1
|
Mazzoni M, Cabanillas L, Costanzini A, Caremoli F, Million M, Larauche M, Clavenzani P, De Giorgio R, Sternini C. Distribution, quantification, and characterization of substance P enteric neurons in the submucosal and myenteric plexuses of the porcine colon. Cell Tissue Res 2024; 395:39-51. [PMID: 37982872 PMCID: PMC10774220 DOI: 10.1007/s00441-023-03842-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/30/2023] [Indexed: 11/21/2023]
Abstract
The pig is an important translational model for studying intestinal physiology and disorders for its many homologies with humans, including the organization of the enteric nervous system (ENS), the major regulator of gastrointestinal functions. This study focused on the quantification and neurochemical characterization of substance P (SP) neurons in the pig ascending (AC) and descending colon (DC) in wholemount preparations of the inner submucosal plexus (ISP), outer submucosal plexus (OSP), and myenteric plexus (MP). We used antibodies for the pan-neuronal marker HuCD, and choline acetyltransferase (ChAT) and neuronal nitric oxide synthase (nNOS), markers for excitatory and inhibitory transmitters, for multiple labeling immunofluorescence and high-resolution confocal microscopy. The highest density of SP immunoreactive (IR) neurons was in the ISP (222/mm2 in the AC, 166/mm2 in the DC), where they make up about a third of HuCD-IR neurons, compared to the OSP and MP (19-22% and 13-17%, respectively, P < 0.001-0.0001). HuCD/SP/ChAT-IR neurons (up to 23%) were overall more abundant than HuCD/SP/nNOS-IR neurons (< 10%). Most SP-IR neurons contained ChAT-IR (62-85%), whereas 18-38% contained nNOS-IR with the highest peak in the OSP. A subpopulation of SP-IR neurons contains both ChAT- and nNOS-IR with the highest peak in the OSP and ISP of DC (33-36%) and the lowest in the ISP of AC (< 10%, P < 0.001). SP-IR varicose fibers were abundant in the ganglia. This study shows that SP-IR neurons are functionally distinct with variable proportions in different plexuses in the AC and DC reflecting diverse functions of specific colonic regions.
Collapse
Affiliation(s)
- Maurizio Mazzoni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, Bologna, Italy
| | - Luis Cabanillas
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Anna Costanzini
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Filippo Caremoli
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Current address: San Raffaele Hospital, Milan, Italy
| | - Mulugeta Million
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, 90095, USA
| | - Muriel Larauche
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Paolo Clavenzani
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, Bologna, Italy
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Catia Sternini
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
2
|
Chen BN, Humenick A, Yew WP, Peterson RA, Wiklendt L, Dinning PG, Spencer NJ, Wattchow DA, Costa M, Brookes SJH. Types of Neurons in the Human Colonic Myenteric Plexus Identified by Multilayer Immunohistochemical Coding. Cell Mol Gastroenterol Hepatol 2023; 16:573-605. [PMID: 37355216 PMCID: PMC10469081 DOI: 10.1016/j.jcmgh.2023.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND AND AIMS Gut functions including motility, secretion, and blood flow are largely controlled by the enteric nervous system. Characterizing the different classes of enteric neurons in the human gut is an important step to understand how its circuitry is organized and how it is affected by disease. METHODS Using multiplexed immunohistochemistry, 12 discriminating antisera were applied to distinguish different classes of myenteric neurons in the human colon (2596 neurons, 12 patients) according to their chemical coding. All antisera were applied to every neuron, in multiple layers, separated by elutions. RESULTS A total of 164 combinations of immunohistochemical markers were present among the 2596 neurons, which could be divided into 20 classes, with statistical validation. Putative functions were ascribed for 4 classes of putative excitatory motor neurons (EMN1-4), 4 inhibitory motor neurons (IMN1-4), 3 ascending interneurons (AIN1-3), 6 descending interneurons (DIN1-6), 2 classes of multiaxonal sensory neurons (SN1-2), and a small, miscellaneous group (1.8% of total). Soma-dendritic morphology was analyzed, revealing 5 common shapes distributed differentially between the 20 classes. Distinctive baskets of axonal varicosities surrounded 45% of myenteric nerve cell bodies and were associated with close appositions, suggesting possible connectivity. Baskets of cholinergic terminals and several other types of baskets selectively targeted ascending interneurons and excitatory motor neurons but were significantly sparser around inhibitory motor neurons. CONCLUSIONS Using a simple immunohistochemical method, human myenteric neurons were shown to comprise multiple classes based on chemical coding and morphology and dense clusters of axonal varicosities were selectively associated with some classes.
Collapse
Affiliation(s)
- Bao Nan Chen
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Adam Humenick
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Wai Ping Yew
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Rochelle A Peterson
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Lukasz Wiklendt
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Phil G Dinning
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia; Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Nick J Spencer
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - David A Wattchow
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, South Australia, Australia
| | - Marcello Costa
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Simon J H Brookes
- Human Physiology, College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
3
|
Heumüller-Klug S, Maurer K, Tapia-Laliena MÁ, Sticht C, Christmann A, Mörz H, Khasanov R, Wink E, Schulte S, Greffrath W, Treede RD, Wessel LM, Schäfer KH. Impact of cryopreservation on viability, gene expression and function of enteric nervous system derived neurospheres. Front Cell Dev Biol 2023; 11:1196472. [PMID: 37377739 PMCID: PMC10291272 DOI: 10.3389/fcell.2023.1196472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Introduction: Impairment of both the central and peripheral nervous system is a major cause of mortality and disability. It varies from an affection of the brain to various types of enteric dysganglionosis. Congenital enteric dysganglionosis is characterized by the local absence of intrinsic innervation due to deficits in either migration, proliferation or differentiation of neural stem cells. Despite surgery, children's quality of life is reduced. Neural stem cell transplantation seems a promising therapeutic approach, requiring huge amounts of cells and multiple approaches to fully colonize the diseased areas completely. A combination of successful expansion and storage of neural stem cells is needed until a sufficient amount of cells is generated. This must be combined with suitable cell transplantation strategies, that cover all the area affected. Cryopreservation provides the possibility to store cells for long time, unfortunately with side effects, i.e., upon vitality. Methods: In this study we investigate the impact of different freezing and thawing protocols (M1-M4) upon enteric neural stem cell survival, protein and gene expression, and cell function. Results: Freezing enteric nervous system derived neurospheres (ENSdN) following slow-freezing protocols (M1-3) resulted in higher survival rates than flash-freezing (M4). RNA expression profiles were least affected by freezing protocols M1/2, whereas the protein expression of ENSdN remained unchanged after treatment with protocol M1 only. Cells treated with the most promising freezing protocol (M1, slow freezing in fetal calf serum plus 10% DMSO) were subsequently investigated using single-cell calcium imaging. Freezing of ENSdN did not alter the increase in intracellular calcium in response to a specific set of stimuli. Single cells could be assigned to functional subgroups according to response patterns and a significant shift towards cells responding to nicotine was observed after freezing. Discussion: The results demonstrate that cryopreservation of ENSdN is possible with reduced viability, only slight changes in protein/gene expression patterns and without an impact on the neuronal function of different enteric nervous system cell subtypes, with the exception of a subtle upregulation of cells expressing nicotinergic acetylcholine receptors. In summary, cryopreservation presents a good method to store sufficient amounts of enteric neural stem cells without neuronal impairment, in order to enable subsequent transplantation of cells into compromised tissues.
Collapse
Affiliation(s)
- Sabine Heumüller-Klug
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Kristina Maurer
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - María Á. Tapia-Laliena
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carsten Sticht
- Medical Research Centre Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Anne Christmann
- AGENS, University of Applied Sciences Kaiserslautern Campus Zweibrücken, Kaiserslautern, Germany
| | - Handan Mörz
- Mannheim Center for Translational Neuroscience (MCTN), Department of Neurophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rasul Khasanov
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Elvira Wink
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Steven Schulte
- AGENS, University of Applied Sciences Kaiserslautern Campus Zweibrücken, Kaiserslautern, Germany
| | - Wolfgang Greffrath
- Mannheim Center for Translational Neuroscience (MCTN), Department of Neurophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rolf-Detlef Treede
- Mannheim Center for Translational Neuroscience (MCTN), Department of Neurophysiology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Lucas M. Wessel
- Department of Pediatric Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Karl-Herbert Schäfer
- AGENS, University of Applied Sciences Kaiserslautern Campus Zweibrücken, Kaiserslautern, Germany
| |
Collapse
|
4
|
Takla M, Saadeh K, Tse G, Huang CLH, Jeevaratnam K. Ageing and the Autonomic Nervous System. Subcell Biochem 2023; 103:201-252. [PMID: 37120470 DOI: 10.1007/978-3-031-26576-1_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
The vertebrate nervous system is divided into central (CNS) and peripheral (PNS) components. In turn, the PNS is divided into the autonomic (ANS) and enteric (ENS) nervous systems. Ageing implicates time-related changes to anatomy and physiology in reducing organismal fitness. In the case of the CNS, there exists substantial experimental evidence of the effects of age on individual neuronal and glial function. Although many such changes have yet to be experimentally observed in the PNS, there is considerable evidence of the role of ageing in the decline of ANS function over time. As such, this chapter will argue that the ANS constitutes a paradigm for the physiological consequences of ageing, as well as for their clinical implications.
Collapse
Affiliation(s)
| | | | - Gary Tse
- Kent and Medway Medical School, Canterbury, UK
- University of Surrey, Guildford, UK
| | | | | |
Collapse
|
5
|
Hibberd TJ, Yew WP, Dodds KN, Xie Z, Travis L, Brookes SJ, Costa M, Hu H, Spencer NJ. Quantification of CGRP-immunoreactive myenteric neurons in mouse colon. J Comp Neurol 2022; 530:3209-3225. [PMID: 36043843 DOI: 10.1002/cne.25403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/08/2022] [Accepted: 08/17/2022] [Indexed: 11/07/2022]
Abstract
Quantitative data of biological systems provide valuable baseline information for understanding pathology, experimental perturbations, and computational modeling. In mouse colon, calcitonin gene-related peptide (CGRP) is expressed by myenteric neurons with multiaxonal (Dogiel type II) morphology, characteristic of intrinsic primary afferent neurons (IPANs). Analogous neurons in other species and gut regions represent 5-35% of myenteric neurons. We aimed to quantify proportions of CGRP-immunopositive (CGRP+) myenteric neurons. Colchicine-treated wholemount preparations of proximal, mid, and distal colon were labeled for HuC/D, CGRP, nitric oxide synthase (NOS), and peripherin (Per). The pan-neuronal markers (Hu+/Per+) co-labeled 94% of neurons. Hu+/Per- neurons comprised ∼6%, but Hu-/Per+ cells were rare. Thus, quantification was based on Hu+ myenteric neurons (8576 total; 1225 ± 239 per animal, n = 7). CGRP+ cell bodies were significantly larger than the average of all Hu+ neurons (329 ± 13 vs. 261 ± 12 μm2 , p < .0001). CGRP+ neurons comprised 19% ± 3% of myenteric neurons without significant regional variation. NOS+ neurons comprised 42% ± 2% of myenteric neurons overall, representing a lower proportion in proximal colon, compared to mid and distal colon (38% ± 2%, 44% ± 2%, and 44% ± 3%, respectively). Peripherin immunolabeling revealed cell body and axonal morphology in some myenteric neurons. Whether all CGRP+ neurons were multiaxonal could not be addressed using peripherin immunolabeling. However, of 118 putatively multiaxonal neurons first identified based on peripherin immunoreactivity, all were CGRP+ (n = 4). In conclusion, CGRP+ myenteric neurons in mouse colon were comprehensively quantified, occurring within a range expected of a putative IPAN marker. All Per+ multiaxonal neurons, characteristic of Dogiel type II/IPAN morphology, were CGRP+.
