gradual increases in distance resulted in a wave-like EJP with EJP amplitudes being maximal at the location of stimulating electrode 6 and a maximal EJP projection distance of approximately 18 mm. The CB(1) receptor agonist WIN 55,212-2 reduced the amplitude of EJP and was responsible for shortening the oral spreading of the excitatory impulse. In a partitioned chamber, WIN 55,212-2 reduced EJP at the separated oral sites, proving that CB(1) activation inhibits interneuron-mediated neurotransmission. These effects were absent in the presence of the CB(1) antagonist SR141716A, which, when given alone, had no effect. WIN 55,212-2 inhibited colonic propulsion in wild-type mice but not in SR141716A-pretreated wild-type or CB(1)(-/-) mice. Activation of the CB(1) receptor modulates excitatory cholinergic neurotransmission in mouse colon by reducing amplitude and spatial spreading of the ascending electrophysiological impulses. This effect on electrophysiological spreading involves CB(1)-mediated effects on motor neurons and ascending interneurons and is likely to underlie the here reported in vivo reduction in colonic propulsion.
Collapse
Affiliation(s)
- Andrei Sibaev
- Dept. of Internal Medicine II, Ludwig Maxmillians University, Munich, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Rivera LR, Thacker M, Furness JB. High- and medium-molecular-weight neurofilament proteins define specific neuron types in the guinea-pig enteric nervous system. Cell Tissue Res 2008; 335:529-38. [DOI: 10.1007/s00441-008-0732-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2008] [Accepted: 10/31/2008] [Indexed: 02/03/2023]
|
47
|
Neal KB, Parry LJ, Bornstein JC. Strain-specific genetics, anatomy and function of enteric neural serotonergic pathways in inbred mice. J Physiol 2008; 587:567-86. [PMID: 19064621 DOI: 10.1113/jphysiol.2008.160416] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Serotonin (5-HT) powerfully affects small intestinal motility and 5-HT-immunoreactive (IR) neurones are highly conserved between species. 5-HT synthesis in central neurones and gastrointestinal mucosa depends on tissue-specific isoforms of the enzyme tryptophan hydroxylase (TPH). RT-PCR identified strain-specific expression of a polymorphism (1473C/G) of the tph2 gene in longitudinal muscle-myenteric plexus preparations of C57Bl/6 and Balb/c mice. The former expressed the high-activity C allele, the latter the low-activity G allele. Confocal microscopy was used to examine close contacts between 5-HT-IR varicosities and myenteric neurones immunoreactive for neuronal nitric oxide synthase (NOS) or calretinin in these two strains. Significantly more close contacts were identified to NOS- (P < 0.05) and calretinin-IR (P < 0.01) neurones in C57Bl/6 jejunum (NOS 1.6 +/- 0.3, n = 52; calretinin 5.2 +/- 0.4, n = 54), than Balb/c jejunum (NOS 0.9 +/- 0.2, n = 78; calretinin 3.5 +/- 0.3, n = 98). Propagating contractile complexes (PCCs) were identified in the isolated jejunum by constructing spatiotemporal maps from video recordings of cannulated segments in vitro. These clusters of contractions usually arose towards the anal end and propagated orally. Regular PCCs were initiated at intraluminal pressures of 6 cmH(2)O, and abolished by tetrodotoxin (1 microm). Jejunal PCCs from C57Bl/6 mice were suppressed by a combination of granisetron (1 microm, 5-HT(3) antagonist) and SB207266 (10 nm, 5-HT(4) antagonist), but PCCs from Balb/c mice were unaffected. There were, however, no strain-specific differences in sensitivity of longitudinal muscle contractions to exogenous 5-HT or blockade of 5-HT(3) and 5-HT(4) receptors. These data associate a genetic difference with significant structural and functional consequences for enteric neural serotonergic pathways in the jejunum.
Collapse
Affiliation(s)
- Kathleen B Neal
- Department of Physiology, The University of Melbourne, Parkville, Victoria, Australia.
| | | | | |
Collapse
|
48
|
Qu ZD, Thacker M, Castelucci P, Bagyánszki M, Epstein ML, Furness JB. Immunohistochemical analysis of neuron types in the mouse small intestine. Cell Tissue Res 2008; 334:147-61. [PMID: 18855018 DOI: 10.1007/s00441-008-0684-7] [Citation(s) in RCA: 243] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2008] [Accepted: 08/22/2008] [Indexed: 12/11/2022]
Abstract
The definition of the nerve cell types of the myenteric plexus of the mouse small intestine has become important, as more researchers turn to the use of mice with genetic mutations to analyze roles of specific genes and their products in enteric nervous system function and to investigate animal models of disease. We have used a suite of antibodies to define neurons by their shapes, sizes, and neurochemistry in the myenteric plexus. Anti-Hu antibodies were used to reveal all nerve cells, and the major subpopulations were defined in relation to the Hu-positive neurons. Morphological Type II neurons, revealed by anti-neurofilament and anti-calcitonin gene-related peptide antibodies, represented 26% of neurons. The axons of the Type II neurons projected through the circular muscle and submucosa to the mucosa. The cell bodies were immunoreactive for choline acetyltransferase (ChAT), and their terminals were immunoreactive for vesicular acetylcholine transporter (VAChT). Nitric oxide synthase (NOS) occurred in 29% of nerve cells. Most were also immunoreactive for vasoactive intestinal peptide, but they were not tachykinin (TK)-immunoreactive, and only 10% were ChAT-immunoreactive. Numerous NOS terminals occurred in the circular muscle. We deduced that 90% of NOS neurons were inhibitory motor neurons to the muscle (26% of all neurons) and 10% (3% of all neurons) were interneurons. Calretinin immunoreactivity was found in a high proportion of neurons (52%). Many of these had TK immunoreactivity. Small calretinin neurons were identified as excitatory neurons to the longitudinal muscle (about 20% of neurons, with ChAT/calretinin/+/- TK chemical coding). Excitatory neurons to the circular muscle (about 10% of neurons) had the same coding. Calretinin immunoreactivity also occurred in a proportion of Type II neurons. Thus, over 90% of neurons in the myenteric plexus of the mouse small intestine can be currently identified by their neurochemistry and shape.
