1
|
Ersson B, Gustafson E, Danielson J, Alafuzoff I. Pathology Seen in Myenteric Plexus in Two Subjects With Waardenburg Syndrome. Neurogastroenterol Motil 2025:e70073. [PMID: 40364458 DOI: 10.1111/nmo.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025]
Abstract
OBJECTIVES The aim was to assess the neuroglial compartment in the myenteric plexus of two subjects with genetically verified Waardenburg syndrome (WS) type 4 (WS4) and to compare the outcome with four "age-matched" controls. DESIGN Gut samples from four control cases and from two newborn subjects with WS4, one with peripheral demyelinating neuropathy, dysmyelinating leukodystrophy, WS and Hirschprung disease (PCWH) (SOX10, c.769A>T, p.Lys257*) and one with Waardenburg-Shah syndrome (WSS) (EDN3, c.472C>T,p.Arg158Cys)-were assessed histologically and immunohistochemically. Antibodies directed to glial cells (SOX10), ganglion cells (HuC/D), and interstitial cells of Cajal (CD117) were applied. RESULTS For the child with PCWH syndrome, both the small and large intestine showed a reduction in the number of glial cells (SOX10), in parallel with hypoganglionosis (HuC/D), when compared with "age-matched" controls. In the child with WSS, a severe reduction in the number of glial cells (SOX10) was observed in both the small and large intestine accompanied by aganglionosis (HuC/D) with a skipped segment. The number of interstitial cells of Cajal (CD117) appeared unaffected in both PCWH and WSS cases. CONCLUSION A severe reduction of glial cells and a severe reduction or loss of ganglion cells (the number of cells assessed per unit length), were seen in our study subjects when compared with "age-matched" controls. Contrary to the above the presence of Cajal cells was unaffected.
Collapse
Affiliation(s)
- Björn Ersson
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Elisabet Gustafson
- Department of Pediatric Surgery, Uppsala University Hospital, Uppsala, Sweden
| | - Johan Danielson
- Department of Pediatric Surgery, Uppsala University Hospital, Uppsala, Sweden
| | - Irina Alafuzoff
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
- Genetics and Pathology, Institution of Immunology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Grant MT, Nelvagal HR, Tecos M, Hamed A, Swanson K, Cooper JD, Vrecenak JD. Cellular trafficking and fate mapping of cells within the nervous system after in utero hematopoietic cell transplantation. Commun Biol 2024; 7:1624. [PMID: 39638879 PMCID: PMC11621337 DOI: 10.1038/s42003-024-06847-6] [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: 12/09/2022] [Accepted: 09/05/2024] [Indexed: 12/07/2024] Open
Abstract
In utero hematopoietic cell transplantation (IUHCT) utilizes fetal immune tolerance to achieve durable chimerism without conditioning or immunosuppression during a unique window in fetal development. Though donor cells have been observed within the nervous system following in utero injection, the timeline and distribution of cellular trafficking across the blood-brain barrier following IUHCT is not well understood. We injected 20 × 106 adult bone marrow mononuclear cells intravenously at gestational age (GA) 12-17 days and found that donor cells were maximally concentrated in the brain with treatment between GA 13-14. Donor cell engraftment persisted within the brain at every timepoint analyzed and concentrated within the hindbrain with significantly more grafted cells than in the forebrain. Additionally, transplanted cells terminally differentiated into various nervous system cellular morphologies and also populated the enteric nervous system. This study is the first to document the timeline and distribution of donor cell trafficking into the immune-protected nervous system and serves as a foundation for the application of IUHCT to treat neurogenetic diseases.
Collapse
Affiliation(s)
- Matthew T Grant
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Hemanth Ramesh Nelvagal
- Washington University in St. Louis School of Medicine, Department of Pediatrics, Division of Genetics and Genomics, St. Louis, MO, USA
- University College London, School of Pharmacy, Department of Pharmacology, London, UK
| | - Maria Tecos
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Amal Hamed
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Kerry Swanson
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA
| | - Jonathan D Cooper
- Washington University in St. Louis School of Medicine, Department of Pediatrics, Division of Genetics and Genomics, St. Louis, MO, USA
| | - Jesse D Vrecenak
- Washington University in St. Louis School of Medicine, Department of Surgery, Division of Pediatric Surgery, St. Louis, MO, USA.
| |
Collapse
|
3
|
Schonkeren SL, Thijssen MS, Idris M, Wouters K, de Vaan J, Teubner A, Gijbels MJ, Boesmans W, Melotte V. Differences in enteric neuronal density in the NSE-Noggin mouse model across institutes. Sci Rep 2024; 14:3686. [PMID: 38355947 PMCID: PMC10866904 DOI: 10.1038/s41598-024-54337-w] [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: 12/20/2023] [Accepted: 02/12/2024] [Indexed: 02/16/2024] Open
Abstract
The enteric nervous system (ENS) is a large and complex part of the peripheral nervous system, and it is vital for gut homeostasis. To study the ENS, different hyper- and hypo-innervated model systems have been developed. The NSE-Noggin mouse model was described as one of the few models with a higher enteric neuronal density in the colon. However, in our hands NSE-Noggin mice did not present with a hyperganglionic phenotype. NSE-Noggin mice were phenotyped based on fur appearance, genotyped and DNA sequenced to demonstrate transgene and intact NSE-Noggin-IRES-EGFP construct presence, and RNA expression of Noggin was shown to be upregulated. Positive EGFP staining in the plexus of NSE-Noggin mice also confirmed Noggin protein expression. Myenteric plexus preparations of the colon were examined to quantify both the overall density of enteric neurons and the proportions of enteric neurons expressing specific subtype markers. The total number of enteric neurons in the colonic myenteric plexus of transgenic mice did not differ significantly from wild types, nor did the proportion of calbindin, calretinin, or serotonin immunoreactive myenteric neurons. Possible reasons as to why the hyperinnervated phenotype could not be observed in contrast with original studies using this mouse model are discussed, including study design, influence of microbiota, and other environmental variables.
Collapse
Affiliation(s)
- Simone L Schonkeren
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Meike S Thijssen
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Musa Idris
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kim Wouters
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joëlle de Vaan
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Andreas Teubner
- Central Animal Facility, Faculty of Health, Medicine & Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Marion J Gijbels
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences: Atherosclerosis & Ischemic Syndrome, Amsterdam Infection and Immunity: Inflammatory Diseases, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Werend Boesmans
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands
- Biomedical Research Institute (BIOMED), Hasselt University, Hasselt, Belgium
| | - Veerle Melotte
- Department of Pathology, GROW-Research Institute for Oncology and Reproduction, Maastricht University Medical Center, Maastricht, The Netherlands.
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
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
|
5
|
Ma J, Nguyen D, Madas J, Kwiat AM, Toledo Z, Bizanti A, Kogut N, Mistareehi A, Bendowski K, Zhang Y, Chen J, Li DP, Powley TL, Furness JB, Cheng Z. Spinal afferent innervation in flat-mounts of the rat stomach: anterograde tracing. Sci Rep 2023; 13:17675. [PMID: 37853008 PMCID: PMC10584867 DOI: 10.1038/s41598-023-43120-y] [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: 04/12/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
The dorsal root ganglia (DRG) project spinal afferent axons to the stomach. However, the distribution and morphology of spinal afferent axons in the stomach have not been well characterized. In this study, we used a combination of state-of-the-art techniques, including anterograde tracer injection into the left DRG T7-T11, avidin-biotin and Cuprolinic Blue labeling, Zeiss M2 Imager, and Neurolucida to characterize spinal afferent axons in flat-mounts of the whole rat stomach muscular wall. We found that spinal afferent axons innervated all regions with a variety of distinct terminal structures innervating different gastric targets: (1) The ganglionic type: some axons formed varicose contacts with individual neurons within myenteric ganglia. (2) The muscle type: most axons ran in parallel with the longitudinal and circular muscles and expressed spherical varicosities. Complex terminal structures were observed within the circular muscle layer. (3) The ganglia-muscle mixed type: some individual varicose axons innervated both myenteric neurons and the circular muscle, exhibiting polymorphic terminal structures. (4) The vascular type: individual varicose axons ran along the blood vessels and occasionally traversed the vessel wall. This work provides a foundation for future topographical anatomical and functional mapping of spinal afferent axon innervation of the stomach under normal and pathophysiological conditions.
Collapse
Affiliation(s)
- Jichao Ma
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Duyen Nguyen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Jazune Madas
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Andrew M Kwiat
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Zulema Toledo
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Ariege Bizanti
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Nicole Kogut
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Anas Mistareehi
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Kohlton Bendowski
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Yuanyuan Zhang
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - Jin Chen
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA
| | - De-Pei Li
- Department of Medicine, Center for Precision Medicine, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Terry L Powley
- Department of Psychological Sciences, Purdue University, West Lafayette, IN, 479062, USA
| | - John B Furness
- Department of Anatomy and Physiology, University of Melbourne, and Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Zixi Cheng
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, 32816, USA.
| |
Collapse
|
6
|
Yamada NO, Wenduerma, Senda T. Altered microbiota caused by disordered gut motility leads to an overactivation of intestinal immune system in APC1638T mice. Med Mol Morphol 2023; 56:177-186. [PMID: 36995439 DOI: 10.1007/s00795-023-00352-1] [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: 01/12/2023] [Accepted: 03/19/2023] [Indexed: 03/31/2023]
Abstract
Adenomatous polyposis coli (APC) is recognized as an antioncogene related to familial adenomatous polyposis and colorectal cancers. However, APC is a large protein with multiple binding partners, indicating APC has diverse roles besides as a tumor suppressor. We have ever studied the roles of APC by using APC1638T/1638T (APC1638T) mice. Through those studies, we have noticed stools of APC1638T mice were smaller than those of APC+/+ mice and hypothesized there be a disturbance in fecal formation processes in APC1638T mice. The gut motility was morphologically analyzed by immunohistochemical staining of the Auerbach's plexus. Gut microbiota was analyzed by terminal restriction fragment length polymorphism (T-RFLP). IgA concentration in stools was determined by enzyme-linked immunosorbent assay (ELISA). As results, macroscopic findings suggestive of large intestinal dysmotility and microscopic findings of disorganization and inflammation of the plexus were obtained in APC1638T mice. An alteration of microbiota composition, especially increased Bacteroidetes population was observed. Increases in IgA positive cells and dendritic cells in the ileum with high fecal IgA concentration were also confirmed, suggesting over-activation of gut immunity. Our findings will contribute to our understanding of APC's functions in the gastrointestinal motility, and lead to a development of novel therapies for gut dysmotility-related diseases.