Collapse
Affiliation(s)
- Timothy J Hibberd
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Wai Ping Yew
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Kelsi N Dodds
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Zili Xie
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Lee Travis
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Simon J Brookes
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Marcello Costa
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| | - Hongzhen Hu
- Department of Anesthesiology, The Center for the Study of Itch & Sensory Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Adelaide, South Australia, Australia
| |
Collapse
|
6
|
Characterization of Neurochemical Signature Alterations in the Enteric Nervous System in Autoimmune Encephalomyelitis. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To date, it has remained unclear whether gastrointestinal symptoms, which are frequently observed in patients with multiple sclerosis (MS), are accompanied by pathology of the enteric nervous system (ENS). Here, the neurotransmitter signature of ENS neurons and morphological alterations of interstitial cells of Cajal (ICCs) were studied in patients with MS and mice with experimental autoimmune encephalomyelitis (EAE), which is an animal model of MS. Immunohistochemical analysis was performed on colonic whole mounts from mice with EAE and on paraffin-embedded sections of intestinal tissue from patients with MS. Antibodies against neurotransmitters or their enzymes (including vasoactive intestinal peptide (VIP), neuronal nitric oxide synthase (nNOS), and choline acetyltransferase (ChAT)) were used in conjunction with pan-neuronal markers. In addition, the presence of anoctamin 1 (ANO1)-expressing ICCs was studied. ENS changes were observed in the myenteric plexus, but they were absent in the submucosal plexus of both EAE mice and patients with MS. There was a significant decrease in the percentage of ChAT-positive neurons in EAE mice as opposed to a trend toward an increase in patients with MS. Moreover, while ANO1 expression was decreased in EAE mice, patients with MS displayed a significant increase. Although additional studies are necessary to accomplish an in-depth characterization of ENS alterations in MS, our results imply that such alterations exist and may reveal novel insights into the pathophysiology of MS.
Collapse
|
7
|
Wattchow DA, Smolilo D, Hibberd T, Spencer NJ, Brookes SJ, De Giorgio R, Heitmann PT, Costa M, Dinning PG. The human enteric nervous system. Historical and modern advances. Collaboration between science and surgery. ANZ J Surg 2022; 92:1365-1370. [PMID: 35403788 DOI: 10.1111/ans.17688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND There are considerable advantages and opportunities for surgeons and trainee surgeons in conducting a period of research allied with basic scientists. Such clinicians are well placed to define relevant clinical questions, provide human material (tissue, biopsy and blood) and translate the techniques derived in experimental animals to human subjects. METHODS This small review explores research conducted on the nervous system of the intestines, with an emphasis on the translation of findings from animal to human. RESULTS This work shows that new techniques of immunohistochemistry and retrograde tracing, developed in animal tissue, have greatly expanded our knowledge of the structure of the human enteric nervous system. CONCLUSIONS Such findings have sparked therapeutic trials for the treatment of gastrointestinal disorders in patients.
Collapse
Affiliation(s)
- David A Wattchow
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Departments of Surgery and Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - David Smolilo
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Departments of Surgery and Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Tim Hibberd
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Nick J Spencer
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Simon Jh Brookes
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| | - Paul T Heitmann
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Departments of Surgery and Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| | - Marcello Costa
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Phil G Dinning
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.,Departments of Surgery and Gastroenterology, Flinders Medical Centre, Adelaide, South Australia, Australia
| |
Collapse
|
8
|
Parker DR, Wiklendt L, Humenick A, Chen BN, Sia TC, Wattchow DA, Dinning PG, Brookes SJH. Sympathetic Pathways Target Cholinergic Neurons in the Human Colonic Myenteric Plexus. Front Neurosci 2022; 16:863662. [PMID: 35368277 PMCID: PMC8970288 DOI: 10.3389/fnins.2022.863662] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/21/2022] [Indexed: 01/01/2023] Open
Abstract
Background The sympathetic nervous system inhibits human colonic motility largely by effects on enteric neurons. Noradrenergic axons, which branch extensively in the myenteric plexus, are integral to this modulatory role, but whether they contact specific types of enteric neurons is unknown. The purpose of this study was to determine the association of noradrenergic varicosities with types of enteric neurons. Methods Human colonic tissue from seven patients was fixed and dissected prior to multi-layer immunohistochemistry for human RNA binding proteins C and D (HuC/D) (pan-neuronal cell body labelling), tyrosine hydroxylase (TH, catecholaminergic labelling), Enkephalin (ENK), choline acetyltransferase (ChAT, cholinergic labelling) and/or nitric oxide synthase (NOS, nitrergic labelling) and imaged using confocal microscopy. TH-immunoreactive varicose nerve endings and myenteric cell bodies were reconstructed as three dimensional digital images. Data was exported to a purpose-built software package which quantified the density of varicosities close to the surface of each myenteric cell body. Results TH-immunoreactive varicosities had a greater mean density within 1 μm of the surface of ChAT +/NOS− nerve cell bodies compared with ChAT−/NOS + cell bodies. Similarly, ENK-immunoreactive varicosities also had a greater mean density close to ChAT +/NOS− cell bodies compared with ChAT−/NOS + cells. Conclusion A method for quantifying close associations between varicosities and nerve cell bodies was developed. Sympathetic axons in the myenteric plexus preferentially target cholinergic excitatory cells compared to nitrergic neurons (which are largely inhibitory). This connectivity is likely to be involved in inhibitory modulation of human colonic motility by the sympathetic nervous system.
Collapse
Affiliation(s)
- Dominic R. Parker
- Laboratory of Neurogastroenterology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Lukasz Wiklendt
- Laboratory of Neurogastroenterology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Adam Humenick
- Laboratory of Neurogastroenterology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Bao Nan Chen
- Laboratory of Neurogastroenterology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Tiong Cheng Sia
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, SA, Australia
| | - David A. Wattchow
- Laboratory of Neurogastroenterology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Colorectal Surgical Unit, Division of Surgery, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Phil G. Dinning
- Laboratory of Neurogastroenterology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Simon J. H. Brookes
- Laboratory of Neurogastroenterology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- *Correspondence: Simon J. H. Brookes,
| |
Collapse
|
9
|
Tarif AMM, Islam MN, Jahan MR, Yanai A, Nozaki K, Masumoto KH, Shinoda K. Immunohistochemical expression and neurochemical phenotypes of huntingtin-associated protein 1 in the myenteric plexus of mouse gastrointestinal tract. Cell Tissue Res 2021; 386:533-558. [PMID: 34665322 DOI: 10.1007/s00441-021-03542-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Huntingtin-associated protein 1 (HAP1) is a neural huntingtin interactor and being considered as a core molecule of stigmoid body (STB). Brain/spinal cord regions with abundant STB/HAP1 expression are usually spared from neurodegeneration in stress/disease conditions, whereas the regions with little STB/HAP1 expression are always neurodegenerative targets. The enteric nervous system (ENS) can act as a potential portal for pathogenesis of neurodegenerative disorders. However, ENS is also a neurodegenerative target in these disorders. To date, the expression of HAP1 and its neurochemical characterization have never been examined there. In the current study, we determined the expression of HAP1 in the ENS of adult mice and characterized the morphological relationships of HAP1-immunoreactive (ir) cells with the markers of motor neurons, sensory neurons, and interneurons in the myenteric plexus using Western blotting and light/fluorescence microscopy. HAP1-immunoreaction was present in both myenteric and submucosal plexuses of ENS. Most of the HAP1-ir neurons exhibited STB in their cytoplasm. In myenteric plexus, a large number of calretinin, calbindin, NOS, VIP, ChAT, SP, somatostatin, and TH-ir neurons showed HAP1-immunoreactivity. In contrast, most of the CGRP-ir neurons were devoid of HAP1-immunoreactivity. Our current study is the first to clarify that HAP1 is highly expressed in excitatory motor neurons, inhibitory motor neurons, and interneurons but almost absent in sensory neurons in myenteric plexus. These suggest that STB/HAP1-ir neurons are mostly Dogiel type I neurons. Due to lack of putative STB/HAP1 protectivity, the sensory neurons (Dogiel type II) might be more vulnerable to neurodegeneration than STB/HAP1-expressing motoneurons/interneurons (Dogiel type I) in myenteric plexus.
Collapse
Affiliation(s)
- Abu Md Mamun Tarif
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
| | - Md Nabiul Islam
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
| | - Mir Rubayet Jahan
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
- Department of Anatomy and Histology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Akie Yanai
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
- Department of Basic Laboratory Sciences, Faculty of Medicine and Health Sciences, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
| | - Kanako Nozaki
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
| | - Koh-Hei Masumoto
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan
| | - Koh Shinoda
- Division of Neuroanatomy, Department of Neuroscience, Yamaguchi University Graduate School of Medicine, 1-1-1 Minami-Kogushi, Ube, 755-8505, Japan.
| |
Collapse
|
10
|
Interactions between the microbiota and enteric nervous system during gut-brain disorders. Neuropharmacology 2021; 197:108721. [PMID: 34274348 DOI: 10.1016/j.neuropharm.2021.108721] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 02/08/2023]
Abstract
For the last 20 years, researchers have focused their intention on the impact of gut microbiota in healthy and pathological conditions. This year (2021), more than 25,000 articles can be retrieved from PubMed with the keywords "gut microbiota and physiology", showing the constant progress and impact of gut microbes in scientific life. As a result, numerous therapeutic perspectives have been proposed to modulate the gut microbiota composition and/or bioactive factors released from microbes to restore our body functions. Currently, the gut is considered a primary site for the development of pathologies that modify brain functions such as neurodegenerative (Parkinson's, Alzheimer's, etc.) and metabolic (type 2 diabetes, obesity, etc.) disorders. Deciphering the mode of interaction between microbiota and the brain is a real original option to prevent (and maybe treat in the future) the establishment of gut-brain pathologies. The objective of this review is to describe recent scientific elements that explore the communication between gut microbiota and the brain by focusing our interest on the enteric nervous system (ENS) as an intermediate partner. The ENS, which is known as the "second brain", could be under the direct or indirect influence of the gut microbiota and its released factors (short-chain fatty acids, neurotransmitters, gaseous factors, etc.). Thus, in addition to their actions on tissue (adipose tissue, liver, brain, etc.), microbes can have an impact on local ENS activity. This potential modification of ENS function has global repercussions in the whole body via the gut-brain axis and represents a new therapeutic strategy.
Collapse
|
11
|
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
|
12
|
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: 5.3] [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
|
13
|
Mazzoni M, Caremoli F, Cabanillas L, de Los Santos J, Million M, Larauche M, Clavenzani P, De Giorgio R, Sternini C. Quantitative analysis of enteric neurons containing choline acetyltransferase and nitric oxide synthase immunoreactivities in the submucosal and myenteric plexuses of the porcine colon. Cell Tissue Res 2020; 383:645-654. [PMID: 32965550 DOI: 10.1007/s00441-020-03286-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022]
Abstract
The enteric nervous system (ENS) controls gastrointestinal functions. In large mammals' intestine, it comprises an inner (ISP) and outer (OSP) submucous plexus and a myenteric plexus (MP). This study quantifies enteric neurons in the ISP, OSP, and MP of the pig ascending (AC) and descending colon (DC) using the HuC/D, choline acetyltransferase (ChAT), and neuronal nitric oxide synthase (nNOS) neuronal markers in whole mount preparations with multiple labeling immunofluorescence. We established that the ISP contains the highest number of HuC/D neurons/mm2, which were more abundant in AC vs. DC, followed by OSP and MP with similar density in AC and DC. In the ISP, the density of ChAT immunoreactive (IR) neurons was very similar in AC and DC (31% and 35%), nNOS-IR neurons were less abundant in AC than DC (15% vs. 42%, P < 0.001), and ChAT/nNOS-IR neurons were 5% and 10%, respectively. In the OSP, 39-44% of neurons were ChAT-IR in AC and DC, while 45% and 38% were nNOS-IR and 10-12% were ChAT/nNOS-IR (AC vs. DC P < 0.05). In the MP, ChAT-IR neurons were 44% in AC and 54% in DC (P < 0.05), nNOS-IR neurons were 50% in both, and ChAT/nNOS-IR neurons were 12 and 18%, respectively. The ENS architecture with multilayered submucosal plexuses and the distribution of functionally distinct groups of neurons in the pig colon are similar to humans, supporting the suitability of the pig as a model and providing the platform for investigating the mechanisms underlying human colonic diseases.