Collapse
Affiliation(s)
- Zheng-Dong Qu
- Department of Anatomy & Cell Biology and Centre for Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | | | | | | | | | | |
Collapse
|
49
|
Jungbauer C, Lindig TM, Schrödl F, Neuhuber W, Brehmer A. Chemical coding of myenteric neurons with different axonal projection patterns in the porcine ileum. J Anat 2007; 209:733-43. [PMID: 17118061 PMCID: PMC2049006 DOI: 10.1111/j.1469-7580.2006.00653.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The aim of this study was to perform an immunohistochemical characterization of two different myenteric neuron types of the pig displaying opposite axonal projections. These were type I neurons equipped with lamellar dendrites that projected mainly orally, and type VI neurons that displayed typical axonal dendrites and projected anally. Double immunostainings of longitudinal muscle/myenteric plexus wholemounts from ileal segments of four pigs were performed to visualize neurofilaments (NF) in combination with calcitonin gene-related peptide (CGRP), leu-enkephalin (ENK) and substance P (SP), respectively. Triple immunostainings of wholemounts, using antibodies against neuronal nitric oxide synthase (nNOS) and vasoactive intestinal peptide (VIP) as well as against VIP and galanin (GAL), were performed. We found that 78% of type I neurons immunoreacted to ENK, 21% to CGRP and 24% to SP. The NF-positive type I neurons co-reactive for one of the three above markers displayed mostly frayed outlines of both their somal contours and their broadened dendritic endings. By contrast, most of the non-coreactive type I neurons displayed rather sharply outlined somata and dendrites. No type I neuron immunoreacted to nNOS, VIP or GAL and none of the type VI NF-reactive neurons reacted to CGRP, ENK or SP. All type VI neurons investigated displayed immunoreactivity for nNOS, 92% of which were co-reactive for VIP. Co-reactivity for VIP and GAL was found in 69% of type VI neurons, 21% were positive for VIP but negative for GAL, 9% were negative for both GAL and VIP, and 1% were positive for GAL but negative for VIP. We conclude that there are two subpopulations of morphological type I neurons. One of these displays mainly oral projections and could not be further characterized in this study. The other, which may correspond to neurons innervating the longitudinal and circular muscle layers, were partly immunoreactive for ENK, CGRP and/or SP. Type VI neurons are immunoreactive for nNOS frequently co-localized with VIP and, partly, also GAL. These may be inhibitory motor neurons and are different from VIP/GAL-coreactive minineurons described earlier.
Collapse
|
50
|
Mulè F, Amato A, Serio R. Role for NK(1) and NK(2) receptors in the motor activity in mouse colon. Eur J Pharmacol 2007; 570:196-202. [PMID: 17597603 DOI: 10.1016/j.ejphar.2007.05.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2007] [Revised: 05/04/2007] [Accepted: 05/22/2007] [Indexed: 01/09/2023]
Abstract
The present study examined the effects induced by endogenous and exogenous activation of NK(1) and NK(2) receptors on the mechanical activity of mouse proximal colon. Experiments were performed in vitro recording the changes in intraluminal pressure from isolated colonic segments. Electrical field stimulation in the presence of atropine and guanethidine produced a small relaxation, followed by nonadrenergic noncholinergic (NANC) contraction. SR140333, NK(1) receptor antagonist, or SR48968, NK(2) receptor antagonist, significantly reduced the contraction, although SR48968 appeared more efficacious. The co-administration of SR140333 and SR48968 virtually abolished the NANC contraction. [Sar(9), Met(O(2))(11)]-substance P, selective NK(1) receptor agonist, induced a concentration-dependent biphasic effect, contraction followed by reduction of the mechanical spontaneous activity. Both effects were antagonized by SR140333, but not by SR48968. [beta-Ala(8)]-neurokinin A (4-10), selective NK(2) receptor agonist, evoked concentration-dependent contraction, which was antagonized by SR48968, but not by SR140333. The contraction induced by [Sar(9), Met(O(2))(11)]-substance P, but not by [beta-Ala(8)]-neurokinin A (4-10), was reduced by tetrodotoxin or atropine, and increased by N(omega)-nitro-L-arginine methyl ester (L-NAME), inhibitor of nitric oxide synthase. The inhibitory effects induced by [Sar(9), Met(O(2))(11)]-substance P were abolished by tetrodotoxin or L-NAME. The results of the present study suggest that in mouse colon both NK(1) and NK(2) receptors are junctionally activated by endogenous tachykinins to cause an additive response. NK(1) receptors appear to be located on cholinergic and on nitrergic neurons as well as on smooth muscle cells, whereas NK(2) receptors seem to be present exclusively on smooth muscle cells.
Collapse
Affiliation(s)
- Flavia Mulè
- Dipartimento di Biologia cellulare e dello Sviluppo, Laboratorio di Fisiologia generale, Università di Palermo, Viale delle Scienze, 90128 Palermo, Italy.
| | | | | |
Collapse
|