Collapse
Affiliation(s)
- Nami O Yamada
- Department of Anatomy, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan.
| | - Wenduerma
- Department of Anatomy, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| | - Takao Senda
- Department of Anatomy, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1194, Japan
| |
Collapse
|
7
|
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: 8] [Impact Index Per Article: 2.7] [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
|
8
|
Chen Y, Zhang S, Li Y, Yan H, Ba Y, Wang X, Shi N, Liu C. Gastric Electrical Stimulation Increases the Proliferation of Interstitial Cells of Cajal and Alters the Enteric Nervous System in Diabetic Rats. Neuromodulation 2022; 25:1106-1114. [DOI: 10.1016/j.neurom.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 09/14/2021] [Accepted: 09/20/2021] [Indexed: 11/26/2022]
|
9
|
Tominaga T, Nagayama S, Takamatsu M, Miyanari S, Nagasaki T, Yamaguchi T, Akiyoshi T, Konishi T, Fujimoto Y, Fukunaga Y, Ueno M. A case of severe megacolon due to acquired isolated hypoganglionosis after low anterior resection for lower rectal cancer. Clin J Gastroenterol 2019; 13:328-333. [PMID: 31828729 PMCID: PMC7239813 DOI: 10.1007/s12328-019-01079-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 11/29/2019] [Indexed: 12/17/2022]
Abstract
Acquired isolated hypoganglionosis is a rare intestinal neurological disease, which presents in adulthood with the clinical symptoms of chronic constipation. A 39-year-old man underwent laparoscopic low anterior resection and covering ileostomy for locally advanced-rectal cancer. A 6-month course of postoperative adjuvant chemotherapy was completed, followed by closure of the ileostoma. After the closure, he developed severe colitis which required 1-month of hospitalization. Mucosal erosions and pseudo-membrane formation were evident on colonoscopy and severe mucosal damage characterized by infiltration of inflammatory cells and crypt degeneration were pathologically confirmed. Even after the remission of the colitis, he suffered from severe constipation and distention. At 4 years after the stoma closure, he decided to undergo laparoscopic total colectomy. Histopathologically, the nerve fibers and ganglion cells became gradually scarcer from the non-dilated to dilated regions. Immunohistochemical staining examination confirmed that the ganglion cells gradually decreased and became degenerated from the normal to dilated region, thereby arriving at the final diagnosis of isolated hypoganglionosis. The patient recovered without any complications and there has been no evidence of any relapse of the symptoms. We present a case of acquired isolated hypoganglionosis-related megacolon, which required laparoscopic total colectomy, due to severe enterocolitis following stoma closure.
Collapse
Affiliation(s)
- Tetsuro Tominaga
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Satoshi Nagayama
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Manabu Takamatsu
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shun Miyanari
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Toshiya Nagasaki
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Tomohiro Yamaguchi
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Takashi Akiyoshi
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Tsuyoshi Konishi
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yoshiya Fujimoto
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Yosuke Fukunaga
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Masashi Ueno
- Department of Gastroenterological Surgery, Gastroenterological Cancer, Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 135-8550, Japan
| |
Collapse
|
10
|
Ohlemacher SK, Langer KB, Fligor CM, Feder EM, Edler MC, Meyer JS. Advances in the Differentiation of Retinal Ganglion Cells from Human Pluripotent Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:121-140. [PMID: 31654388 DOI: 10.1007/978-3-030-28471-8_5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human pluripotent stem cell (hPSC) technology has revolutionized the field of biology through the unprecedented ability to study the differentiation of human cells in vitro. In the past decade, hPSCs have been applied to study development, model disease, develop drugs, and devise cell replacement therapies for numerous biological systems. Of particular interest is the application of this technology to study and treat optic neuropathies such as glaucoma. Retinal ganglion cells (RGCs) are the primary cell type affected in these diseases, and once lost, they are unable to regenerate in adulthood. This necessitates the development of strategies to study the mechanisms of degeneration as well as develop translational therapeutic approaches to treat early- and late-stage disease progression. Numerous protocols have been established to derive RGCs from hPSCs, with the ability to generate large populations of human RGCs for translational applications. In this review, the key applications of hPSCs within the retinal field are described, including the use of these cells as developmental models, disease models, drug development, and finally, cell replacement therapies. In greater detail, the current report focuses on the differentiation of hPSC-derived RGCs and the many unique characteristics associated with these cells in vitro including their genetic identifiers, their electrophysiological activity, and their morphological maturation. Also described is the current progress in the use of patient-specific hPSCs to study optic neuropathies affecting RGCs, with emphasis on the use of these RGCs for studying disease mechanisms and pathogenesis, drug screening, and cell replacement therapies in future studies.
Collapse
Affiliation(s)
- Sarah K Ohlemacher
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Kirstin B Langer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Clarisse M Fligor
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Elyse M Feder
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Michael C Edler
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA.,Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA
| | - Jason S Meyer
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA. .,Department of Medical and Molecular Genetics, Indiana University, Indianapolis, IN, USA. .,Stark Neurosciences Research Institute, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
11
|
HuC/D expression in small round cell tumors and neuroendocrine tumors: a useful tool for distinguishing neuroblastoma from childhood small round cell tumors. Hum Pathol 2019; 85:162-167. [DOI: 10.1016/j.humpath.2018.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/30/2018] [Accepted: 11/02/2018] [Indexed: 12/13/2022]
|
12
|
Ceccotti C, Giaroni C, Bistoletti M, Viola M, Crema F, Terova G. Neurochemical characterization of myenteric neurons in the juvenile gilthead sea bream (Sparus aurata) intestine. PLoS One 2018; 13:e0201760. [PMID: 30075006 PMCID: PMC6075763 DOI: 10.1371/journal.pone.0201760] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
We evaluated the chemical coding of the myenteric plexus in the proximal and distal intestine of gilthead sea bream (Sparus aurata), which represents one of the most farmed fish in the Mediterranean area. The presence of nitric oxide (NO), acetylcholine (ACh), serotonin (5-HT), calcitonin-gene-related peptide (CGRP), substance P (SP) and vasoactive intestinal peptide (VIP) containing neurons, was investigated in intestinal whole mount preparations of the longitudinal muscle with attached the myenteric plexus (LMMP) by means of immunohistochemical fluorescence staining. The main excitatory and inhibitory neurochemicals identified in intestinal smooth muscle were ACh, SP, 5HT, and NO, VIP, CGRP. Some neurons displayed morphological features of ascending and descending interneurons and of putative sensory neurons. The expression of these pathways in the two intestinal regions is largely superimposable, although some differences emerged, which may be relevant to the morphological properties of each region. The most important variances are the higher neuronal density and soma size in the proximal intestine, which may depend on the volume of the target tissue. Since in the fish gut the submucosal plexus is less developed, myenteric neurons substantially innervate also the submucosal and epithelial layers, which display a major thickness and surface in the proximal intestine. In addition, myenteric neurons containing ACh and SP, which mainly represent excitatory motor neurons and interneurons innervating the smooth muscle were more numerous in the distal intestine, possibly to sustain motility in the thicker smooth muscle coat. Overall, this study expands our knowledge of the intrinsic innervation that regulates intestinal secretion, absorption and motility in gilthead sea bream and provides useful background information for rational design of functional feeds aimed at improving fish gut health.
Collapse
Affiliation(s)
- Chiara Ceccotti
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, Pavia, Italy
| | - Genciana Terova
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Inter-University Centre for Research in Protein Biotechnologies "The Protein Factory"- Polytechnic University of Milan and University of Insubria, Varese, Italy
| |
Collapse
|
13
|
Lindborg JA, Niemi JP, Howarth MA, Liu KW, Moore CZ, Mahajan D, Zigmond RE. Molecular and cellular identification of the immune response in peripheral ganglia following nerve injury. J Neuroinflammation 2018; 15:192. [PMID: 29945607 PMCID: PMC6019520 DOI: 10.1186/s12974-018-1222-5] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 06/14/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Neuroinflammation accompanies neural trauma and most neurological diseases. Axotomy in the peripheral nervous system (PNS) leads to dramatic changes in the injured neuron: the cell body expresses a distinct set of genes known as regeneration-associated genes, the distal axonal segment degenerates and its debris is cleared, and the axons in the proximal segment form growth cones and extend neurites. These processes are orchestrated in part by immune and other non-neuronal cells. Macrophages in ganglia play an integral role in supporting regeneration. Here, we explore further the molecular and cellular components of the injury-induced immune response within peripheral ganglia. METHODS Adult male wild-type (WT) and Ccr2 -/- mice were subjected to a unilateral transection of the sciatic nerve and axotomy of the superior cervical ganglion (SCG). Antibody arrays were used to determine the expression of chemokines and cytokines in the dorsal root ganglion (DRG) and SCG. Flow cytometry and immunohistochemistry were utilized to identify the cellular composition of the injury-induced immune response within ganglia. RESULTS Chemokine expression in the ganglia differed 48 h after nerve injury with a large increase in macrophage inflammatory protein-1γ in the SCG but not in the DRG, while C-C class chemokine ligand 2 was highly expressed in both ganglia. Differences between WT and Ccr2 -/- mice were also observed with increased C-C class chemokine ligand 6/C10 expression in the WT DRG compared to C-C class chemokine receptor 2 (CCR2)-/- DRG and increased CXCL5 expression in CCR2-/- SCG compared to WT. Diminished macrophage accumulation in the DRG and SCG of Ccr2 -/- mice was found compared to WT ganglia 7 days after nerve injury. Interestingly, neutrophils were found in the SCG but not in the DRG. Cytokine expression, measured 7 days after injury, differed between ganglion type and genotype. Macrophage activation was assayed by colabeling ganglia with the anti-inflammatory marker CD206 and the macrophage marker CD68, and an almost complete colocalization of the two markers was found in both ganglia. CONCLUSIONS This study demonstrates both molecular and cellular differences in the nerve injury-induced immune response between DRG and SCG and between WT and Ccr2 -/- mice.