Collapse
Affiliation(s)
- Maurizio Mazzoni
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, Bologna, Italy
| | - Filippo Caremoli
- Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Luis Cabanillas
- Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Janira de Los Santos
- Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Mulugeta Million
- Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.,Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, 90095, USA
| | - Muriel Larauche
- Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Paolo Clavenzani
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, 40064, Bologna, Italy
| | - Roberto De Giorgio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Catia Sternini
- Division of Digestive Diseases, Department Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA. .,Department of Neurobiology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
14
|
Graham KD, López SH, Sengupta R, Shenoy A, Schneider S, Wright CM, Feldman M, Furth E, Valdivieso F, Lemke A, Wilkins BJ, Naji A, Doolin E, Howard MJ, Heuckeroth RO. Robust, 3-Dimensional Visualization of Human Colon Enteric Nervous System Without Tissue Sectioning. Gastroenterology 2020; 158:2221-2235.e5. [PMID: 32113825 PMCID: PMC7392351 DOI: 10.1053/j.gastro.2020.02.035] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Small, 2-dimensional sections routinely used for human pathology analysis provide limited information about bowel innervation. We developed a technique to image human enteric nervous system (ENS) and other intramural cells in 3 dimensions. METHODS Using mouse and human colon tissues, we developed a method that combines tissue clearing, immunohistochemistry, confocal microscopy, and quantitative analysis of full-thickness bowel without sectioning to quantify ENS and other intramural cells in 3 dimensions. RESULTS We provided 280 adult human colon confocal Z-stacks from persons without known bowel motility disorders. Most of our images were of myenteric ganglia, captured using a 20× objective lens. Full-thickness colon images, viewed with a 10× objective lens, were as large as 4 × 5 mm2. Colon from 2 pediatric patients with Hirschsprung disease was used to show distal colon without enteric ganglia, as well as a transition zone and proximal pull-through resection margin where ENS was present. After testing a panel of antibodies with our method, we identified 16 antibodies that bind to molecules in neurons, glia, interstitial cells of Cajal, and muscularis macrophages. Quantitative analyses demonstrated myenteric plexus in 24.5% ± 2.4% of flattened colon Z-stack area. Myenteric ganglia occupied 34% ± 4% of myenteric plexus. Single myenteric ganglion volume averaged 3,527,678 ± 573,832 mm3 with 38,706 ± 5763 neuron/mm3 and 129,321 ± 25,356 glia/mm3. Images of large areas provided insight into why published values of ENS density vary up to 150-fold-ENS density varies greatly, across millimeters, so analyses of small numbers of thin sections from the same bowel region can produce varying results. Neuron subtype analysis revealed that approximately 56% of myenteric neurons stained with neuronal nitric oxide synthase antibody and approximately 33% of neurons produce and store acetylcholine. Transition zone regions from colon tissues of patients with Hirschsprung disease had ganglia in multiple layers and thick nerve fiber bundles without neurons. Submucosal neuron distribution varied among imaged colon regions. CONCLUSIONS We developed a 3-dimensional imaging method for colon that provides more information about ENS structure than tissue sectioning. This approach could improve diagnosis for human bowel motility disorders and may be useful for other bowel diseases as well.
Collapse
Affiliation(s)
- Kahleb D. Graham
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Cincinnati Children’s Hospital Medical Center and the Department of Pediatrics at University of Cincinnati College of Medicine, Cincinnati, OH 45229
| | - Silvia Huerta López
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318
| | - Rajarshi Sengupta
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,American Association for Cancer Research, 615 Chestnut Street, 17th Floor, Philadelphia, PA 19106-4404
| | - Archana Shenoy
- Department of Pathology, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A., 19104-4318
| | - Sabine Schneider
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| | - Christina M. Wright
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| | - Michael Feldman
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Emma Furth
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Federico Valdivieso
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, University of Pennsylvania Medical Center, 3400 Spruce Street, Philadelphia, PA, U.S.A., 19104-4238
| | - Amanda Lemke
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318
| | - Benjamin J. Wilkins
- Department of Pathology, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A., 19104-4318
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104-4318
| | - Edward Doolin
- Pediatric General, Thoracic and Fetal Surgery, The Children’s Hospital of Philadelphia, 3401 Civic Center Boulevard, Philadelphia, PA, U.S.A. 19104-4318
| | - Marthe J. Howard
- Department of Neurosciences, University of Toledo, Mail Stop # 1007, 3000 Arlington Avenue, Toledo, OH, U.S.A, 43614-2598
| | - Robert O. Heuckeroth
- Children’s Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center – Suite # 1116I, Philadelphia, PA, U.S.A., 19104-4318,Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, 3401 Civic Center Boulevard, Philadelphia, PA, 19104-4318
| |
Collapse
|
15
|
Boschetti E, Malagelada C, Accarino A, Malagelada JR, Cogliandro RF, Gori A, Bonora E, Giancola F, Bianco F, Tugnoli V, Clavenzani P, Azpiroz F, Stanghellini V, Sternini C, De Giorgio R. Enteric neuron density correlates with clinical features of severe gut dysmotility. Am J Physiol Gastrointest Liver Physiol 2019; 317:G793-G801. [PMID: 31545923 PMCID: PMC6962493 DOI: 10.1152/ajpgi.00199.2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Gastrointestinal (GI) symptoms can originate from severe dysmotility due to enteric neuropathies. Current methods used to demonstrate enteric neuropathies are based mainly on classic qualitative histopathological/immunohistochemical evaluation. This study was designed to identify an objective morphometric method for paraffin-embedded tissue samples to quantify the interganglionic distance between neighboring myenteric ganglia immunoreactive for neuron-specific enolase, as well as the number of myenteric and submucosal neuronal cell bodies/ganglion in jejunal specimens of patients with severe GI dysmotility. Jejunal full-thickness biopsies were collected from 32 patients (22 females; 16-77 yr) with well-characterized severe dysmotility and 8 controls (4 females; 47-73 yr). A symptom questionnaire was filled before surgery. Mann-Whitney U test, Kruskal-Wallis coupled with Dunn's posttest and nonparametric linear regression tests were used for analyzing morphometric data and clinical correlations, respectively. Compared with controls, patients with severe dysmotility exhibited a significant increase in myenteric interganglionic distance (P = 0.0005) along with a decrease in the number of myenteric (P < 0.00001) and submucosal (P < 0.0004) neurons. A 50% reduction in the number of submucosal and myenteric neurons correlated with an increased interganglionic distance and severity of dysmotility. Our study proposes a relatively simple tool that can be applied for quantitative evaluation of paraffin sections from patients with severe dysmotility. The finding of an increased interganglionic distance may aid diagnosis and limit the direct quantitative analysis of neurons per ganglion in patients with an interganglionic distance within the control range.NEW & NOTEWORTHY Enteric neuropathies are challenging conditions characterized by a severe impairment of gut physiology, including motility. An accurate, unambiguous assessment of enteric neurons provided by quantitative analysis of routine paraffin sections may help to define neuropathy-related gut dysmotility. We showed that patients with severe gut dysmotility exhibited an increased interganglionic distance associated with a decreased number of myenteric and submucosal neurons, which correlated with symptoms and clinical manifestations of deranged intestinal motility.
Collapse
Affiliation(s)
- Elisa Boschetti
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Carolina Malagelada
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | - Anna Accarino
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | - Juan R. Malagelada
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | | | - Alessandra Gori
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elena Bonora
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Fiorella Giancola
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Francesca Bianco
- 1Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Vitaliano Tugnoli
- 3Department of Biomedical and Neuro Motor Sciences, University of Bologna, Bologna, Italy
| | - Paolo Clavenzani
- 4Department of Veterinary Medicine, University of Bologna, Ozzano, Italy
| | - Fernando Azpiroz
- 2Digestive System Research Unit, University Hospital Vall d'Hebron, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), Barcelona, Spain
| | | | - Catia Sternini
- 5Digestive Disease Division, Departments of Medicine and Neurobiology, University of California, Los Angeles, California
| | | |
Collapse
|
16
|
Humenick A, Chen BN, Lauder CIW, Wattchow DA, Zagorodnyuk VP, Dinning PG, Spencer NJ, Costa M, Brookes SJH. Characterization of projections of longitudinal muscle motor neurons in human colon. Neurogastroenterol Motil 2019; 31:e13685. [PMID: 31355986 DOI: 10.1111/nmo.13685] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 07/07/2019] [Accepted: 07/08/2019] [Indexed: 12/08/2022]
Abstract
BACKGROUND The enteric nervous system contains inhibitory and excitatory motor neurons which modulate smooth muscle contractility. Cell bodies of longitudinal muscle motor neurons have not been identified in human intestine. METHODS We used retrograde tracing ex vivo with DiI, with multiple labeling immunohistochemistry, to characterize motor neurons innervating tenial and inter-tenial longitudinal muscle of human colon. KEY RESULTS The most abundant immunohistochemical markers in the tertiary plexus were vesicular acetylcholine transporter, nitric oxide synthase (NOS), and vasoactive intestinal polypeptide (VIP). Of retrogradely traced motor neurons innervating inter-tenial longitudinal muscle, 95% were located within 6mm oral or anal to the DiI application site. Excitatory motor neuron cell bodies, immunoreactive for choline acetyltransferase (ChAT), were clustered aborally, whereas NOS-immunoreactive cell bodies were distributed either side of the DiI application site. Motor neurons had small cell bodies, averaging 438 + 18µm2 in cross-sectional area, similar for ChAT- and NOS-immunoreactive subtypes. Motor neurons innervating the tenia had slightly longer axial projections, with 95% located within 9mm. ChAT-immunoreactive excitatory motor neurons to tenia were clustered aborally, whereas NOS-immunoreactive inhibitory motor neurons had both ascending and descending projections. VIP immunoreactivity was rarely present without NOS immunoreactivity in motor neurons. CONCLUSIONS AND INFERENCES Tenial and inter-tenial motor neurons innervating the longitudinal muscle have short projections. Inhibitory motor neurons have less polarized projections than cholinergic excitatory motor neurons. Longitudinal and circular muscle layers are innervated by distinct local populations of excitatory and inhibitory motor neurons. A population of human enteric neurons that contribute significantly to colonic motility has been characterized.
Collapse
Affiliation(s)
- Adam Humenick
- Human Physiology, Medical Bioscience, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Bao Nan Chen
- Human Physiology, Medical Bioscience, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Chris I W Lauder
- Department of Surgery, Flinders Medical Centre, Adelaide, SA, Australia
| | - David A Wattchow
- Department of Surgery, Flinders Medical Centre, Adelaide, SA, Australia
| | - Vladimir P Zagorodnyuk
- Human Physiology, Medical Bioscience, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Phil G Dinning
- Department of Surgery, Flinders Medical Centre, Adelaide, SA, Australia
| | - Nick J Spencer
- Human Physiology, Medical Bioscience, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Marcello Costa
- Human Physiology, Medical Bioscience, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Simon J H Brookes
- Human Physiology, Medical Bioscience, Centre for Neuroscience, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
17
|
Anetsberger D, Kürten S, Jabari S, Brehmer A. Morphological and Immunohistochemical Characterization of Human Intrinsic Gastric Neurons. Cells Tissues Organs 2019; 206:183-195. [PMID: 31230045 DOI: 10.1159/000500566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 04/24/2019] [Indexed: 11/19/2022] Open
Abstract
Our knowledge about human gastric enteric neuron types is even more limited than that of human intestinal types. Here, we immunohistochemically stained wholemounts and sections of gastric specimens obtained from 18 tumor-resected patients. Myenteric wholemounts were labeled for choline acetyl transferase (ChAT), neuronal nitric oxide synthase (NOS), and the human neuronal protein HuC/D (as pan-neuronal marker for quantitative analysis) or alternatively for neurofilament (for morphological evaluation). ChAT-positive neurons outnumbered NOS-positive neurons (56 vs. 27%), and neurons negative for both markers accounted for 17%. Two larger groups of neurons (each between 12 and 14%) costained for ChAT and vasoactive intestinal peptide (VIP) or for NOS and VIP, respectively. Clear morphochemical correlation was found for uniaxonal stubby type I neurons (ChAT+; putative excitatory inter- or motor neurons), for uniaxonal spiny type I neurons (NOS+/VIP+; putative inhibitory motor or interneurons), and for multiaxonal type II neurons (ChAT+; putative afferent neurons; immunostaining of additional wholemounts revealed their coreactivity for somatostatin). Whereas these latter neuron types were already known from the human intestine, the morphology of gastric myenteric neurons coreactive for ChAT and VIP was newly described: they had numerous short, extremely thin dendrites and resembled, together with their cell bodies, a "hairy" head. In our sections, nerve fibers coreactive for ChAT and VIP were commonly found only in the mucosa. We suggest these myenteric ChAT+/VIP+/hairy neurons to be mucosal effector neurons. In contrast to myenteric neurons, the much less common submucosal neurons were not embedded in a continuous plexus and did not display any clear morphochemical phenotypes.
Collapse
Affiliation(s)
- Daniel Anetsberger
- Institute of Anatomy and Cell Biology, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Stefanie Kürten
- Institute of Anatomy and Cell Biology, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Samir Jabari
- Institute of Neuropathology, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Axel Brehmer
- Institute of Anatomy and Cell Biology, Friedrich Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany,
| |
Collapse
|
18
|
Ng KS, Montes-Adrian NA, Mahns DA, Gladman MA. Quantification and neurochemical coding of the myenteric plexus in humans: No regional variation between the distal colon and rectum. Neurogastroenterol Motil 2018; 30. [PMID: 28836741 DOI: 10.1111/nmo.13193] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/28/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND It remains unclear whether regional variation exists in the human enteric nervous system (ENS) ie, whether intrinsic innervation varies along the gut. Recent classification of gastrointestinal neuropathies has highlighted inadequacies in the quantification of the human ENS. This study used paired wholemounts to accurately quantify and neurochemically code the hindgut myenteric plexus, comparing human distal colon and rectum. METHODS Paired human descending colonic/rectal specimens were procured from 15 patients undergoing anterior resection. Wholemounts of myenteric plexi were triple-immunostained with anti-Hu/NOS/ChAT antibodies. Images were acquired by motorized epifluorescence microscopy, allowing assessment of ganglionic density/size, ganglionic area density, and neuronal density. 'Stretch-corrected' values were calculated using stretched/relaxed tissue dimensions. KEY RESULTS Tile-stitching created a collage with average area 99 300 000 μm2 . Stretch-corrected ganglionic densities were similar (colon: median 510 ganglia/100 mm2 [range 386-1170], rectum: 585 [307-923]; P = .99), as were average ganglionic sizes (colon: 57 593 μm2 [40 301-126 579], rectum: 54 901 [38 701-90 211], P = .36). Ganglionic area density (colon: 11.92 mm2 per 100 mm2 [7.53-18.64], rectum: 9.84 [5.80-17.19], P = .10) and stretch-corrected neuronal densities (colon: 189 neurons/mm2 [117-388], rectum: 182 [89-361], P = .31) were also similar, as were the neurochemical profiles of myenteric ganglia, with comparable proportions of NOS+ and ChAT+ neurons (P > .10). CONCLUSIONS AND INFERENCES This study has revealed similar neuronal and ganglionic densities and neurochemical profiles in human distal colon and rectum. Further investigation of other components of the ENS, incorporating additional immunohistochemical markers are required to confirm that there is no regional variation in the human hindgut ENS.
Collapse
Affiliation(s)
- K-S Ng
- Academic Colorectal Unit, Sydney Medical School - Concord, University of Sydney, Concord, New South Wales, Australia
| | - N A Montes-Adrian
- Academic Colorectal Unit, Sydney Medical School - Concord, University of Sydney, Concord, New South Wales, Australia
| | - D A Mahns
- Department of Integrative Physiology, School of Medicine, University of Western Sydney, Campbelltown, New South Wales, Australia
| | - M A Gladman
- Academic Colorectal Unit, Sydney Medical School - Concord, University of Sydney, Concord, New South Wales, Australia.,Enteric Neuroscience and Gastrointestinal Research Group, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
19
|
Zséli G, Vida B, Szilvásy-Szabó A, Tóth M, Lechan RM, Fekete C. Neuronal connections of the central amygdalar nucleus with refeeding-activated brain areas in rats. Brain Struct Funct 2018; 223:391-414. [PMID: 28852859 PMCID: PMC5773374 DOI: 10.1007/s00429-017-1501-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 08/18/2017] [Indexed: 12/22/2022]
Abstract
Following fasting, satiety is accompanied by neuronal activation in brain areas including the central amygdalar nucleus (CEA). Since CEA is known to inhibit food intake, we hypothesized that CEA contributes to the termination of meal during refeeding. To better understand the organization of this satiety-related circuit, the interconnections of the CEA with refeeding-activated neuronal groups were elucidated using retrograde (cholera toxin-β subunit, CTB) and anterograde (phaseolus vulgaris leucoagglutinin, PHA-L) tracers in male rats. C-Fos-immunoreactivity was used as marker of neuronal activation. The refeeding-activated input of the CEA primarily originated from the paraventricular thalamic, parasubthalamic and parabrachial nuclei. Few CTB-c-Fos double-labeled neurons were detected in the prefrontal cortex, lateral hypothalamic area, nucleus of the solitary tract (NTS) and the bed nuclei of the stria terminalis (BNST). Only few refeeding-activated proopiomelanocortin-producing neurons of the arcuate nucleus projected to the CEA. Anterograde tract tracing revealed a high density of PHAL-labeled axons contacted with refeeding-activated neurons in the BNST, lateral hypothalamic area, parasubthalamic, paraventricular thalamic and parabrachial nuclei and NTS; a low density of labeled axons was found in the paraventricular hypothalamic nucleus. Chemogenetic activation of the medial CEA (CEAm) inhibited food intake during the first hour of refeeding, while activation of lateral CEA had no effect. These data demonstrate the existence of reciprocal connections between the CEA and distinct refeeding-activated hypothalamic, thalamic and brainstem nuclei, suggesting the importance of short feedback loops in the regulation of satiety and importance of the CEAm in the regulation of food intake during refeeding.
Collapse
Affiliation(s)
- Györgyi Zséli
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St, Budapest, 1083, Hungary
- Neuroendocrinology Program, Semmelweis University Neurosciences Doctoral School, Budapest, Hungary
| | - Barbara Vida
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St, Budapest, 1083, Hungary
- Department of Neuroscience, Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, Hungary
| | - Anett Szilvásy-Szabó
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St, Budapest, 1083, Hungary
- Neuroendocrinology Program, Semmelweis University Neurosciences Doctoral School, Budapest, Hungary
| | - Mónika Tóth
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St, Budapest, 1083, Hungary
| | - Ronald M Lechan
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Tupper Research Institute, Tufts Medical Center, Boston, MA, USA
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Csaba Fekete
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, 43 Szigony St, Budapest, 1083, Hungary.
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Tupper Research Institute, Tufts Medical Center, Boston, MA, USA.
| |
Collapse
|
20
|
SchÄfer BT, Silveira MP, Palombit K, Mendes CE, Watanabe IS, Miglino MA, Castelucci P. Morphological Characterization of the Myenteric Plexus of the Ileum and Distal colon of Dogs Affected by Muscular Dystrophy. Anat Rec (Hoboken) 2017; 301:673-685. [DOI: 10.1002/ar.23708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Revised: 02/02/2017] [Accepted: 05/03/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Bárbara Tavares SchÄfer
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of São Paulo; Brazil
| | - Mariana Póvoa Silveira
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of São Paulo; Brazil
| | - Kelly Palombit
- Department of Anatomy/Biomedical Sciences Institute; University of São Paulo; Brazil
- Deparment of Morphology; University Federal do Piaui; Teresina-PI Brazil
| | | | - Ii Sei Watanabe
- Department of Anatomy/Biomedical Sciences Institute; University of São Paulo; Brazil
| | - Maria Angélica Miglino
- Department of Surgery, Faculty of Veterinary Medicine and Animal Science; University of São Paulo; Brazil
| | - Patricia Castelucci
- Department of Anatomy/Biomedical Sciences Institute; University of São Paulo; Brazil
| |
Collapse
|
21
|
Kapoor A, Auer DR, Lee D, Chatterjee S, Chakravarti A. Testing the Ret and Sema3d genetic interaction in mouse enteric nervous system development. Hum Mol Genet 2017; 26:1811-1820. [PMID: 28334784 DOI: 10.1093/hmg/ddx084] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/02/2017] [Indexed: 12/20/2022] Open
Abstract
For most multigenic disorders, clinical manifestation (penetrance) and presentation (expressivity) are likely to be an outcome of genetic interaction between multiple susceptibility genes. Here, using gene knockouts in mice, we evaluated genetic interaction between loss of Ret and loss of Sema3d, two Hirschsprung disease susceptibility genes. We intercrossed Ret and Sema3d double null heterozygotes to generate mice with the nine possible genotypes and assessed survival by counting various genotypes, myenteric plexus presence by acetylcholinesterase staining and embryonic day 12.5 (E12.5) intestine transcriptome by RNA-sequencing. Survival rates of Ret wild-type, null heterozygote and null homozygote mice at E12.5, birth and weaning were not influenced by the genotypes at Sema3d locus and vice versa. Loss of myenteric plexus was observed only in all Ret null homozygotes, irrespective of the genotypes at Sema3d locus, and Sema3d null heterozygote and homozygote mice had normal intestinal innervation. As compared with wild-type mice intestinal gene expression, loss of Ret in null homozygotes led to differential expression of ∼300 genes, whereas loss of Sema3d in null homozygotes had no major consequence and there was no evidence supporting major interaction between the two genes influencing intestine transcriptome. Overall, given the null alleles and phenotypic assays used, we did not find evidence for genetic interaction between Ret and Sema3d affecting survival, presence of myenteric plexus or intestine transcriptome.
Collapse
Affiliation(s)
- Ashish Kapoor
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dallas R Auer
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Dongwon Lee
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sumantra Chatterjee
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Aravinda Chakravarti
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
22
|
Hutson JM, Farmer PJ, Peck CJ, Chow CW, Southwell BR. Multiple endocrine neoplasia 2B: Differential increase in enteric nerve subgroups in muscle and mucosa. World J Gastrointest Pathophysiol 2017; 8:142-149. [PMID: 28868184 PMCID: PMC5561435 DOI: 10.4291/wjgp.v8.i3.142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 05/24/2017] [Accepted: 07/17/2017] [Indexed: 02/06/2023] Open
Abstract
Multiple endocrine neoplasia 2B (MEN2B) is a rare syndrome caused by an activating mutation of the RET gene, leading to enteric gangliomatosis. This child presented with constipation at 1-mo old, was diagnosed with MEN2B by rectal biopsy at 4 mo, had thyroidectomy at 9 mo and a colectomy at 4 years. We studied the extent of neuronal and nerve fibre proliferation and which classes of enteric nerves are affected by examining the colon with multiple neuronal antibodies. Resected transverse colon was fixed, frozen, sectioned and processed for fluorescence immunohistochemistry labelling with antibodies against TUJ1, Hu, ChAT, NOS, VIP, SP and CGRP and cKit. Control transverse colon was from the normal margin of Hirschsprung (HSCR) colon (4-year-old) and a child with familial adenomatous polyposis (FAP, 12 year). Myenteric ganglia were increased in size to as wide as the circular muscle. There was a large increase in nerve cells and nerve fibres. ChAT-, NOS-, VIP- and SP-immunoreactive nerve fibres all increased in the myenteric ganglia. NOS-IR nerves preferentially increased in the muscle, while VIP and SP increased in submucosal ganglia and mucosal nerve fibres. The density of ICC was normal. RET overactivation in MEN2B lead to a large increase in intrinsic nerve fibres in the myenteric and submucosal ganglia, with a relative increase in NOS-IR nerve fibres in the circular muscle and VIP and SP in the submucosal ganglia and mucosa. The changes were associated with severe constipation resulting in colectomy at 4 years.