Collapse
Affiliation(s)
- Jane A Lindborg
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jon P Niemi
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Madeline A Howarth
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA.,Science and Engineering Program, Hathaway Brown School, Shaker Heights, OH, USA
| | - Kevin W Liu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Christian Z Moore
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Deepti Mahajan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Richard E Zigmond
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, USA. .,Present Address: Department Neurosciences, School of Medicine, 10900 Euclid Avenue, Robbins E701, Cleveland, OH, 44106-4975, USA.
| |
Collapse
|
14
|
Furukawa T, Takeuchi Y, Tanaka T, Yoshimaru K, Taguchi T, Tajiri T. Localized isolated hypoganglionosis in an infant. JOURNAL OF PEDIATRIC SURGERY CASE REPORTS 2018. [DOI: 10.1016/j.epsc.2017.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
15
|
Immunostaining for Hu C/D and CD56 is useful for a definitive histopathological diagnosis of congenital and acquired isolated hypoganglionosis. Virchows Arch 2017; 470:679-685. [PMID: 28424865 DOI: 10.1007/s00428-017-2128-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 03/03/2017] [Accepted: 04/11/2017] [Indexed: 12/12/2022]
Abstract
Isolated hypoganglionosis (IHG) has been proposed as a distinct entity with two subtypes: congenital IHG (CIHG) and acquired IHG (AIHG). However, due to the rarity of the disease and the lack of defining histological criteria, the concept of IHG is not widely accepted. We studied paraffin-embedded intestinal specimens from 79 patients diagnosed with Hirschsprung's disease (HD) (n = 49), CIHG (n = 25), and AIHG (n = 5) collected between January 1996 and December 2015. Histopathological diagnosis of HD, CIHG, and AIHG was confirmed by hematoxylin and eosin staining and immunohistochemical staining using Hu C/D and CD56. We evaluated (immuno)histopathological findings, counted the number of ganglion cells, and measured the size of Auerbach's plexus. Hu C/D labeled neuronal cell bodies, whereas CD56 was detected in all neuronal components. In HD, all ganglion cells in Auerbach's plexus in the normoganglionic segment (NGS) were immunoreactive for Hu C/D, whereas in the aganglionic segment (AGS), these were replaced by CD56-positive extrinsic nerve fibers and bundles. The number of ganglion cells in AIHG and CIHG was significantly lower than in the NGS of HD (p < 0.05). Auerbach's plexus was significantly smaller in CIHG (p < 0.05) but in AIHG equivalent to the NGS in HD. In summary, immunostaining for Hu C/D and CD56 is useful for definitive histopathological diagnosis of IHG.
Collapse
|
16
|
Roy-Carson S, Natukunda K, Chou HC, Pal N, Farris C, Schneider SQ, Kuhlman JA. Defining the transcriptomic landscape of the developing enteric nervous system and its cellular environment. BMC Genomics 2017; 18:290. [PMID: 28403821 PMCID: PMC5389105 DOI: 10.1186/s12864-017-3653-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 03/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Motility and the coordination of moving food through the gastrointestinal tract rely on a complex network of neurons known as the enteric nervous system (ENS). Despite its critical function, many of the molecular mechanisms that direct the development of the ENS and the elaboration of neural network connections remain unknown. The goal of this study was to transcriptionally identify molecular pathways and candidate genes that drive specification, differentiation and the neural circuitry of specific neural progenitors, the phox2b expressing ENS cell lineage, during normal enteric nervous system development. Because ENS development is tightly linked to its environment, the transcriptional landscape of the cellular environment of the intestine was also analyzed. RESULTS Thousands of zebrafish intestines were manually dissected from a transgenic line expressing green fluorescent protein under the phox2b regulatory elements [Tg(phox2b:EGFP) w37 ]. Fluorescence-activated cell sorting was used to separate GFP-positive phox2b expressing ENS progenitor and derivatives from GFP-negative intestinal cells. RNA-seq was performed to obtain accurate, reproducible transcriptional profiles and the unbiased detection of low level transcripts. Analysis revealed genes and pathways that may function in ENS cell determination, genes that may be identifiers of different ENS subtypes, and genes that define the non-neural cellular microenvironment of the ENS. Differential expression analysis between the two cell populations revealed the expected neuronal nature of the phox2b expressing lineage including the enrichment for genes required for neurogenesis and synaptogenesis, and identified many novel genes not previously associated with ENS development. Pathway analysis pointed to a high level of G-protein coupled pathway activation, and identified novel roles for candidate pathways such as the Nogo/Reticulon axon guidance pathway in ENS development. CONCLUSION We report the comprehensive gene expression profiles of a lineage-specific population of enteric progenitors, their derivatives, and their microenvironment during normal enteric nervous system development. Our results confirm previously implicated genes and pathways required for ENS development, and also identify scores of novel candidate genes and pathways. Thus, our dataset suggests various potential mechanisms that drive ENS development facilitating characterization and discovery of novel therapeutic strategies to improve gastrointestinal disorders.
Collapse
Affiliation(s)
- Sweta Roy-Carson
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Kevin Natukunda
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Hsien-Chao Chou
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA.,Present Address: National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Narinder Pal
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA.,Present address: North Central Regional Plant Introduction Station, 1305 State Ave, Ames, IA, 50014, USA
| | - Caitlin Farris
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA.,Present address: Pioneer Hi-Bred International, Johnson, IA, 50131, USA
| | - Stephan Q Schneider
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Julie A Kuhlman
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA, 50011, USA. .,642 Science II, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
17
|
Li Q, Michel K, Annahazi A, Demir IE, Ceyhan GO, Zeller F, Komorowski L, Stöcker W, Beyak MJ, Grundy D, Farrugia G, De Giorgio R, Schemann M. Anti-Hu antibodies activate enteric and sensory neurons. Sci Rep 2016; 6:38216. [PMID: 27905561 PMCID: PMC5131267 DOI: 10.1038/srep38216] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/07/2016] [Indexed: 02/07/2023] Open
Abstract
IgG of type 1 anti-neuronal nuclear antibody (ANNA-1, anti-Hu) specificity is a serological marker of paraneoplastic neurological autoimmunity (including enteric/autonomic) usually related to small-cell lung carcinoma. We show here that IgG isolated from such sera and also affinity-purified anti-HuD label enteric neurons and cause an immediate spike discharge in enteric and visceral sensory neurons. Both labelling and activation of enteric neurons was prevented by preincubation with the HuD antigen. Activation of enteric neurons was inhibited by the nicotinic receptor antagonists hexamethonium and dihydro-β-erythroidine and reduced by the P2X antagonist pyridoxal phosphate-6-azo (benzene-2,4-disulfonic acid (PPADS) but not by the 5-HT3 antagonist tropisetron or the N-type Ca-channel blocker ω-Conotoxin GVIA. Ca++ imaging experiments confirmed activation of enteric neurons but not enteric glia. These findings demonstrate a direct excitatory action of ANNA-1, in particular anti-HuD, on visceral sensory and enteric neurons, which involves nicotinic and P2X receptors. The results provide evidence for a novel link between nerve activation and symptom generation in patients with antibody-mediated gut dysfunction.
Collapse
Affiliation(s)
- Qin Li
- Human Biology, Technical University of Munich, Freising, Germany.,Department of Physiology, Shandong University School of Medicine, Jinan, China
| | - Klaus Michel
- Human Biology, Technical University of Munich, Freising, Germany
| | - Anita Annahazi
- Human Biology, Technical University of Munich, Freising, Germany
| | - Ihsan E Demir
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich; Munich, Germany
| | - Güralp O Ceyhan
- Department of Surgery, Klinikum Rechts der Isar, Technical University of Munich; Munich, Germany
| | | | - Lars Komorowski
- Institute for Experimental Immunology, Euroimmun AG, Lübeck, Germany
| | - Winfried Stöcker
- Institute for Experimental Immunology, Euroimmun AG, Lübeck, Germany
| | - Michael J Beyak
- GI Diseases Research Unit, Queen's University, Kingston, ON, Canada
| | - David Grundy
- Department of Biomedical Sciences, University of Sheffield, Sheffield, UK
| | | | - Roberto De Giorgio
- Department of Medical and Surgical Sciences and Center for Applied Biomedical Research, University of Bologna, Bologna, Italy
| | - Michael Schemann
- Human Biology, Technical University of Munich, Freising, Germany
| |
Collapse
|
18
|
Walter GC, Phillips RJ, McAdams JL, Powley TL. Individual sympathetic postganglionic neurons coinnervate myenteric ganglia and smooth muscle layers in the gastrointestinal tract of the rat. J Comp Neurol 2016; 524:2577-603. [PMID: 26850701 DOI: 10.1002/cne.23978] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 01/21/2016] [Accepted: 02/02/2016] [Indexed: 01/25/2023]
Abstract
A full description of the terminal architecture of sympathetic axons innervating the gastrointestinal (GI) tract has not been available. To label sympathetic fibers projecting to the gut muscle wall, dextran biotin was injected into the celiac and superior mesenteric ganglia (CSMG) of rats. Nine days postinjection, animals were euthanized and stomachs and small intestines were processed as whole mounts (submucosa and mucosa removed) to examine CSMG efferent terminals. Myenteric neurons were counterstained with Cuprolinic Blue; catecholaminergic axons were stained immunohistochemically for tyrosine hydroxylase. Essentially all dextran-labeled axons (135 of 136 sampled) were tyrosine hydroxylase-positive. Complete postganglionic arbors (n = 154) in the muscle wall were digitized and analyzed morphometrically. Individual sympathetic axons formed complex arbors of varicose neurites within myenteric ganglia/primary plexus and, concomitantly, long rectilinear arrays of neurites within circular muscle/secondary plexus or longitudinal muscle/tertiary plexus. Very few CSMG neurons projected exclusively (i.e., ∼100% of an arbor's varicose branches) to myenteric plexus (∼2%) or smooth muscle (∼14%). With less stringent inclusion criteria (i.e., ≥85% of an axon's varicose branches), larger minorities of neurons projected predominantly to either myenteric plexus (∼13%) or smooth muscle (∼27%). The majority (i.e., ∼60%) of all individual CSMG postganglionics formed mixed, heterotypic arbors that coinnervated extensively (>15% of their varicose branches per target) both myenteric ganglia and smooth muscle. The fact that ∼87% of all sympathetics projected either extensively or even predominantly to smooth muscle, while simultaneously contacting myenteric plexus, is consistent with the view that these neurons control GI muscle directly, if not exclusively. J. Comp. Neurol. 524:2577-2603, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Gary C Walter
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Robert J Phillips
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Jennifer L McAdams
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| | - Terry L Powley
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
19
|
Levandis G, Balestra B, Siani F, Rizzo V, Ghezzi C, Ambrosi G, Cerri S, Bonizzi A, Vicini R, Vairetti M, Ferrigno A, Pastoris O, Blandini F. Response of colonic motility to dopaminergic stimulation is subverted in rats with nigrostriatal lesion: relevance to gastrointestinal dysfunctions in Parkinson's disease. Neurogastroenterol Motil 2015; 27:1783-95. [PMID: 26433214 DOI: 10.1111/nmo.12691] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Accepted: 08/25/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND Constipation is extremely common in patients with Parkinson's disease (PD) and has been described in PD animal models. In this study, we investigated whether a PD-like degeneration of dopaminergic neurons of the substantia nigra can influence peristalsis in colonic segments of rats by impacting on enteric dopaminergic transmission. METHODS Male, Sprague-Dawley rats received a unilateral injection of neurotoxin 6-hydroxydopamine (6-OHDA), or saline, into the medial-forebrain-bundle. Peristaltic activity was recorded in isolated colonic segments, in baseline conditions and following exposure to combinations of D2 receptor (DRD2) agonist sumanirole and antagonist L-741626. Dopamine levels and DRD2 expression were assessed in the ileum and colon of animals. We also investigated the involvement of the dorsal motor nucleus of the vagus (DMV) - a potential relay station between central dopaminergic denervation and gastrointestinal (GI) dysfunction - by analyzing cytochrome c oxidase activity and FosB/DeltaFosB expression in DMV neurons. KEY RESULTS We observed profound alterations in the response of colonic segments of 6-OHDA lesioned animals to DRD2 stimulation. In fact, the inhibition of colonic peristalsis elicited by sumanirole in control rats was absent in 6-OHDA-lesioned animals. These animals also showed reduced DRD2 expression in the colon, along with elevation of dopamine levels. No significant changes were detected within the DMV. CONCLUSIONS & INFERENCES Our results demonstrate that selective lesion of the nigrostriatal dopaminergic pathway subverts the physiological response of the colon to dopaminergic stimulation, opening new perspectives in the comprehension and treatment of GI dysfunctions associated with PD.