Collapse
|
23
|
Reichardt F, Chassaing B, Nezami BG, Li G, Tabatabavakili S, Mwangi S, Uppal K, Liang B, Vijay-Kumar M, Jones D, Gewirtz AT, Srinivasan S. Western diet induces colonic nitrergic myenteric neuropathy and dysmotility in mice via saturated fatty acid- and lipopolysaccharide-induced TLR4 signalling. J Physiol 2017; 595:1831-1846. [PMID: 28000223 PMCID: PMC5330876 DOI: 10.1113/jp273269] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/01/2016] [Indexed: 01/01/2023] Open
Abstract
KEY POINTS A high-fat diet (60% kcal from fat) is associated with motility disorders inducing constipation and loss of nitrergic myenteric neurons in the proximal colon. Gut microbiota dysbiosis, which occurs in response to HFD, contributes to endotoxaemia. High levels of lipopolysaccharide lead to apoptosis in cultured myenteric neurons that express Toll-like receptor 4 (TLR4). Consumption of a Western diet (WD) (35% kcal from fat) for 6 weeks leads to gut microbiota dysbiosis associated with altered bacterial metabolites and increased levels of plasma free fatty acids. These disorders precede the nitrergic myenteric cell loss observed in the proximal colon. Mice lacking TLR4 did not exhibit WD-induced myenteric cell loss and dysmotility. Lipopolysaccharide-induced in vitro enteric neurodegeneration requires the presence of palmitate and may be a result of enhanced NO production. The present study highlights the critical role of plasma saturated free fatty acids that are abundant in the WD with respect to driving enteric neuropathy and colonic dysmotility. ABSTRACT The consumption of a high-fat diet (HFD) is associated with myenteric neurodegeneration, which in turn is associated with delayed colonic transit and constipation. We examined the hypothesis that an inherent increase in plasma free fatty acids (FFA) in the HFD together with an HFD-induced alteration in gut microbiota contributes to the pathophysiology of these disorders. C57BL/6 mice were fed a Western diet (WD) (35% kcal from fat enriched in palmitate) or a purified regular diet (16.9% kcal from fat) for 3, 6, 9 and 12 weeks. Gut microbiota dysbiosis was investigated by fecal lipopolysaccharide (LPS) measurement and metabolomics (linear trap quadrupole-Fourier transform mass spectrometer) analysis. Plasma FFA and LPS levels were assessed, in addition to colonic and ileal nitrergic myenteric neuron quantifications and motility. Compared to regular diet-fed control mice, WD-fed mice gained significantly more weight without blood glucose alteration. Dysbiosis was exhibited after 6 weeks of feeding, as reflected by increased fecal LPS and bacterial metabolites and concomitant higher plasma FFA. The numbers of nitrergic myenteric neurons were reduced in the proximal colon after 9 and 12 weeks of WD and this was also associated with delayed colonic transit. WD-fed Toll-like receptor 4 (TLR4)-/- mice did not exhibit myenteric cell loss or dysmotility. Finally, LPS (0.5-2 ng·ml-1 ) and palmitate (20 and 30 μm) acted synergistically to induce neuronal cell death in vitro, which was prevented by the nitric oxide synthase inhibitor NG-nitro-l-arginine methyl ester. In conclusion, WD-feeding results in increased levels of FFA and microbiota that, even in absence of hyperglycaemia or overt endotoxaemia, synergistically induce TLR4-mediated neurodegeneration and dysmotility.
Collapse
Affiliation(s)
- François Reichardt
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta & Atlanta VA Medical Center, Decatur, GA, USA
| | - Benoit Chassaing
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, GA, USA
| | - Behtash Ghazi Nezami
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta & Atlanta VA Medical Center, Decatur, GA, USA
| | - Ge Li
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta & Atlanta VA Medical Center, Decatur, GA, USA
| | - Sahar Tabatabavakili
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta & Atlanta VA Medical Center, Decatur, GA, USA
| | - Simon Mwangi
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta & Atlanta VA Medical Center, Decatur, GA, USA
| | - Karan Uppal
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, GA, USA
| | - Bill Liang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, GA, USA
| | - Matam Vijay-Kumar
- Department of Nutritional Sciences & Medicine, Pennsylvania State University, University Park, PA, USA
| | - Dean Jones
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Emory University, GA, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, GA, USA
| | - Shanthi Srinivasan
- Department of Digestive Diseases, Emory University School of Medicine, Atlanta & Atlanta VA Medical Center, Decatur, GA, USA
| |
Collapse
|
24
|
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: 1.1] [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
|
25
|
Cheng LS, Schwartz DM, Hotta R, Graham HK, Goldstein AM. Bowel dysfunction following pullthrough surgery is associated with an overabundance of nitrergic neurons in Hirschsprung disease. J Pediatr Surg 2016; 51:1834-1838. [PMID: 27570241 PMCID: PMC5065396 DOI: 10.1016/j.jpedsurg.2016.08.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Revised: 08/01/2016] [Accepted: 08/01/2016] [Indexed: 12/29/2022]
Abstract
PURPOSE Recent evidence suggests that patients with Hirschsprung disease (HD) have abnormal neurotransmitter expression in the ganglionated proximal colon. These alterations may cause persistent bowel dysfunction even after pullthrough surgery. We sought to quantify the proportion of nitrergic neurons in the ganglionic colon of HD patients and relate these findings to functional outcome. METHODS The proximal resection margin from 17 patients with colonic HD who underwent a pullthrough procedure and colorectal tissue from 4 age-matched controls were immunohistochemically examined to quantify the proportion of nitrergic neurons. The incidence of constipation, incontinence, and enterocolitis in HD patients was assessed retrospectively and correlated with the proportion of nitric oxide synthase (NOS) expressing neurons. Neuronal subtypes in the ganglionic colon of the Edrnb-/- mouse model of HD were also studied. RESULTS Mice with HD had a significantly higher proportion of NOS+ neurons in ganglionic colon than normal littermates (32.0±5.6% vs. 19.8±1.2%, p<0.01). Patients with HD also had significantly more NOS+ neurons than controls (18.4±4.6% vs. 13.1±1.9%, p<0.01). Patients who experienced constipation or enterocolitis postoperatively tended toward a higher proportion of NOS+ neurons (21.4±3.9% vs. 17.1±4.1%, p=0.06). Furthermore, patients with a proportion of NOS+ neurons above the median of all HD patients (18.3%) were significantly more likely to have constipation than those below the median (75% vs. 14%, p<0.05). CONCLUSION An overabundance of nitrergic neurons in the proximal resection margin is associated with HD and may predict bowel dysfunction following pullthrough surgery.
Collapse
Affiliation(s)
- Lily S Cheng
- Division of Pediatric Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114; Department of Surgery, University of California San Francisco, 500 Parnassus Ave, San Francisco, CA 94143
| | - Dana M Schwartz
- Division of Pediatric Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114
| | - Ryo Hotta
- Division of Pediatric Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114
| | - Hannah K Graham
- Division of Pediatric Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114
| | - Allan M Goldstein
- Division of Pediatric Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114.
| |
Collapse
|
26
|
Carbone SE, Jovanovska V, Brookes SJH, Nurgali K. Electrophysiological and morphological changes in colonic myenteric neurons from chemotherapy-treated patients: a pilot study. Neurogastroenterol Motil 2016; 28:975-84. [PMID: 26909894 PMCID: PMC5215581 DOI: 10.1111/nmo.12795] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 01/14/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Patients receiving anticancer chemotherapy experience a multitude of gastrointestinal side-effects. However, the causes of these symptoms are uncertain and whether these therapeutics directly affect the enteric nervous system is unknown. Our aim was to determine whether the function and morphology of myenteric neurons are altered in specimens of the colon from chemotherapy-treated patients. METHODS Colon specimens were compared from chemotherapy-treated and non-treated patients following colorectal resections for removal of carcinoma. Intracellular electrophysiological recordings from myenteric neurons and immunohistochemistry were performed in whole mount preparations. KEY RESULTS Myenteric S neurons from chemotherapy-treated patients were hyperexcitable; more action potentials (11.4 ± 9.4, p < 0.05) were fired in response to depolarising current pulses than in non-treated patients (1.4 ± 0.5). The rheobase and the threshold to evoke action potentials were significantly lower for neurons from chemotherapy-treated patients compared to neurons from non-treated patients (p < 0.01). Fast excitatory postsynaptic potential reversal potential was more positive in neurons from chemotherapy-treated patients (p < 0.05). An increase in the number of neurons with translocation of Hu protein from the cytoplasm to the nucleus was observed in specimens from chemotherapy-treated patients (103 ± 25 neurons/mm(2) , 37.2 ± 7.0%, n = 8) compared to non-treated (26 ± 5 neurons/mm(2) , 11.9 ± 2.7%, n = 12, p < 0.01). An increase in the soma size of neuronal nitric oxide synthase-immunoreactive neurons was also observed in these specimens. CONCLUSIONS & INFERENCES This is the first study suggesting functional and structural changes in human myenteric neurons in specimens of colon from patients receiving anticancer chemotherapy. These changes may contribute to the causation of gastrointestinal symptoms experienced by chemotherapy-treated patients.
Collapse
Affiliation(s)
- S. E. Carbone
- Centre for Chronic DiseaseCollege of Health and BiomedicineVictoria UniversityMelbourneVICAustralia
| | - V. Jovanovska
- Centre for Chronic DiseaseCollege of Health and BiomedicineVictoria UniversityMelbourneVICAustralia
| | - S. J. H. Brookes
- Discipline of Human Physiology and Centre for NeuroscienceFlinders UniversityAdelaideSAAustralia
| | - K. Nurgali
- Centre for Chronic DiseaseCollege of Health and BiomedicineVictoria UniversityMelbourneVICAustralia
| |
Collapse
|
27
|
Zséli G, Vida B, Martinez A, Lechan RM, Khan AM, Fekete C. Elucidation of the anatomy of a satiety network: Focus on connectivity of the parabrachial nucleus in the adult rat. J Comp Neurol 2016; 524:2803-27. [PMID: 26918800 DOI: 10.1002/cne.23992] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 10/09/2015] [Accepted: 02/22/2016] [Indexed: 01/01/2023]
Abstract
We hypothesized that brain regions showing neuronal activation after refeeding comprise major nodes in a satiety network, and tested this hypothesis with two sets of experiments. Detailed c-Fos mapping comparing fasted and refed rats was performed to identify candidate nodes of the satiety network. In addition to well-known feeding-related brain regions such as the arcuate, dorsomedial, and paraventricular hypothalamic nuclei, lateral hypothalamic area, parabrachial nucleus (PB), nucleus of the solitary tract and central amygdalar nucleus, other refeeding activated regions were also identified, such as the parastrial and parasubthalamic nuclei. To begin to understand the connectivity of the satiety network, the interconnectivity of PB with other refeeding-activated neuronal groups was studied following administration of anterograde or retrograde tracers into the PB. After allowing for tracer transport time, the animals were fasted and then refed before sacrifice. Refeeding-activated neurons that project to the PB were found in the agranular insular area; bed nuclei of terminal stria; anterior hypothalamic area; arcuate, paraventricular, and dorsomedial hypothalamic nuclei; lateral hypothalamic area; parasubthalamic nucleus; central amygdalar nucleus; area postrema; and nucleus of the solitary tract. Axons originating from the PB were observed to closely associate with refeeding-activated neurons in the agranular insular area; bed nuclei of terminal stria; anterior hypothalamus; paraventricular, arcuate, and dorsomedial hypothalamic nuclei; lateral hypothalamic area; central amygdalar nucleus; parasubthalamic nucleus; ventral posterior thalamic nucleus; area postrema; and nucleus of the solitary tract. These data indicate that the PB has bidirectional connections with most refeeding-activated neuronal groups, suggesting that short-loop feedback circuits exist in this satiety network. J. Comp. Neurol. 524:2803-2827, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Györgyi Zséli
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary, 1083.,Neuroendocrinology Program, Semmelweis University Neurosciences Doctoral School, Budapest, Hungary, 1085
| | - Barbara Vida
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary, 1083.,Department of Neuroscience, Faculty of Information Technology, Pázmány Péter Catholic University, Budapest, Hungary, 1088
| | - Anais Martinez
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, 79902.,Graduate Program in Pathobiology, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, 79902
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts, 02111.,Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts, 02111
| | - Arshad M Khan
- UTEP Systems Neuroscience Laboratory, Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, 79902
| | - Csaba Fekete
- Department of Endocrine Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary, 1083.,Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tupper Research Institute, Tufts Medical Center, Boston, Massachusetts, 02111
| |
Collapse
|
28
|
Excitatory and inhibitory enteric innervation of horse lower esophageal sphincter. Histochem Cell Biol 2015; 143:625-35. [PMID: 25578519 DOI: 10.1007/s00418-014-1306-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2014] [Indexed: 12/30/2022]
Abstract
The lower esophageal sphincter (LES) is a specialized, thickened muscle region with a high resting tone mediated by myogenic and neurogenic mechanisms. During swallowing or belching, the LES undergoes strong inhibitory innervation. In the horse, the LES seems to be organized as a "one-way" structure, enabling only the oral-anal progression of food. We characterized the esophageal and gastric pericardial inhibitory and excitatory intramural neurons immunoreactive (IR) for the enzymes neuronal nitric oxide synthase (nNOS) and choline acetyltransferase. Large percentages of myenteric plexus (MP) and submucosal (SMP) plexus nNOS-IR neurons were observed in the esophagus (72 ± 9 and 69 ± 8 %, respectively) and stomach (57 ± 17 and 45 ± 3 %, respectively). In the esophagus, cholinergic MP and SMP neurons were 29 ± 14 and 65 ± 24 vs. 36 ± 8 and 38 ± 20 % in the stomach, respectively. The high percentage of nitrergic inhibitory motor neurons observed in the caudal esophagus reinforces the role of the enteric nervous system in the horse LES relaxation. These findings might allow an evaluation of whether selective groups of enteric neurons are involved in horse neurological disorders such as megaesophagus, equine dysautonomia, and white lethal foal syndrome.