Collapse
Affiliation(s)
- G Levandis
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino", National Neurological Institute, Pavia, Italy
| | - B Balestra
- Dept. of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - F Siani
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino", National Neurological Institute, Pavia, Italy
| | - V Rizzo
- Department of Molecular Medicine, IRCCS San Matteo Foundation, University of Pavia, Pavia, Italy
| | - C Ghezzi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino", National Neurological Institute, Pavia, Italy
| | - G Ambrosi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino", National Neurological Institute, Pavia, Italy
| | - S Cerri
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino", National Neurological Institute, Pavia, Italy
| | - A Bonizzi
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino", National Neurological Institute, Pavia, Italy
| | - R Vicini
- Dept. of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - M Vairetti
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - A Ferrigno
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - O Pastoris
- Dept. of Biology and Biotechnology, University of Pavia, Pavia, Italy
| | - F Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino", National Neurological Institute, Pavia, Italy
| |
Collapse
|
20
|
Powley TL, Hudson CN, McAdams JL, Baronowsky EA, Phillips RJ. Vagal Intramuscular Arrays: The Specialized Mechanoreceptor Arbors That Innervate the Smooth Muscle Layers of the Stomach Examined in the Rat. J Comp Neurol 2015; 524:713-37. [PMID: 26355387 DOI: 10.1002/cne.23892] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 01/14/2023]
Abstract
The fundamental roles that the stomach plays in ingestion and digestion notwithstanding, little morphological information is available on vagal intramuscular arrays (IMAs), the afferents that innervate gastric smooth muscle. To characterize IMAs better, rats were given injections of dextran biotin in the nodose ganglia, and, after tracer transport, stomach whole mounts were collected. Specimens were processed for avidin-biotin permanent labeling, and subsets of the whole mounts were immunohistochemically processed for c-Kit or stained with cuprolinic blue. IMAs (n = 184) were digitized for morphometry and mapping. Throughout the gastric muscle wall, IMAs possessed common phenotypic features. Each IMA was generated by a parent neurite arborizing extensively, forming an array of multiple (mean = 212) branches averaging 193 µm in length. These branches paralleled, and coursed in apposition with, bundles of muscle fibers and interstitial cells of Cajal. Individual arrays averaged 4.3 mm in length and innervated volumes of muscle sheet, presumptive receptive fields, averaging 0.1 mm(3) . Evaluated by region and by muscle sheet, IMAs displayed architectural adaptations to the different loci. A subset (32%) of circular muscle IMAs issued specialized polymorphic collaterals to myenteric ganglia, and a subset (41%) of antral longitudinal muscle IMAs formed specialized net endings associated with the serosal boundary. IMAs were concentrated in regional patterns that correlated with the unique biomechanical adaptations of the stomach, specifically proximal stomach reservoir functions and antral emptying operations. Overall, the structural adaptations and distributions of the IMAs were consonant with the hypothesized stretch receptor roles of the afferents.
Collapse
Affiliation(s)
- Terry L Powley
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, 47907-2081
| | - Cherie N Hudson
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, 47907-2081
| | - Jennifer L McAdams
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, 47907-2081
| | - Elizabeth A Baronowsky
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, 47907-2081
| | - Robert J Phillips
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana, 47907-2081
| |
Collapse
|
21
|
Evidence for neuronal and structural changes in submucous ganglia of patients with functional dyspepsia. Am J Gastroenterol 2015; 110:1205-15. [PMID: 26077177 DOI: 10.1038/ajg.2015.158] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 04/02/2015] [Indexed: 02/07/2023]
Abstract
OBJECTIVES An intact and well-functioning enteric nervous system is necessary to efficiently organize gut function. Functional gastrointestinal disorders are pathological entities in which gut function is impaired without a clearly established pathophysiology. On the basis of the relative ease with which intestinal biopsies can be obtained, and taking advantage of a recently developed optical recording technique, we evaluated whether functional neuronal defects exist in enteric nerves of patients with functional dyspepsia (FD). METHODS The submucous plexus isolated from duodenal biopsies taken from FD patients and control subjects was used to functionally and morphologically examine nerves and ganglionic architecture (neurons and glial cells). In light of previous studies reporting eosinophil and mast cell infiltration in the gut mucosa of FD patients, we also examined whether these cells infiltrated the submucous plexus and whether this correlated with neuronal activity and specific clinical symptoms. RESULTS We demonstrate that neuronal functioning is impaired in the submucous plexus of FD patients, as shown by decreased calcium responses to depolarization and electrical stimulation. Glial (S100) and neuronal (HuCD) markers show signs of gliosis, altered ganglionic architecture, and neuronal abnormalities in the submucous plexus of FD patients. We found that eosinophils and mast cells infiltrated the submucous layer of FD patients to a much larger extent than in controls. A significant correlation was found between the number of these cells and the calcium transient amplitudes measured in submucous ganglia. CONCLUSIONS We provide the first direct evidence that FD is characterized by functional and structural abnormalities within the submucous ganglion plexus, which may be of future predictive and diagnostic value in the treatment of FD patients.
Collapse
|
22
|
Genç B, Lagrimas AKB, Kuru P, Hess R, Tu MW, Menichella DM, Miller RJ, Paller AS, Özdinler PH. Visualization of Sensory Neurons and Their Projections in an Upper Motor Neuron Reporter Line. PLoS One 2015. [PMID: 26222784 PMCID: PMC4519325 DOI: 10.1371/journal.pone.0132815] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Visualization of peripheral nervous system axons and cell bodies is important to understand their development, target recognition, and integration into complex circuitries. Numerous studies have used protein gene product (PGP) 9.5 [a.k.a. ubiquitin carboxy-terminal hydrolase L1 (UCHL1)] expression as a marker to label sensory neurons and their axons. Enhanced green fluorescent protein (eGFP) expression, under the control of UCHL1 promoter, is stable and long lasting in the UCHL1-eGFP reporter line. In addition to the genetic labeling of corticospinal motor neurons in the motor cortex and degeneration-resistant spinal motor neurons in the spinal cord, here we report that neurons of the peripheral nervous system are also fluorescently labeled in the UCHL1-eGFP reporter line. eGFP expression is turned on at embryonic ages and lasts through adulthood, allowing detailed studies of cell bodies, axons and target innervation patterns of all sensory neurons in vivo. In addition, visualization of both the sensory and the motor neurons in the same animal offers many advantages. In this report, we used UCHL1-eGFP reporter line in two different disease paradigms: diabetes and motor neuron disease. eGFP expression in sensory axons helped determine changes in epidermal nerve fiber density in a high-fat diet induced diabetes model. Our findings corroborate previous studies, and suggest that more than five months is required for significant skin denervation. Crossing UCHL1-eGFP with hSOD1G93A mice generated hSOD1G93A-UeGFP reporter line of amyotrophic lateral sclerosis, and revealed sensory nervous system defects, especially towards disease end-stage. Our studies not only emphasize the complexity of the disease in ALS, but also reveal that UCHL1-eGFP reporter line would be a valuable tool to visualize and study various aspects of sensory nervous system development and degeneration in the context of numerous diseases.
Collapse
Affiliation(s)
- Barış Genç
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Amiko Krisa Bunag Lagrimas
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Pınar Kuru
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Robert Hess
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Michael William Tu
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Daniela Maria Menichella
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Richard J. Miller
- Department of Molecular Pharmacology and Biological Chemistry, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - Amy S. Paller
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
- Departments of Dermatology and Pediatrics, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
- Skin Disease Research Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
- Center for Genetic Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
| | - P. Hande Özdinler
- Davee Department of Neurology and Clinical Neurological Sciences, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Feinberg School of Medicine, Chicago, IL, United States of America
- Cognitive Neurology and Alzheimer's Disease Center, Northwestern University, Chicago, IL, United States of America
- * E-mail:
| |
Collapse
|
23
|
McVey Neufeld KA, Perez-Burgos A, Mao YK, Bienenstock J, Kunze WA. The gut microbiome restores intrinsic and extrinsic nerve function in germ-free mice accompanied by changes in calbindin. Neurogastroenterol Motil 2015; 27:627-36. [PMID: 25727007 DOI: 10.1111/nmo.12534] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 02/02/2015] [Indexed: 02/08/2023]
Abstract
BACKGROUND The microbiome is essential for normal myenteric intrinsic primary afferent neuron (IPAN) excitability. These neurons control gut motility and modulate gut-brain signaling by exciting extrinsic afferent fibers innervating the enteric nervous system via an IPAN to extrinsic fiber sensory synapse. We investigated effects of germ-free (GF) status and conventionalization on extrinsic sensory fiber discharge in the mesenteric nerve bundle and IPAN electrophysiology, and compared these findings with those from specific pathogen-free (SPF) mice. As we have previously shown that the IPAN calcium-dependent slow afterhyperpolarization (sAHP) is enhanced in GF mice, we also examined the expression of the calcium-binding protein calbindin in these neurons in these different animal groups. METHODS IPAN sAHP and mesenteric nerve multiunit discharge were recorded using ex vivo jejunal gut segments from SPF, GF, or conventionalized (CONV) mice. IPANs were excited by adding 5 μM TRAM-34 to the serosal superfusate. We probed for calbindin expression using immunohistochemical techniques. KEY RESULTS SPF mice had a 21% increase in mesenteric nerve multiunit firing rate and CONV mice a 41% increase when IPANs were excited by TRAM-34. For GF mice, this increase was barely detectable (2%). TRAM-34 changed sAHP area under the curve by -77 for SPF, +3 for GF, or -54% for CONV animals. Calbindin-immunopositive neurons per myenteric ganglion were 36% in SPF, 24% in GF, and 52% in CONV animals. CONCLUSIONS & INFERENCES The intact microbiome is essential for normal intrinsic and extrinsic nerve function and gut-brain signaling.