Collapse
|
29
|
Abstract
Megacolon, the irreversible dilation of a colonic segment, is a structural sign associated with various gastrointestinal disorders. In its hereditary, secondary form (e.g. in Hirschsprung's disease), dilation occurs in an originally healthy colonic segment due to an anally located, aganglionic zone. In contrast, in chronic Chagas' disease, the dilated segment itself displays pathohistological changes, and the earliest and most prominent being found was massive loss of myenteric neurons. This neuron loss was partial and selective, i.e. some neurons containing neuronal nitric oxide synthase and/or vasoactive intestinal peptide (VIP) were spared from neuron death. This disproportionate survival of inhibitory neurons, however, did not completely correlate with the calibre change along the surgically removed, megacolonic segments. A better correlation was observed as to potentially contractile muscle tissue elements and the interstitial cells of Cajal. Therefore, the decreased densities of α-smooth muscle actin- and c-kit-immunoreactive profiles were estimated along resected megacolonic segments. Their lowest values were observed in the megacolonic zones itself, whereas less pronounced decreases were found in the non-dilated, transitional zones (oral and anal to dilation). In contrast to the myenteric plexus, the submucosal plexus displayed only a moderate neuron loss. Neurons co-immunoreactive for VIP and calretinin survived disproportionately. As a consequence, these neurons may have contributed to maintain the epithelial barrier and allowed the chagasic patients to survive for decades, despite their severe disturbance of colonic motility. Due to its neuroprotective and neuroeffectory functions, VIP may play a key role in the development and duration of chagasic megacolon.
Collapse
|
30
|
Effects of inflammation and axotomy on expression of acetylcholine transferase and nitric oxide synthetase within the cocaine- and amphetamine-regulated transcript-immunoreactive neurons of the porcine descending colon. J Comp Pathol 2013; 150:287-96. [PMID: 24650891 DOI: 10.1016/j.jcpa.2013.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 08/05/2013] [Accepted: 08/30/2013] [Indexed: 12/14/2022]
Abstract
This study reports changes in expression of acetylcholine transferase (AChT) and nitric oxide synthetase (NOS) in neurons immunoreactive for cocaine- and amphetamine-regulated transcript (CART) peptides during chemically-driven inflammation and axotomy in the porcine descending colon. The co-localization of the neurotransmitters with CART was studied by double immunofluorescence in the myenteric plexus (MP) and outer submucosal plexus (OSP) of the porcine descending colon under physiological and selected pathological conditions. In control animals, neurons expressing CART also expressed AChT in 25.37 ± 0.98% and 26.73 ± 0.96% in the MP and OSP, respectively. Neuronal co-expression of CART with NOS occurred in 90.66 ± 2.13% and 88.09 ± 2.96% in the MP and OSP, respectively. Following axotomy the number of neurons co-expressing CART and AChT decreased to 16.50 ± 3.20% in the MP and increased to 35.49 ± 2.04% in the OSP, while the number of neurons co-expressing CART and NOS increased to 96.66 ± 2.38% in the MP and 97.46 ± 2.22% in the OSP. Experimentally-induced colitis resulted in an increase in the number of neurons co-expressing CART and AChT to 42.40 ± 2.28% in the MP and 63.62 ± 1.83% in the OSP. Similarly, in these animals the number of neurons co-expressing CART and NOS increased to 93.9 ± 2.58% in the MP and 90.43 ± 2.09% in the OSP. Sham-operated controls showed expression levels of 26.22 ± 0.66% (MP) and 27.02 ± 1.73% (OSP) for simultaneous CART and AChT expression and 94.18 ± 0.93% (MP) and 88.21 ± 0.81% (OSP) for CART and NOS co-localization. These data confirm that the examined neurotransmitters have a role in traumatic and inflammatory responses of enteric neurons.
Collapse
|
31
|
Histopathology in gastrointestinal neuromuscular diseases: methodological and ontological issues. Adv Anat Pathol 2013; 20:17-31. [PMID: 23232568 DOI: 10.1097/pap.0b013e31827b65c0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gastrointestinal neuromuscular diseases (GINMDs) comprise a heterogenous group of chronic conditions associated with impaired gut motility. These gastrointestinal (GI) disorders, differing for etiopathogenic mechanisms, pathologic lesions, and region of gut involvement, represent a relevant matter for public health, because they are very common, can be disabling, and determine major social and economic burdens. GINMDs are presumed or proven to arise as a result of a dysfunctioning GI neuromuscular apparatus, which includes myenteric ganglia (neurons and glial cells), interstitial cells of Cajal and smooth muscle cells. Despite the presence of symptoms related to gut dysmotility in the clinical phenotype of these patients, in the diagnostic setting scarce attention is usually paid to the morphologic pattern of the GI neuromuscular apparatus. It is also objectively difficult to collect full-thickness gut tissue samples from patients with GINMDs, because their disease, which can be only functional in nature, may not justify invasive diagnostic procedures as a first-line approach. As a consequence, whenever available, bioptic gut specimens, retrieved from these patients, must be regarded as a unique chance for obtaining relevant diagnostic information. On the basis of these arguments, there is an urgent need of standardized and validated histopathologic methods, aiming at overcoming the discrepancies affecting current approaches, which usually lead to conflicting definitions of normality and hamper the identification of disease-specific pathologic patterns. This review article intends to address current methodological and ontological issues in the histopathologic diagnosis of GINMDs, to foster the debate on how to discriminate normal morphology from abnormalities.
Collapse
|
32
|
Parkinson's disease is not associated with gastrointestinal myenteric ganglion neuron loss. Acta Neuropathol 2012; 124:665-80. [PMID: 22941241 DOI: 10.1007/s00401-012-1040-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 08/22/2012] [Accepted: 08/22/2012] [Indexed: 12/14/2022]
Abstract
Gastrointestinal dysfunction is a prominent non-motor feature of Parkinson's disease (PD) that contributes directly to the morbidity of patients, complicates management of motor symptoms, and may herald incipient PD in patients without motor disability. Although PD has traditionally been considered a disease of dopaminergic neurons in the substantia nigra, analyses of gastrointestinal samples from PD patients have consistently revealed pathology in the enteric nervous system. The relationship of PD pathology to GI dysmotility is poorly understood, and this lack of understanding has led to limited success in developing treatments for PD-related GI symptoms. We have quantitatively compared myenteric neuron density and relative abundance of NO, VIP, and catecholamine neurons between patients with PD and control individuals along the length of the GI tract. In addition, we have examined the frequency of GI α-synuclein neuritic pathology and its co-localization with the same neuronal markers. We have included a comparison with a small population of patients with incidental Lewy bodies found at autopsy. These data indicate that there is no neuronal loss in the myenteric plexus in PD. Lewy body pathology parallels parasympathetic autonomic input from the dorsal motor nucleus of the vagus, not the distribution of extrinsic sympathetic input or intrinsic enteric neurons, and is only rarely co-localized with tyrosine hydroxylase. These data provide a critical background to which further analyses of the effect of PD on the GI tract may be compared and suggest that neuropathology in myenteric neurons is unlikely to be a causative factor in PD-related GI dysmotility.
Collapse
|
33
|
Preponderance of inhibitory versus excitatory intramuscular nerve fibres in human chagasic megacolon. Int J Colorectal Dis 2012; 27:1181-9. [PMID: 22729712 DOI: 10.1007/s00384-012-1500-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2012] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Megacolon, chronic dilation of a colonic segment, is a frequent sign of Chagas disease. It is accompanied by an extensive neuron loss which, as shown recently, results in a partial, selective survival of nitrergic myenteric neurons. Here, we focused on the balance of intramuscular excitatory (choline acetyltransferase [ChAT]-immunoreactive) and inhibitory (neuronal nitric oxide synthase [NOS]- as well as vasoactive intestinal peptide [VIP]-immunoreactive) nerve fibres. MATERIALS AND METHODS From surgically removed megacolonic segments of seven patients, three sets of cryosections (from non-dilated oral, megacolonic and non-dilated anal parts) were immunhistochemically triple-stained for ChAT, NOS and VIP. Separate area measurements of nerve profiles within the circular and longitudinal muscle layers, respectively, were compared with those of seven non-chagasic control patients. Additionally, wholemounts from the same regions were stained for NOS, VIP and neurofilaments (NF). RESULTS The intramuscular nerve fibre density was significantly reduced in all three chagasic segments. The proportions of inhibitory (NOS only, VIP only, or NOS/VIP-coimmunoreactive) intramuscular nerves were 68 %/58 % (circular/longitudinal muscle, respectively) in the controls and increased to 75 %/69 % (oral parts), 84 %/76 % (megacolonic) and 87 %/94 % (anal) in chagasic specimens. In the myenteric plexus, NF-positive neurons co-staining for NOS and VIP also increased proportionally. The almost complete lack of dendritic structures in ganglia of chagasic specimens hampered morphological identification. DISCUSSION AND CONCLUSION We suggest that preponderance of inhibitory, intramuscular nerve fibres may be one factor explaining the chronic dilation. Since the nerve fibre imbalance is most pronounced in the anal, non-dilated segment, other components of the motor apparatus (musculature, interstitial cells, submucosal neurons) have to be considered.
Collapse
|
34
|
Noorian AR, Taylor GM, Annerino DM, Greene JG. Neurochemical phenotypes of myenteric neurons in the rhesus monkey. J Comp Neurol 2012; 519:3387-401. [PMID: 21618236 DOI: 10.1002/cne.22679] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Understanding the neurochemical composition of the enteric nervous system (ENS) is critical for elucidating neurological function in the gastrointestinal (GI) tract in health and disease. Despite their status as the closest models of human neurological systems, relatively little is known about enteric neurochemistry in nonhuman primates. We describe neurochemical coding of the enteric nervous system, specifically the myenteric plexus, of the rhesus monkey (Macaca mulatta) by immunohistochemistry and directly compare it to human tissues. There are considerable differences in the myenteric plexus along different segments of the monkey GI tract. While acetylcholine neurons make up the majority of myenteric neurons in the stomach (70%), they are a minority in the rectum (47%). Conversely, only 22% of gastric myenteric neurons express nitric oxide synthase (NOS) compared to 52% in the rectum. Vasoactive intestinal peptide (VIP) is more prominent in the stomach (37%) versus the rest of the GI tract (≈10%), and catecholamine neurons are rare (≈1%). There is significant coexpression of NOS and VIP in myenteric neurons that is more prominent in the proximal GI tract. Taken as a whole, these data provide insight into the neurochemical anatomy underlying GI motility. While overall similarity to other mammalian species is clear, there are some notable differences between the ENS of rhesus monkeys, humans, and other species that will be important to take into account when evaluating models of human diseases in animals.