Collapse
Affiliation(s)
- K A McVey Neufeld
- McMaster Brain-Body Institute at St Joseph's Healthcare, Hamilton, ON, Canada; Department of Psychiatry and Behavioral Neurosciences, McMaster University, Hamilton, ON, Canada
| | | | | | | | | |
Collapse
|
24
|
Aqueous Extract of Agaricus blazei Murrill Prevents Age-Related Changes in the Myenteric Plexus of the Jejunum in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 2015:287153. [PMID: 25960748 PMCID: PMC4415631 DOI: 10.1155/2015/287153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/15/2014] [Accepted: 12/16/2014] [Indexed: 01/27/2023]
Abstract
This study evaluated the effects of the supplementation with aqueous extract of Agaricus blazei Murrill (ABM) on biometric and blood parameters and quantitative morphology of the myenteric plexus and jejunal wall in aging Wistar rats. The animals were euthanized at 7 (C7), 12 (C12 and CA12), and 23 months of age (C23 and CA23). The CA12 and CA23 groups received a daily dose of ABM extract (26 mg/animal) via gavage, beginning at 7 months of age. A reduction in food intake was observed with aging, with increases in the Lee index, retroperitoneal fat, intestinal length, and levels of total cholesterol and total proteins. Aging led to a reduction of the total wall thickness, mucosa tunic, villus height, crypt depth, and number of goblet cells. In the myenteric plexus, aging quantitatively decreased the population of HuC/D(+) neuronal and S100(+) glial cells, with maintenance of the nNOS(+) nitrergic subpopulation and increase in the cell body area of these populations. Supplementation with the ABM extract preserved the myenteric plexus in old animals, in which no differences were detected in the density and cell body profile of neurons and glial cells in the CA12 and CA23 groups, compared with C7 group. The supplementation with the aqueous extract of ABM efficiently maintained myenteric plexus homeostasis, which positively influenced the physiology and prevented the death of the neurons and glial cells.
Collapse
|
25
|
Swaminathan M, Oron AP, Chatterjee S, Piper H, Cope-Yokoyama S, Chakravarti A, Kapur RP. Intestinal Neuronal Dysplasia-Like Submucosal Ganglion Cell Hyperplasia at the Proximal Margins of Hirschsprung Disease Resections. Pediatr Dev Pathol 2015; 18:466-76. [PMID: 26699691 PMCID: PMC4809533 DOI: 10.2350/15-07-1675-oa.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Intestinal neuronal dysplasia type B (IND) denotes an increased proportion of hyperplastic submucosal ganglia, as resolved histochemically in 15-μm-thick frozen sections. IND has been reported proximal to the aganglionic segment in patients with Hirschsprung disease (HSCR) and is putatively associated with a higher rate of postsurgical dysmotility. We developed and validated histological criteria to diagnose IND-like submucosal ganglion cell hyperplasia (IND-SH) in paraffin sections and used the approach to study the incidence and clinical and/or genetic associations of IND-SH at the proximal margins of HSCR pull-through resection specimens. Full-circumference paraffin sections from the proximal margins of 64 HSCR colonic pull-through specimens and 24 autopsy controls were immunostained for neuron-specific Hu antigen, and nucleated ganglion cells in each submucosal ganglion were counted. In controls, an age-related decline in the relative abundance of "giant" ganglia (≥7 nucleated Hu-positive [Hu+] ganglion cells) was observed. A conservative diagnostic threshold for IND-SH (control mean ± 3× standard deviation) was derived from 15 controls less than 25 weeks of age. No control exceeded this threshold, whereas in the same age range, IND-SH was observed at the proximal margins in 15% (7 of 46) of HSCR resections, up to 15 cm proximal to the aganglionic segment. No significant correlation was observed between IND-SH and length of or distance from the aganglionic segment, sex, trisomy 21, RET or SEMA3C/D polymorphisms, or clinical outcome, but analysis of more patients, with better long-term follow-up will be required to clarify the significance of this histological phenotype.
Collapse
Affiliation(s)
| | | | - Sumantra Chatterjee
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine ,Balimore, MD
| | - Hannah Piper
- University of Texas Southwestern, Children's Health, Dallas, TX
| | | | - Aravinda Chakravarti
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine ,Balimore, MD
| | - Raj P. Kapur
- Seattle Children's Research Institute, Seattle, WA,University of Washington, Pathology, Seattle, WA
| |
Collapse
|
26
|
Welch MG, Margolis KG, Li Z, Gershon MD. Oxytocin regulates gastrointestinal motility, inflammation, macromolecular permeability, and mucosal maintenance in mice. Am J Physiol Gastrointest Liver Physiol 2014; 307:G848-62. [PMID: 25147234 PMCID: PMC4200316 DOI: 10.1152/ajpgi.00176.2014] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 08/18/2014] [Indexed: 01/31/2023]
Abstract
Enteric neurons express oxytocin (OT); moreover, enteric neurons and enterocytes express developmentally regulated OT receptors (OTRs). Although OT (with secretin) opposes intestinal inflammation, physiological roles played by enteric OT/OTR signaling have not previously been determined. We tested hypotheses that OT/OTR signaling contributes to enteric nervous system (ENS)-related gastrointestinal (GI) physiology. GI functions and OT effects were compared in OTR-knockout (OTRKO) and wild-type (WT) mice. Stool mass and water content were greater in OTRKO mice than in WT. GI transit time in OTRKO animals was faster than in WT; OT inhibited in vitro generation of ENS-dependent colonic migrating motor complexes in WT but not in OTRKO mice. Myenteric neurons were hyperplastic in OTRKO animals, and mucosal exposure to cholera toxin (CTX) in vitro activated Fos in more myenteric neurons in OTRKO than WT than in WT mice; OT inhibited the CTX response in WT but not in OTRKO mice. Villi and crypts were shorter in OTRKO than in WT mice, and transit-amplifying cell proliferation in OTRKO crypts was deficient. Macromolecular intestinal permeability in OTRKO was greater than WT mice, and experimental colitis was more severe in OTRKO mice; moreover, OT protected WT animals from colitis. Observations suggest that OT/OTR signaling acts as a brake on intestinal motility, decreases mucosal activation of enteric neurons, and promotes enteric neuronal development and/or survival. It also regulates proliferation of crypt cells and mucosal permeability; moreover OT/OTR signaling is protective against inflammation. Oxytocinergic signaling thus appears to play an important role in multiple GI functions that are subject to neuronal regulation.
Collapse
Affiliation(s)
- Martha G Welch
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Kara G Margolis
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Zhishan Li
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| | - Michael D Gershon
- Department of Psychiatry, Pediatrics, and Pathology and Cell Biology, Columbia University, College of Physicians and Surgeons, New York, New York
| |
Collapse
|
27
|
Desmet AS, Cirillo C, Vanden Berghe P. Distinct subcellular localization of the neuronal marker HuC/D reveals hypoxia-induced damage in enteric neurons. Neurogastroenterol Motil 2014; 26:1131-43. [PMID: 24861242 DOI: 10.1111/nmo.12371] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/29/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Correct neuronal identification is essential to study neurons in health and disease. Although commonly used as pan-neuronal marker, HuC/D's expression pattern varies substantially between healthy and (patho)physiological conditions. This heterogenic labeling has received very little attention. We sought to investigate the subcellular HuC/D localization in enteric neurons in different conditions. METHODS The localization of neuronal RNA-binding proteins HuC/D was investigated by immunohistochemistry in the mouse myenteric plexus using different toxins and caustic agents. Preparations were also stained with Sox10 and glial fibrillary acidic protein (GFAP) antibodies to assess enteric glial cell appearance. KEY RESULTS Mechanically induced tissue damage, interference with the respiratory chain and oxygen (O2 ) deprivation increased nuclear HuC/D immunoreactivity. This effect was paralleled by a distortion of the GFAP-labeled glial network along with a loss of Sox10 expression and coincided with the activation of a non-apoptotic genetic program. Chemically induced damage and specific noxious stimuli did not induce a change in HuC/D immunoreactivity, supporting the specific nature of the nuclear HuC/D localization. CONCLUSIONS & INFERENCES HuC/D is not merely a pan-neuronal marker but its subcellular localization also reflects the condition of a neuron at the time of fixation. The functional meaning of this change in HuC/D localization is not entirely clear, but disturbance in O2 supply in combination with the support of enteric glial cells seems to play a crucial role. The molecular consequence of changes in HuC/D expression needs to be further investigated.
Collapse
Affiliation(s)
- A-S Desmet
- Laboratory for Enteric NeuroScience (LENS), Translational Research Center for GastroIntestinal Disorders (TARGID), University of Leuven, Leuven, Belgium
| | | | | |
Collapse
|
28
|
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
|
29
|
Powley TL, Hudson CN, McAdams JL, Baronowsky EA, Martin FN, Mason JK, Phillips RJ. Organization of vagal afferents in pylorus: mechanoreceptors arrayed for high sensitivity and fine spatial resolution? Auton Neurosci 2014; 183:36-48. [PMID: 24656895 DOI: 10.1016/j.autneu.2014.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 01/16/2023]
Abstract
The pylorus is innervated by vagal mechanoreceptors that project to gastrointestinal smooth muscle, but the distributions and specializations of vagal endings in the sphincter have not been fully characterized. To evaluate their organization, the neural tracer dextran biotin was injected into the nodose ganglia of rats. Following tracer transport, animals were perfused, and their pylori and antra were prepared as whole mounts. Specimens were processed to permanently label the tracer, and subsets were counterstained with Cuprolinic blue or immunostained for c-Kit. Intramuscular arrays (IMAs) in the circular muscle comprised the principal vagal afferent innervation of the sphincter. These pyloric ring IMAs were densely distributed and evidenced a variety of structural specializations. Morphometric comparisons between the arbors innervating the pylorus and a corresponding sample of IMAs in the adjacent antral circular muscle highlighted that sphincter IMAs branched profusely, forming more than twice as many branches as did antral IMAs (means of 405 vs. 165, respectively), and condensed their numerous neurites into compact receptive fields (∼48% of the area of antral IMAs) deep in the circular muscle (∼6μm above the submucosa). Separate arbors of IMAs in the sphincter interdigitated and overlapped to form a 360° band of mechanoreceptors encircling the pyloric canal. The annulus of vagal IMA arbors, putative stretch receptors tightly intercalated in the sphincter ring and situated near the lumen of the pyloric canal, creates an architecture with the potential to generate gut reflexes on the basis of pyloric sensory maps of high sensitivity and fine spatial resolution.