Collapse
Affiliation(s)
- Ali Reza Noorian
- Department of Neurology and the Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | |
Collapse
|
35
|
Bernardini N, Segnani C, Ippolito C, De Giorgio R, Colucci R, Faussone-Pellegrini MS, Chiarugi M, Campani D, Castagna M, Mattii L, Blandizzi C, Dolfi A. Immunohistochemical analysis of myenteric ganglia and interstitial cells of Cajal in ulcerative colitis. J Cell Mol Med 2012; 16:318-27. [PMID: 21426484 PMCID: PMC3823295 DOI: 10.1111/j.1582-4934.2011.01298.x] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease with alterations of colonic motility, which influence clinical symptoms. Although morpho-functional abnormalities in the enteric nervous system have been suggested, in UC patients scarce attention has been paid to possible changes in the cells that control colonic motility, including myenteric neurons, glial cells and interstitial cells of Cajal (ICC). This study evaluated the neural-glial components of myenteric ganglia and ICC in the colonic neuromuscular compartment of UC patients by quantitative immunohistochemical analysis. Full-thickness archival samples of the left colon were collected from 10 patients with UC (5 males, 5 females; age range 45–62 years) who underwent elective bowel resection. The colonic neuromuscular compartment was evaluated immunohistochemically in paraffin cross-sections. The distribution and number of neurons, glial cells and ICC were assessed by anti-HuC/D, -S100β and -c-Kit antibodies, respectively. Data were compared with findings on archival samples of normal left colon from 10 sex- and age-matched control patients, who underwent surgery for uncomplicated colon cancer. Compared to controls, patients with UC showed: (i) reduced density of myenteric HuC/D+ neurons and S100β+ glial cells, with a loss over 61% and 38%, respectively, and increased glial cell/neuron ratio; (ii) ICC decrease in the whole neuromuscular compartment. The quantitative variations of myenteric neuro-glial cells and ICC indicate considerable alterations of the colonic neuromuscular compartment in the setting of mucosal inflammation associated with UC, and provide a morphological basis for better understanding the motor abnormalities often observed in UC patients.
Collapse
Affiliation(s)
- Nunzia Bernardini
- Department of Human Morphology and Applied Biology, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hübner S, Efthymiadis A. Recent progress in histochemistry and cell biology. Histochem Cell Biol 2012; 137:403-57. [PMID: 22366957 DOI: 10.1007/s00418-012-0933-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2012] [Indexed: 01/06/2023]
Abstract
Studies published in Histochemistry and Cell Biology in the year 2011 represent once more a manifest of established and newly sophisticated techniques being exploited to put tissue- and cell type-specific molecules into a functional context. The review is therefore the Histochemistry and Cell Biology's yearly intention to provide interested readers appropriate summaries of investigations touching the areas of tissue biology, developmental biology, the biology of the immune system, stem cell research, the biology of subcellular compartments, in order to put the message of such studies into natural scientific-/human- and also pathological-relevant correlations.
Collapse
Affiliation(s)
- Stefan Hübner
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany.
| | | |
Collapse
|
37
|
Beyond hematoxylin and eosin: the importance of immunohistochemical techniques for evaluating surgically resected constipated patients. Tech Coloproctol 2011; 15:371-5. [PMID: 21766200 DOI: 10.1007/s10151-011-0721-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Accepted: 06/30/2011] [Indexed: 12/18/2022]
Abstract
Chronic constipation requiring surgical ablation for intractability is often a frustrating condition from the pathologist's point of view. In fact, limiting the histological examination to only hematoxylin-eosin staining usually yields only the information that there are no abnormalities. By employing some simple and widely available immunohistochemical methods, discussed in this review, it is possible to gather data that may help in explaining the pathophysiological basis of constipation in these patients.
Collapse
|
38
|
Kapeller J, Möller D, Lasitschka F, Autschbach F, Hovius R, Rappold G, Brüss M, Gershon MD, Niesler B. Serotonin receptor diversity in the human colon: Expression of serotonin type 3 receptor subunits 5-HT3C, 5-HT3D, and 5-HT3E. J Comp Neurol 2011; 519:420-32. [PMID: 21192076 DOI: 10.1002/cne.22525] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since the first description of 5-HT₃ receptors more than 50 years ago, there has been speculation about the molecular basis of their receptor heterogeneity. We have cloned the genes encoding novel 5-HT3 subunits 5-HT3C, 5-HT3D, and 5-HT3E and have shown that these subunits are able to form functional heteromeric receptors when coexpressed with the 5-HT3A subunit. However, whether these subunits are actually expressed in human tissue remained to be confirmed. In the current study, we performed immunocytochemistry to locate the 5-HT3A as well as the 5-HT3C, 5-HT3D, and 5-HT3E subunits within the human colon. Western blot analysis was used to confirm subunit expression, and RT-PCR was employed to detect transcripts encoding 5-HT₃ receptor subunits in microdissected tissue samples. This investigation revealed, for the first time, that 5-HT3C, 5-HT3D, and 5-HT3E subunits are coexpressed with 5-HT3A in cell bodies of myenteric neurons. Furthermore, 5-HT3A and 5-HT3D were found to be expressed in submucosal plexus of the human large intestine. These data provide a strong basis for future studies of the roles that specific 5-HT₃ receptor subtypes play in the function of the enteric and central nervous systems and the contribution that specific 5-HT₃ receptors make to the pathophysiology of gastrointestinal disorders such as irritable bowel syndrome and dyspepsia.
Collapse
Affiliation(s)
- Johannes Kapeller
- Department of Human Molecular Genetics, University of Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kustermann A, Neuhuber W, Brehmer A. Calretinin and somatostatin immunoreactivities label different human submucosal neuron populations. Anat Rec (Hoboken) 2011; 294:858-69. [PMID: 21416629 DOI: 10.1002/ar.21365] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2010] [Accepted: 01/24/2011] [Indexed: 01/13/2023]
Abstract
In human myenteric plexus, calretinin (CALR) and somatostatin (SOM) coexist in Dogiel Type II neurons, which were considered as intrinsic primary afferent neurons in the guinea pig. The aims of this study were to test if also human submucosal neurons costain immunohistochemically for CALR and SOM and whether these or other neurons display Type II morphology. Two sets of submucosal wholemounts of small and large intestine from 29 patients (median age 65 years) were triple stained for CALR, SOM, and human neuronal protein Hu C/D (HU, a pan-neuronal marker) as well as for CALR, SOM, and peripherin (PER), respectively. Only exceptionally, neurons coreactive for both CALR and SOM were found. The three major groups of neurons were CALR-/HU-coreactive (CALR-neurons), SOM-/HU-coreactive (SOM-neurons), and HU-alone-positive neurons. We observed significantly more CALR-neurons in the external submucosal plexus (ESP) of all regions and more SOM-neurons in the internal submucosal plexus (ISP), although with substantial interindividual variations. Comparisons of small vs. large intestine revealed more SOM-neurons (ESP: 29% vs. 4%, ISP: 40% vs. 13%) but fewer CALR-neurons (ESP: 37% vs. 77%, ISP: 21% vs. 67%) in small intestine. Morphologically, CALR-neurons had multiple processes; in some cases, we identified multidendritic/uniaxonal neurons. In contrast, SOM-neurons had mostly only one process. The functions of both populations as possible primary afferent neurons, interneurons, secretomotor neurons, or vasomotor neurons are discussed. Future morphochemical distinction of these groups may reveal different subgroups.
Collapse
|
40
|
Knowles CH, Farrugia G. Gastrointestinal neuromuscular pathology in chronic constipation. Best Pract Res Clin Gastroenterol 2011; 25:43-57. [PMID: 21382578 PMCID: PMC4175481 DOI: 10.1016/j.bpg.2010.12.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 12/15/2010] [Indexed: 01/31/2023]
Abstract
Some patients with chronic constipation may undergo colectomy yielding tissue appropriate to diagnosis of underlying neuromuscular pathology. The analysis of such tissue has, over the past 40 years, fueled research that has explored the presence of neuropathy, myopathy and more recently changes in interstitial cells of Cajal (ICC). In this chapter, the data from these studies have been critically reviewed in the context of the significant methodological and interpretative issues that beset the field of gastrointestinal neuromuscular pathology. On this basis, reductions in ICC appear to a consistent finding but one whose role as a primary cause of slow-transit constipation requires further evaluation. Findings indicative of significant neuropathy or myopathy are variable and in many studies subject to considerable methodological bias. Methods with practical diagnostic utility in the individual patient have rarely been employed and require further validation in respect of normative data.
Collapse
Affiliation(s)
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
41
|
Jabari S, da Silveira ABM, de Oliveira EC, Neto SG, Quint K, Neuhuber W, Brehmer A. Partial, selective survival of nitrergic neurons in chagasic megacolon. Histochem Cell Biol 2010; 135:47-57. [PMID: 21184236 PMCID: PMC3019355 DOI: 10.1007/s00418-010-0774-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2010] [Indexed: 02/06/2023]
Abstract
One frequent chronic syndrome of Chagas’ disease is megacolon, an irreversible dilation of a colonic segment. Extensive enteric neuron loss in the affected segment is regarded as key factor for deficient motility. Here, we assessed the quantitative balance between cholinergic and nitrergic neurons representing the main limbs of excitatory and inhibitory colonic motor innervation, respectively. From surgically removed megacolonic segments of four patients, each three myenteric wholemounts (from non-dilated oral, megacolonic and non-dilated anal parts) was immunohistochemically triple-stained for choline acetyltransferase, neuronal nitric oxide synthase (NOS) and the panneuronal human neuronal protein Hu C/D. Degenerative changes were most pronounced in the megacolonic and anal regions, e.g. bulked, honeycomb-like ganglia with few neurons which were partly enlarged or atrophic or vacuolated. Neuron counts from each 15 ganglia of 12 megacolonic wholemounts were compared with those of 12 age- and region-matched controls. Extensive neuron loss, mainly in megacolonic and anal wholemounts, was obvious. In all three regions derived from megacolonic samples, the proportion of NOS-positive neurons (control: 55%) was significantly increased: in non-dilated oral parts to 61% (p = 0.003), in megacolonic regions to 72% (p < 0.001) and in non-dilated anal regions to 78% (p < 0.001). We suggest the chronic dilation of megacolonic specimens to be due to the preponderance of the nitrergic, inhibitory input to the intestinal muscle. However, the observed neuronal imbalance was not restricted to the dilated regions: the non-dilated anal parts may be innervated by ascending, cholinergic axons emerging from less affected, more anally located regions.
Collapse
Affiliation(s)
- Samir Jabari
- Institute of Anatomy I, University of Erlangen-Nuremberg, Krankenhausstr. 9, 91054, Erlangen, Germany
| | | | | | | | | | | | | |
Collapse
|
42
|
King SK, Sutcliffe JR, Ong SY, Lee M, Koh TL, Wong SQ, Farmer PJ, Peck CJ, Stanton MP, Keck J, Cook DJ, Chow CW, Hutson JM, Southwell BR. Substance P and vasoactive intestinal peptide are reduced in right transverse colon in pediatric slow-transit constipation. Neurogastroenterol Motil 2010; 22:883-92, e234. [PMID: 20529207 DOI: 10.1111/j.1365-2982.2010.01524.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Slow-transit constipation (STC) is recognized in children but the etiology is unknown. Abnormalities in substance P (SP), vasoactive intestinal peptide (VIP) and nitric oxide (NO) have been implicated. The density of nerve fibers in circular muscle containing these transmitters was examined in colon from children with STC and compared to other pediatric and adult samples. METHODS Fluorescence immunohistochemistry using antibodies to NO synthase (NOS), VIP and SP was performed on colonic biopsies (transverse and sigmoid colon) from 33 adults with colorectal cancer, 11 children with normal colonic transit and anorectal retention (NAR) and 51 with chronic constipation and slow motility in the proximal colon (STC). The percentage area of nerve fibers in circular muscle containing each transmitter was quantified in confocal images. KEY RESULTS In colon circular muscle, the percentage area of nerve fibers containing NOS > VIP > SP (6 : 2 : 1). Pediatric groups had a higher density of nerve fibers than adults. In pediatric samples, there were no regional differences in NOS and VIP, while SP nerve fiber density was higher in sigmoid than proximal colon. STC children had lower SP and VIP nerve fiber density in the proximal colon than NAR children. Twenty-three percent of STC children had low SP nerve fiber density. CONCLUSIONS & INFERENCES There are age-related reductions in nerve fiber density in human colon circular muscle. NOS and VIP do not show regional variations, while SP nerve fiber density is higher in distal colon. 1/3 of pediatric STC patients have low SP or VIP nerve fiber density in proximal colon.
Collapse
Affiliation(s)
- S K King
- Department of General Surgery, Royal Children's Hospital, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Harrington AM, Lee M, Ong SY, Yong E, Farmer P, Peck CJ, Chow CW, Hutson JM, Southwell BR. Immunoreactivity for high-affinity choline transporter colocalises with VAChT in human enteric nervous system. Cell Tissue Res 2010; 341:33-48. [PMID: 20490865 DOI: 10.1007/s00441-010-0981-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 04/08/2010] [Indexed: 01/08/2023]
Abstract
Cholinergic nerves are identified by labelling molecules in the ACh synthesis, release and destruction pathway. Recently, antibodies against another molecule in this pathway have been developed. Choline reuptake at the synapse occurs via the high-affinity choline transporter (CHT1). CHT1 immunoreactivity is present in cholinergic nerve fibres containing vesicular acetylcholine transporter (VAChT) in the human and rat central nervous system and rat enteric nervous system. We have examined whether CHT1 immunoreactivity is present in nerve fibres in human intestine and whether it is colocalised with markers of cholinergic, tachykinergic or nitrergic circuitry. Human ileum and colon were fixed, sectioned and processed for fluorescence immunohistochemistry with antibodies against CHT1, class III beta-tubulin (TUJ1), synaptophysin, common choline acetyl-transferase (cChAT), VAChT, nitric oxide synthase (NOS), substance P (SP) and vasoactive intestinal peptide (VIP). CHT1 immunoreactivity was present in many nerve fibres in the circular and longitudinal muscle, myenteric and submucosal ganglia, submucosa and mucosa in human colon and ileum and colocalised with immunoreactivity for TUJ1 and synaptophysin confirming its presence in nerve fibres. In nerve fibres in myenteric ganglia and muscle, CHT1 immunoreactivity colocalised with immunoreactivity for VAChT and cChAT. Some colocalisation occurred with SP immunoreactivity, but little with immunoreactivity for VIP or NOS. In the mucosa, CHT1 immunoreactivity colocalised with that for VIP and SP in nerve fibres and was also present in vascular nerve fibres in the submucosa and on epithelial cells on the luminal border of crypts. The colocalisation of CHT1 immunoreactivity with VAChT immunoreactivity in cholinergic enteric nerves in the human bowel thus suggests that CHT1 represents another marker of cholinergic nerves.
Collapse
Affiliation(s)
- Andrea M Harrington
- F Douglas Stephens Surgical Research Laboratory, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Smooth-muscle-specific expression of neurotrophin-3 in mouse embryonic and neonatal gastrointestinal tract. Cell Tissue Res 2010; 340:267-86. [PMID: 20387078 DOI: 10.1007/s00441-010-0959-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2009] [Accepted: 02/26/2010] [Indexed: 12/20/2022]
Abstract
Vagal gastrointestinal (GI) afferents are essential for the regulation of eating, body weight, and digestion. However, their functional organization and the way that this develops are poorly understood. Neurotrophin-3 (NT-3) is crucial for the survival of vagal sensory neurons and is expressed in the developing GI tract, possibly contributing to their survival and to other aspects of vagal afferent development. The identification of the functions of this peripheral NT-3 thus requires a detailed understanding of the localization and timing of its expression in the developing GI tract. We have studied embryos and neonates expressing the lacZ reporter gene from the NT-3 locus and found that NT-3 is expressed predominantly in the smooth muscle of the outer GI wall of the stomach, intestines, and associated blood vessels and in the stomach lamina propria and esophageal epithelium. NT-3 expression has been detected in the mesenchyme of the GI wall by embryonic day 12.5 (E12.5) and becomes restricted to smooth muscle and lamina propria by E15.5, whereas its expression in blood vessels and esophageal epithelium is first observed at E15.5. Expression in most tissues is maintained at least until postnatal day 4. The lack of colocalization of beta-galactosidase and markers for myenteric ganglion cell types suggests that NT-3 is not expressed in these ganglia. Therefore, NT-3 expression in the GI tract is largely restricted to smooth muscle at ages when vagal axons grow into the GI tract, and when vagal mechanoreceptors form in smooth muscle, consistent with its role in these processes and in vagal sensory neuron survival.
Collapse
|
45
|
Wedel T, Büsing V, Heinrichs G, Nohroudi K, Bruch HP, Roblick UJ, Böttner M. Diverticular disease is associated with an enteric neuropathy as revealed by morphometric analysis. Neurogastroenterol Motil 2010; 22:407-14, e93-4. [PMID: 20040058 DOI: 10.1111/j.1365-2982.2009.01445.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The pathogenesis of diverticular disease (DD) is attributed to several aetiological factors (e.g. age, diet, connective tissue disorders) but also includes distinct intestinal motor abnormalities. Although the enteric nervous system (ENS) is the key-regulator of intestinal motility, data on neuropathological alterations are limited. The study aimed to investigate the ENS by a systematic morphometric analysis. METHODS Full-thickness sigmoid specimens obtained from patients with symptomatic DD (n = 27) and controls (n = 27) were processed for conventional histology and immunohistochemistry using anti-HuC/D as pan-neuronal marker. Enteric ganglia, nerve and glial cells were quantified separately in the myenteric, external and internal submucosal plexus compartments. KEY RESULTS Compared to controls, patients with DD showed significantly (P < 0.05) (i) reduced neuronal density in all enteric nerve plexus, (ii) decrease of ganglionic nerve cell content in the myenteric plexus, (iii) decreased ganglionic density in the internal submucosal plexus, (iv) reduced glial cell density in the myenteric plexus, (v) decrease of ganglionic glial cell content in the myenteric plexus and increase in submucosal plexus compartments, (vi) increased glia index in all enteric nerve plexus. About 44.4% of patients with DD exhibited myenteric ganglia displaying enteric gliosis. CONCLUSIONS & INFERENCES Patients with DD show substantial structural alterations of the ENS mainly characterized by myenteric and submucosal oligo-neuronal hypoganglionosis which may account for intestinal motor abnormalities reported in DD. The morphometric data give evidence that DD is associated with structural alterations of the ENS which may complement established pathogenetic concepts.
Collapse
Affiliation(s)
- T Wedel
- Department of Anatomy, Christian-Albrechts University of Kiel, Kiel, Germany.
| | | | | | | | | | | | | |
Collapse
|
46
|
Beck M, Schlabrakowski A, Schrödl F, Neuhuber W, Brehmer A. ChAT and NOS in human myenteric neurons: co-existence and co-absence. Cell Tissue Res 2009; 338:37-51. [DOI: 10.1007/s00441-009-0852-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Accepted: 07/27/2009] [Indexed: 12/31/2022]
|
47
|
Drolet RE, Cannon JR, Montero L, Greenamyre JT. Chronic rotenone exposure reproduces Parkinson's disease gastrointestinal neuropathology. Neurobiol Dis 2009; 36:96-102. [PMID: 19595768 DOI: 10.1016/j.nbd.2009.06.017] [Citation(s) in RCA: 155] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 06/26/2009] [Accepted: 06/30/2009] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal disorders, particularly severe constipation and delayed gastric emptying, are core symptoms of Parkinson's disease that affect most patients. However, the neuropathological substrate and physiological basis for this dysfunction are poorly defined. To begin to explore these phenomena in laboratory models of PD, rats were treated with either vehicle or rotenone (2.0 mg/kg, i.p.; 5 days/week) for 6-weeks. Myenteric plexus alpha-synuclein aggregate pathology and neuron loss were assessed 3-days and 6-months after the last rotenone injection. Gastrointestinal motility was assessed at 3-days, 1-month and 6-months after the last rotenone injection. Rotenone treatment caused an acute reduction in alpha-synuclein-immunoreactivity, but this was followed 6 months later by a robust increase in aggregate pathology and cytoplasmic inclusions that were similar in appearance to enteric Lewy-bodies in idiopathic PD. Rotenone-treated rats also had a moderate but permanent loss of small intestine myenteric neurons and an associated modest slowing of gastrointestinal motility 6-months after treatment. Our results suggest that a circumscribed exposure to an environmental toxicant can cause the delayed appearance of parkinsonian alpha-synuclein pathology in the enteric nervous system and an associated functional deficit in gastrointestinal motility. The rotenone model may therefore, provide a means to investigate pathogenic mechanisms and to test new therapeutic interventions into gastrointestinal dysfunction in PD.
Collapse
Affiliation(s)
- Robert E Drolet
- Pittsburgh Institute for Neurodegenerative Disease, Department of Neurology, University of Pittsburgh, Pittsburgh, PA 1526, USA.
| | | | | | | |
Collapse
|
48
|
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
|
49
|
Bernard CE, Gibbons SJ, Gomez-Pinilla PJ, Lurken MS, Schmalz PF, Roeder JL, Linden D, Cima RR, Dozois EJ, Larson DW, Camilleri M, Zinsmeister AR, Pozo MJ, Hicks GA, Farrugia G. Effect of age on the enteric nervous system of the human colon. Neurogastroenterol Motil 2009; 21:746-e46. [PMID: 19220755 PMCID: PMC2776702 DOI: 10.1111/j.1365-2982.2008.01245.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The effect of age on the anatomy and function of the human colon is incompletely understood. The prevalence of disorders in adults such as constipation increase with age but it is unclear if this is due to confounding factors or age-related structural defects. The aim of this study was to determine number and subtypes of enteric neurons and neuronal volumes in the human colon of different ages. Normal colon (descending and sigmoid) from 16 patients (nine male) was studied; ages 33-99. Antibodies to HuC/D, choline acetyltransferase (ChAT), neuronal nitric oxide synthase (nNOS), and protein gene product 9.5 were used. Effect of age was determined by testing for linear trends using regression analysis. In the myenteric plexus, number of Hu-positive neurons declined with age (slope = -1.3 neurons/mm/10 years, P = 0.03). The number of ChAT-positive neurons also declined with age (slope = -1.1 neurons/mm/10 years of age, P = 0.02). The number of nNOS-positive neurons did not decline with age. As a result, the ratio of nNOS to Hu increased (slope = 0.03 per 10 years of age, P = 0.01). In the submucosal plexus, the number of neurons did not decline with age (slope = -0.3 neurons/mm/10 years, P = 0.09). Volume of nerve fibres in the circular muscle and volume of neuronal structures in the myenteric plexus did not change with age. In conclusion, the number of neurons in the human colon declines with age with sparing of nNOS-positive neurons. This change was not accompanied by changes in total volume of neuronal structures suggesting compensatory changes in the remaining neurons.
Collapse
Affiliation(s)
- Cheryl E. Bernard
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| | - Simon J. Gibbons
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| | - Pedro J. Gomez-Pinilla
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain,Department of Physiology, Nursing School, University of Extremadura, Caceres, Spain
| | - Matthew S. Lurken
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| | - Philip F. Schmalz
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| | - Jaime L. Roeder
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| | - David Linden
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| | - Robert R. Cima
- Department of Surgery, University of Extremadura, Caceres, Spain
| | - Eric J. Dozois
- Department of Surgery, University of Extremadura, Caceres, Spain
| | - David W. Larson
- Department of Surgery, University of Extremadura, Caceres, Spain
| | - Michael Camilleri
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| | - Alan R Zinsmeister
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain,Division of Biostatistics, Mayo Clinic College of Medicine, Rochester, MN, University of Extremadura, Caceres, Spain
| | - Maria J Pozo
- Department of Physiology, Nursing School, University of Extremadura, Caceres, Spain,Red Tematica de Investigacion Cooperative en Envejecimiento y, Fragilidad, East Hanover,, NJ
| | | | - Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, University of Extremadura, Caceres, Spain
| |
Collapse
|
50
|
The Role of Immunohistochemistry in Idiopathic Chronic Intestinal Pseudoobstruction (CIPO). Am J Surg Pathol 2009; 33:749-58. [DOI: 10.1097/pas.0b013e31819b381a] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|