Collapse
Affiliation(s)
- Terry L Powley
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States.
| | - Cherie N Hudson
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States
| | - Jennifer L McAdams
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States
| | - Elizabeth A Baronowsky
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States
| | - Felecia N Martin
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States
| | - Jacqueline K Mason
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States
| | - Robert J Phillips
- Purdue University, Department of Psychological Sciences, West Lafayette, IN 47907-2081, United States.
| |
Collapse
|
30
|
Kapur RP. Calretinin-immunoreactive mucosal innervation in very short-segment Hirschsprung disease: a potentially misleading observation. Pediatr Dev Pathol 2014; 17:28-35. [PMID: 24168728 DOI: 10.2350/13-10-1387-oa.1] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Absent calretinin-immunoreactive (CRir) mucosal innervation in aganglionic rectal biopsies is considered a useful diagnostic finding for Hirschsprung disease. Analysis of a series of rectosigmoid resections from patients with short-segment (>2-cm aganglionic, n = 9) and very short-segment (≤2-cm aganglionic, n = 9) Hirschsprung disease demonstrates that CRir mucosal nerves extend into the proximal 1-2 cm of aganglionic bowel, where their presence in distal rectal biopsies could complicate diagnosis of very short-segment disease. Indeed, retrospective analysis of preoperative, aganglionic, distal rectal biopsies from 4 of 9 patients with very short-segment Hirschsprung disease revealed CRir mucosal innervation. Accurate diagnosis was possible based on generous histopathological submucosal sampling to exclude ganglion cells and the presence of abundant large-caliber submucosal nerves (more than 4 nerves >30 µm thick/×200 field or more than 2 nerves >40 µm thick/×200 field). Absent CRir mucosal innervation supports the diagnosis of Hirschsprung disease, but the presence of CRir mucosal nerves does not exclude aganglionosis, especially in distal rectal biopsies from patients with very short-segment Hirschsprung disease.
Collapse
Affiliation(s)
- Raj P Kapur
- Department of Laboratories, 0C.8.720, Seattle Children's Hospital, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| |
Collapse
|
31
|
Zhang Y, Bitner D, Pontes Filho AA, Li F, Liu S, Wang H, Yang F, Adhikari S, Gordon J, Srinivasan S, Hu W. Expression and function of NIK- and IKK2-binding protein (NIBP) in mouse enteric nervous system. Neurogastroenterol Motil 2014; 26:77-97. [PMID: 24011459 PMCID: PMC3962790 DOI: 10.1111/nmo.12234] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Accepted: 08/15/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND NIK- and IKK2-binding protein (NIBP)/TRAPPC9 is expressed in brain neurons, and human NIBP mutations are associated with neurodevelopmental disorders. The cellular distribution and function of NIBP in the enteric nervous system (ENS) remain unknown. METHODS Western blot and reverse transcription-polymerase chain reaction analysis were used respectively to identify the protein and mRNA expression of NIBP and other neuronal markers. Multi-labeled immunofluorescent microscopy and confocal image analysis were used to examine the cellular distribution of NIBP-like immunoreactivity (IR) in whole mount intestine. Enteric neuronal cell line (ENC) was infected with lentivirus carrying NIBP or its shRNA expression vectors and treated with vehicle or tumor necrosis factor (TNF)α. KEY RESULTS NIBP is expressed at both mRNA and protein levels in different regions and layers of the mouse intestine. NIBP-like-IR was co-localized with various neuronal markers, but not with glial, smooth muscular, or interstitial cells of Cajal markers. A small population of NIBP-expressing cells and fibers in extra-ganglionic and intra-ganglionic area were negative for pan-neuronal markers HuD or Peripherin. Relatively high NIBP-like-IR was found in 35-44% of myenteric neurons and 9-10% of submucosal neurons. Approximately 98%, 87%, and 43% of these relatively high NIBP-expressing neurons were positive for choline acetyltransferase, neuronal nitric oxide synthase and Calretinin, respectively. NIBP shRNA knockdown in ENC inhibited TNFα-induced NFκB activation and neuronal differentiation, whereas NIBP overexpression promoted it. CONCLUSIONS & INFERENCES NIBP is extensively expressed in the ENS with relatively high level in a subpopulation of enteric neurons. Various NIBP expression levels in different neurons may represent dynamic trafficking or posttranslational modification of NIBP in some functionally active neurons and ultimately regulate ENS plasticity.
Collapse
Affiliation(s)
- Yonggang Zhang
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Daniel Bitner
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Adalto Alfredo Pontes Filho
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Fang Li
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Shu Liu
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Hong Wang
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Fan Yang
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Sam Adhikari
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Jennifer Gordon
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| | - Shanthi Srinivasan
- Division of Digestive Diseases, Emory University, 615 Michael St., Atlanta, GA 30322 and Atlanta VAMC, Decatur, GA, 30331
| | - Wenhui Hu
- Department of Neuroscience, Temple University School of Medicine, 3500 N Broad Street, Philadelphia, PA 19140, USA
| |
Collapse
|
32
|
Phillips RJ, Martin FN, Billingsley CN, Powley TL. Alpha-synuclein expression patterns in the colonic submucosal plexus of the aging Fischer 344 rat: implications for biopsies in aging and neurodegenerative disorders? Neurogastroenterol Motil 2013; 25:e621-33. [PMID: 23809578 PMCID: PMC3735646 DOI: 10.1111/nmo.12176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 06/01/2013] [Indexed: 12/17/2022]
Abstract
BACKGROUND This experiment assessed normative expression patterns of alpha-synuclein (SYNC), including ganglionic remodeling and development of SYNC pathologies, in the submucosal plexus (SMP) of the colon during healthy aging. The observations address age-associated changes in bowel function and are relevant to evaluations of SMP-containing colonic biopsies for SYNC or synucleinopathies associated with aging and peripheral neurodegenerative diseases. METHODS Colonic submucosal whole mounts from groups of virgin male Fischer 344 rats (n ≥ 8 per group) at 4, 8, 16, and 24 months of age were processed immunohistochemically for SYNC and the pan-neuronal marker HuC/D. In addition, macrophages immunoreactive for MHCII were examined. Stereological protocols were used to generate unbiased estimates of neuron density, neurons per ganglion, neurons per ganglionic area, and neuron size. KEY RESULTS The protein SYNC was expressed in a subpopulation of SMP neurons, in both nucleus and cytoplasm. The general age-associated pattern across different cell counts was an increase in the number of SYNC+ neurons between 4 and 8 months of age, with progressively decreasing numbers of both SYNC+ and SYNC- neurons over the remaining lifespan. The soma size of SYNC+ neurons increased progressively with age. Aggregated SYNC occurred in the aging SMP, and macrophages with alternatively activated profiles were located adjacent to pathological SYNC deposits, consistent with ongoing phagocytosis. CONCLUSIONS & INFERENCES Changes in SYNC expression with age, including a baseline of accumulating synucleinopathies in the healthy aging SMP, need to be considered when interpreting either functional disturbances or biopsies of the aging colon.
Collapse
Affiliation(s)
- Robert J. Phillips
- Corresponding author: Robert J Phillips Purdue University 703 Third Street West Lafayette, IN 47907-2091 Phone: 765-494-6268 Fax: 765-496-1264
| | | | | | | |
Collapse
|
33
|
High-fat diet ingestion correlates with neuropathy in the duodenum myenteric plexus of obese mice with symptoms of type 2 diabetes. Cell Tissue Res 2013; 354:381-94. [PMID: 23881404 DOI: 10.1007/s00441-013-1681-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 06/13/2013] [Indexed: 02/08/2023]
Abstract
Obesity and type 2 diabetes are increasing in prevalence at an alarming rate in developed and developing nations and over 50% of patients with prolonged stages of disease experience forms of autonomic neuropathy. These patients have symptoms indicating disrupted enteric nervous system function including gastric discomfort, gastroparesis and intestinal dysmotility. Previous assessments have examined enteric neuronal injury within either type 1 diabetic or transgenic type 2 diabetic context. This study aims to assess damage to myenteric neurons within the duodenum of high-fat diet ingesting mice experiencing symptoms of type 2 diabetes, as this disease context is most parallel to the human condition and disrupted duodenal motility underlies negative gastrointestinal symptoms. Mice fed a high-fat diet developed symptoms of obesity and diabetes by 4 weeks. After 8 weeks, the total number of duodenal myenteric neurons and the synaptophysin density index were reduced and transmission electron microscopy showed axonal swelling and loss of neurofilaments and microtubules, suggesting compromised neuronal health. High-fat diet ingestion correlated with a loss of neurons expressing VIP and nNOS but did not affect the expression of ChAT, substance P, calbindin and CGRP. These results correlate high-fat diet ingestion, obesity and type 2 diabetes symptoms with a loss of duodenal neurons, biasing towards those with inhibitory nature. This pathology may underlie dysmotility and other negative GI symptoms experienced by human type 2 diabetic and obese patients.
Collapse
|
34
|
Gamage PPKM, Ranson RN, Patel BA, Yeoman MS, Saffrey MJ. Myenteric neuron numbers are maintained in aging mouse distal colon. Neurogastroenterol Motil 2013; 25:e495-e505. [PMID: 23517051 DOI: 10.1111/nmo.12114] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 02/15/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND Age-associated myenteric neuronal loss has been described in several species. In some studies,cholinergic neurons have been reported to be selectively vulnerable, whereas nitrergic neurons are spared. Aging of the mouse enteric nervous system(ENS) and the subtypes of mouse myenteric neurons that may be lost have been little studied. We therefore investigated changes in the numbers of total neurons and two neuronal subpopulations in the mouse distal colon during aging. METHODS Wholemount preparations from 3–4-, 12–13-, 18–19-, and 24–25-month-old C57BL/6 mice were double immunolabeled with HuC/D antibody to identify the total neuronal population and antisera to either calbindin or neuronal nitric oxide synthase (nNOS) to identify myenteric neuronal subpopulations. Samples were analyzed by confocal microscopy. New procedures were employed to ensure unbiased counting and to correct for changes in gut dimensions with age and stretch during sample preparation. The density of nerve fibers in the tertiary plexus was also studied. KEY RESULTS No significant change in numbers of total neurons or of either subpopulation with age was measured, but because of gut growth, the density of myenteric neurons decreased between 3–4 and 12–13 months. The density of nNOS-immunoreactive nerve fibers in the tertiary plexus increased significantly with age, up to 18–19 months. Numerous swollen processes of CB and nNOS-immunoreactive neurons were observed in 18–19- and 24–25-month-old animals. Conclusions &Inferences These results indicate that aging does not result in a loss of myenteric neurons in mouse distal colon at the ages studied, although neurodegenerative changes, which may impact on neuronal function, do occur.
Collapse
Affiliation(s)
- P. P. K. M. Gamage
- Department of Life, Health and Chemical Sciences; Biomedical Research Network; Open University; Walton Hall; Milton Keynes; MK7 6AA; UK
| | - R. N. Ranson
- Faculty of Health and Life Sciences; Northumbria University; Ellison Building; Newcastle upon Tyne; NE1 8ST; UK
| | - B. A. Patel
- School of Pharmacy and Biomolecular Sciences, Huxley Building; University of Brighton; Lewes Road; Brighton; BN2 4GJ; UK
| | - M. S. Yeoman
- School of Pharmacy and Biomolecular Sciences, Huxley Building; University of Brighton; Lewes Road; Brighton; BN2 4GJ; UK
| | - M. J. Saffrey
- Department of Life, Health and Chemical Sciences; Biomedical Research Network; Open University; Walton Hall; Milton Keynes; MK7 6AA; UK
| |
Collapse
|
35
|
Kapur RP, Kennedy AJ. Histopathologic delineation of the transition zone in short-segment Hirschsprung disease. Pediatr Dev Pathol 2013; 16:252-66. [PMID: 23495711 DOI: 10.2350/12-12-1282-oa.1] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Failure to completely resect the transition zone (TZ) between aganglionic and neuroanatomically normal bowel ("TZ pull-through") is considered one reason for postoperative obstructive symptoms in Hirschsprung disease (HD). Despite years of study, the proximal boundary of the TZ remains nebulous, complicated by discordant, often subjective, histopathologic definitions. In order to objectively delineate the TZ, transverse sections at 1 cm intervals from the rectums of 9 non-HD autopsy subjects and resections from 15 infants with short-segment HD were immunostained with Hu (ganglion cell bodies) and glucose transporter 1 (Glut1) (perineurium of extrinsic nerves), and 6 putative features of TZ were examined: (1) aganglionosis of ≥1/8th circumference; (2) myenteric or submucosal hypoganglionosis; (3) hypertrophic submucosal nerves; (4) Glut1+ submucosal innervation; (5) submucosal hyperganglionosis; and (6) "ectopic" ganglia in lamina propria, muscularis propria, or serosa. In non-HD controls, Glut1+ submucosal innervation, hypertrophic nerves, partial circumferential aganglionosis, and hypoganglionosis were absent or restricted to the distal 2 cm. In contrast, all 6 neuropathologic features of TZ were identified proximal to the aganglionic segment in the majority of HD resections, but the length of the TZ ranged from 0 to 12 cm, depending on which neuropathologic feature was considered. Excluding submucosal hyperganglionosis and ectopic ganglia, the TZ was generally ≤5 cm. Many features of TZ cannot be excluded intraoperatively with a biopsy or a full-circumference frozen section. However, partial circumferential aganglionosis, severe myenteric hypoganglionosis, and hypertrophic submucosal nerves can, and probably should, be assessed in full-circumference frozen sections of the proximal resection margin, to reduce the likelihood of TZ pull-through.
Collapse
Affiliation(s)
- Raj P Kapur
- Department of Laboratories, Seattle Children's Hospital, Seattle, WA, USA.
| | | |
Collapse
|
36
|
Stenkamp-Strahm C, Patterson S, Boren J, Gericke M, Balemba O. High-fat diet and age-dependent effects on enteric glial cell populations of mouse small intestine. Auton Neurosci 2013; 177:199-210. [PMID: 23726157 DOI: 10.1016/j.autneu.2013.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Revised: 04/26/2013] [Accepted: 04/29/2013] [Indexed: 02/09/2023]
Abstract
Diabetes and obesity are increasing in prevalence at an alarming rate throughout the world. Autonomic diabetic neuropathy is evident in individuals that experience a long-standing diabetic disease state, and gastrointestinal (GI) dysmotility is thought to be the outcome of neuropathies within the enteric nervous system (ENS) of these patients. To date, an analysis of enteric glial cell population changes during diabetic symptoms has not been performed, and may bring insight into disease pathology and neuropathy, given glial cell implications in gastrointestinal and neuronal homeostasis. Diabetes and obesity were monitored in C57Bl/6J mice fed a 72% high-fat diet, and duodenal glial expression patterns were evaluated by immunohistochemistry and RT-PCR for S100β, Sox10 and GFAP proteins and transcripts, as well as transmission electron microscopy (TEM). The high-fat diet caused obesity, hyperglycemia and insulin resistance after 4 weeks. These changes were associated with a significant decline in the area density indices of mucosa-associated glial cell networks, evidenced by S100β staining at 8 and 20 weeks. All three markers and TEM showed that myenteric glial cells were unaffected by early and late disease periods. However, analysis of Sox10 transcript expression and immunoreactivity showed a diet independent, age-associated decline in glial cell populations. This is the first study showing that mucosal glia cell damage occurs during diabetic symptoms, suggesting that mucosal enteric glia injury may have a pathophysiological significance during this disease. Our results also provide support for age-associated changes in longitudinal studies of enteric glial cells.
Collapse
|
37
|
Saffrey MJ. Cellular changes in the enteric nervous system during ageing. Dev Biol 2013; 382:344-55. [PMID: 23537898 DOI: 10.1016/j.ydbio.2013.03.015] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Revised: 02/22/2013] [Accepted: 03/19/2013] [Indexed: 02/06/2023]
Abstract
The intrinsic neurons of the gut, enteric neurons, have an essential role in gastrointestinal functions. The enteric nervous system is plastic and continues to undergo changes throughout life, as the gut grows and responds to dietary and other environmental changes. Detailed analysis of changes in the ENS during ageing suggests that enteric neurons are more vulnerable to age-related degeneration and cell death than neurons in other parts of the nervous system, although there is considerable variation in the extent and time course of age-related enteric neuronal loss reported in different studies. Specific neuronal subpopulations, particularly cholinergic myenteric neurons, may be more vulnerable than others to age-associated loss or damage. Enteric degeneration and other age-related neuronal changes may contribute to gastrointestinal dysfunction that is common in the elderly population. Evidence suggests that caloric restriction protects against age-associated loss of enteric neurons, but recent advances in the understanding of the effects of the microbiota and the complex interactions between enteric ganglion cells, mucosal immune system and intestinal epithelium indicate that other factors may well influence ageing of enteric neurons. Much remains to be understood about the mechanisms of neuronal loss and damage in the gut, although there is evidence that reactive oxygen species, neurotrophic factor dysregulation and/or activation of a senescence associated phenotype may be involved. To date, there is no evidence for ongoing neurogenesis that might replace dying neurons in the ageing gut, although small local sites of neurogenesis would be difficult to detect. Finally, despite the considerable evidence for enteric neurodegeneration during ageing, and evidence for some physiological changes in animal models, the ageing gut appears to maintain its function remarkably well in animals that exhibit major neuronal loss, indicating that the ENS has considerable functional reserve.
Collapse
Affiliation(s)
- M Jill Saffrey
- Department Life, Health & Chemical Sciences, Open University, Walton Hall, Milton Keynes MK7 6AA, United Kingdom.
| |
Collapse
|
38
|
Eisenman ST, Gibbons SJ, Singh RD, Bernard CE, Wu J, Sarr MG, Kendrick ML, Larson DW, Dozois EJ, Shen KR, Farrugia G. Distribution of TMEM100 in the mouse and human gastrointestinal tract--a novel marker of enteric nerves. Neuroscience 2013; 240:117-28. [PMID: 23485812 DOI: 10.1016/j.neuroscience.2013.02.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 01/30/2013] [Accepted: 02/14/2013] [Indexed: 02/02/2023]
Abstract
Identification of markers of enteric neurons has contributed substantially to our understanding of the development, normal physiology, and pathology of the gut. Previously identified markers of the enteric nervous system can be used to label all or most neuronal structures or for examining individual cells by labeling just the nucleus or cell body. Most of these markers are excellent but have some limitations. Transmembrane protein 100 (TMEM100) is a gene at locus 17q32 encoding a 134-amino acid protein with two hypothetical transmembrane domains. TMEM100 expression has not been reported in adult mammalian tissues but does appear in the ventral neural tube of embryonic mice and plays a role in signaling pathways associated with development of the enteric nervous system. We showed that TMEM100 messenger RNA is expressed in the gastrointestinal tract and demonstrated that TMEM100 is a membrane-associated protein. Furthermore TMEM100 immunoreactivity was restricted to enteric neurons and vascular tissue in the muscularis propria of all regions of the mouse and human gastrointestinal tract. TMEM100 immunoreactivity colocalized with labeling for the pan-neuronal marker protein gene product 9.5 (PGP9.5) but not with the glial marker S100ß or Kit, a marker of interstitial cells of Cajal. The signaling molecule, bone morphogenetic protein (BMP) 4, was also expressed in enteric neurons of the human colon and co-localized with TMEM100. TMEM100 is also expressed in neuronal cell bodies and fibers in the mouse brain and dorsal root ganglia. We conclude that TMEM100 is a novel, membrane-associated marker for enteric nerves and is as effective as PGP9.5 for identifying neuronal structures in the gastrointestinal tract. The expression of TMEM100 in the enteric nervous system may reflect a role in the development and differentiation of cells through a transforming growth factor β, BMP or related signaling pathway.
Collapse
Affiliation(s)
- S T Eisenman
- Enteric Neuroscience Program, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Coron E, Auksorius E, Pieretti A, Mahé MM, Liu L, Steiger C, Bromberg Y, Bouma B, Tearney G, Neunlist M, Goldstein AM. Full-field optical coherence microscopy is a novel technique for imaging enteric ganglia in the gastrointestinal tract. Neurogastroenterol Motil 2012; 24:e611-21. [PMID: 23106847 PMCID: PMC3866795 DOI: 10.1111/nmo.12035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Noninvasive methods are needed to improve the diagnosis of enteric neuropathies. Full-field optical coherence microscopy (FFOCM) is a novel optical microscopy modality that can acquire 1 μm resolution images of tissue. The objective of this research was to demonstrate FFOCM imaging for the characterization of the enteric nervous system (ENS). METHODS Normal mice and EdnrB(-/-) mice, a model of Hirschsprung's disease (HD), were imaged in three-dimensions ex vivo using FFOCM through the entire thickness and length of the gut. Quantitative analysis of myenteric ganglia was performed on FFOCM images obtained from whole-mount tissues and compared with immunohistochemistry imaged by confocal microscopy. KEY RESULTS Full-field optical coherence microscopy enabled visualization of the full thickness gut wall from serosa to mucosa. Images of the myenteric plexus were successfully acquired from the stomach, duodenum, colon, and rectum. Quantification of ganglionic neuronal counts on FFOCM images revealed strong interobserver agreement and identical values to those obtained by immunofluorescence microscopy. In EdnrB(-/-) mice, FFOCM analysis revealed a significant decrease in ganglia density along the colorectum and a significantly lower density of ganglia in all colorectal segments compared with normal mice. CONCLUSIONS & INFERENCES Full-field optical coherence microscopy enables optical microscopic imaging of the ENS within the bowel wall along the entire intestine. FFOCM is able to differentiate ganglionic from aganglionic colon in a mouse model of HD, and can provide quantitative assessment of ganglionic density. With further refinements that enable bowel wall imaging in vivo, this technology has the potential to revolutionize the characterization of the ENS and the diagnosis of enteric neuropathies.
Collapse
Affiliation(s)
- E Coron
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
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: 151] [Impact Index Per Article: 11.6] [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
|
41
|
Powley TL, Gilbert JM, Baronowsky EA, Billingsley CN, Martin FN, Phillips RJ. Vagal sensory innervation of the gastric sling muscle and antral wall: implications for gastro-esophageal reflux disease? Neurogastroenterol Motil 2012; 24:e526-37. [PMID: 22925069 PMCID: PMC3776304 DOI: 10.1111/nmo.12003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The gastric sling muscle has not been investigated for possible sensory innervation, in spite of the key roles the structure plays in lower esophageal sphincter (LES) function and gastric physiology. Thus, the present experiment used tracing techniques to label vagal afferents and survey their projections in the lesser curvature. METHODS Sprague-Dawley rats received injections of dextran biotin into the nodose ganglia. Fourteen days postinjection, animals were euthanized and their stomachs were processed to visualize the vagal afferent innervation. In different cases, neurons, muscle cells, or interstitial cells of Cajal (ICC) were counterstained. KEY RESULTS The sling muscle is innervated throughout its length by vagal afferent intramuscular arrays (IMAs) associated with ICC. In addition, the distal antral attachment site of the sling muscle is innervated by a novel vagal afferent terminal specialization, an antral web ending. The muscle wall of the distal antrum is also innervated by conventional IMAs and intraganglionic laminar endings, the two types of mechanoreceptors found throughout stomach smooth muscle. CONCLUSIONS & INFERENCES The innervation of sling muscle by IMAs, putative stretch receptors, suggests that sling sensory feedback may generate vago-vagal or other reflexes with vagal afferent limbs. The restricted distribution of afferent web endings near the antral attachments of sling fibers suggests the possibility of specialized mechanoreceptor functions linking antral and pyloric activity to the operation of the LES. Dysfunctional sling afferents could generate LES motor disturbances, or normative compensatory sensory feedback from the muscle could compromise therapies targeting only effectors.
Collapse
Affiliation(s)
- Terry L. Powley
- Corresponding author: Terry L. Powley, Purdue University, 703 Third Street, West Lafayette, IN 47907-2091, , Phone: 765-494-6269, Fax: 765-496-1264
| | | | | | | | | | | |
Collapse
|
42
|
Martin GR, Alvarez AL, Bashashati M, Keenan CM, Jirik FR, Sharkey KA. Endogenous cellular prion protein regulates contractility of the mouse ileum. Neurogastroenterol Motil 2012; 24:e412-24. [PMID: 22762267 DOI: 10.1111/j.1365-2982.2012.01970.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Cellular prion protein (PrP(C) ) is expressed in the enteric nervous system (ENS), however, its physiological role has not been identified. Studies suggest that PrP(C) can function as a metal-binding protein, as absence of the protein has been linked to altered copper metabolism and atypical synaptic activity. Because copper is known to modulate smooth muscle relaxation, we tested the hypothesis that PrP(C) deficiency would alter intestinal contractility. METHODS We examined electrically evoked ileal contractility in Prnp(-/-) or wild type littermate mice and the effects of copper or copper chelation. PrP(C) expression was studied in whole mount ileal preparations of mice and guinea pigs by immunohistochemistry. KEY RESULTS Relative to wild type mice, ileal tissues of Prnp(-/-) mice exhibited reduced electrical field stimulation (EFS)-evoked contractility. Furthermore, EFS-induced relaxation, as a percentage of that induced by a nitric oxide donor, was enhanced. Addition of a copper donor to the organ bath increased, whereas the addition of a copper chelator inhibited, nitric oxide donor-induced ileal relaxation in Prnp(-/-) mice. PrP(C) was expressed on nerve fibers or terminals, and some cell bodies in the myenteric and submucosal plexuses of wild type mice. PrP(C) colocalized with a neuron-specific ectonucleotidase, nucleoside triphosphate diphosphohydrolase 3 (NTPDase3), but to only a limited extent with GFAP, a marker of enteric glia. Guinea pigs expressed PrP(C) in nerve fibers or terminals and enteric glia in the myenteric and submucosal plexuses. CONCLUSIONS & INFERENCES Our findings suggest that PrP(C) , which is abundant in the ENS, has a role in the regulation of ileal contractility.
Collapse
Affiliation(s)
- G R Martin
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| | | | | | | | | | | |
Collapse
|
43
|
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: 1.5] [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
|
44
|
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: 83] [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
|
45
|
Colucci M, Cervio M, Faniglione M, De Angelis S, Pajoro M, Levandis G, Tassorelli C, Blandini F, Feletti F, De Giorgio R, Dellabianca A, Tonini S, Tonini M. Intestinal dysmotility and enteric neurochemical changes in a Parkinson's disease rat model. Auton Neurosci 2012; 169:77-86. [PMID: 22608184 DOI: 10.1016/j.autneu.2012.04.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 04/20/2012] [Accepted: 04/24/2012] [Indexed: 01/28/2023]
Abstract
Gastrointestinal disorders, constipation in particular, are the most common non-motor dysfunctions affecting Parkinson's disease (PD) patients. We have previously reported that rats bearing unilateral nigrostriatal lesion caused by 6-hydroxydopamine (6-OHDA) stereotaxic injection develop severe constipation together with a region-specific decrease of neuronal nitric oxide synthase (nNOS) in enteric neurons of the lower intestinal tract. Here, we extend these observations on other enteric neuronal subpopulations, investigating also the propulsive activity of isolated colonic specimens. Four weeks post 6-OHDA injection, lesioned rats showed a significant increase of vasoactive intestinal polypeptide (VIP) concomitant with the reduced expression of nNOS in the myenteric plexus of distal ileum and proximal colon; in particular VIP increased in a subpopulation of neurons actively expressing nNOS. On the other hand, choline acetyltransferase (ChAT) was not modified in any of the intestinal segments analyzed. Interestingly, we found a reduced expression of dopamine receptor type 2 (D2R) in proximal (-66.8%) and distal (-54.5%) colon, together with reduced peristalsis efficiency (decrease in intraluminal pressure and frequency of peristaltic events) in the 6-OHDA-lesioned rats. The selective depletion of dopaminergic nigrostriatal neurons is associated with changes in the expression of enteric inhibitory neurotransmitters, as well as of the D2R in intestinal specific regions. Moreover, 6-OHDA-lesioned rats demonstrated altered colon propulsive activity referable to the D2R decrease. Our findings unveil subtle mechanisms underlying the enteric neurochemical plasticity events evoked by disruption of the normal brain-gut cross-talk, giving a peculiar point of view on the pathophysiology of the severe constipation that frequently affects PD patients.
Collapse
Affiliation(s)
- Mario Colucci
- Department of Legal Medicine, Forensic Sciences and Pharmaco-Toxicology, University of Pavia, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
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
|
47
|
Phillips RJ, Powley TL. Macrophages associated with the intrinsic and extrinsic autonomic innervation of the rat gastrointestinal tract. Auton Neurosci 2012; 169:12-27. [PMID: 22436622 DOI: 10.1016/j.autneu.2012.02.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Revised: 02/22/2012] [Accepted: 02/23/2012] [Indexed: 12/28/2022]
Abstract
Interactions between macrophages and the autonomic innervation of gastrointestinal (GI) tract smooth muscle have received little experimental attention. To better understand this relationship, immunohistochemistry was performed on GI whole mounts from rats at three ages. The phenotypes, morphologies, and distributions of gut macrophages are consistent with the cells performing extensive housekeeping functions in the smooth muscle layers. Specifically, a dense population of macrophages was located throughout the muscle wall where they were distributed among the muscle fibers and along the vasculature. Macrophages were also associated with ganglia and connectives of the myenteric plexus and with the sympathetic innervation. Additionally, these cells were in tight registration with the dendrites and axons of the myenteric neurons as well as the varicosities along the length of the sympathetic axons, suggestive of a contribution by the macrophages to the homeostasis of both synapses and contacts between the various elements of the enteric circuitry. Similarly, macrophages were involved in the presumed elimination of neuropathies as indicated by their association with dystrophic neurons and neurites which are located throughout the myenteric plexus and smooth muscle wall of aged rats. Importantly, the patterns of macrophage-neuron interactions in the gut paralleled the much more extensively characterized interactions of macrophages (i.e., microglia) and neurons in the CNS. The present observations in the PNS as well as extrapolations from homologous microglia in the CNS suggest that GI macrophages play significant roles in maintaining the nervous system of the gut in the face of wear and tear, disease, and aging.
Collapse
Affiliation(s)
- Robert J Phillips
- Department of Psychological Sciences, Purdue University, West Lafayette, Indiana 47907-2081, USA
| | | |
Collapse
|
48
|
Chen JJ, Gershon AA, Li Z, Cowles RA, Gershon MD. Varicella zoster virus (VZV) infects and establishes latency in enteric neurons. J Neurovirol 2011; 17:578-89. [PMID: 22190254 PMCID: PMC3324263 DOI: 10.1007/s13365-011-0070-1] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Revised: 12/01/2011] [Accepted: 12/05/2011] [Indexed: 01/27/2023]
Abstract
Case reports have linked varicella-zoster virus (VZV) to gastrointestinal disorders, including severe abdominal pain preceding fatal varicella and acute colonic pseudoobstruction (Ogilvie's syndrome). Because we had previously detected DNA and transcripts encoding latency-associated VZV gene products in the human gut, we sought to determine whether latent VZV is present in the human enteric nervous system (ENS) and, if so, to identify the cells in which it is located and its route to the bowel. Neither DNA, nor transcripts encoding VZV gene products, could be detected in resected gut from any of seven control children (<1 year old) who had not received the varicella vaccine or experienced varicella; however, VZV DNA and transcripts were each found to be present in resected bowel from 6/6 of children with a past history of varicella and in that of 6/7 of children who received the varicella vaccine. Both wild-type (WT) and vaccine-type (vOka) VZV thus establish latent infection in human gut. To determine routes by which VZV might gain access to the bowel, we injected guinea pigs with human or guinea pig lymphocytes expressing green fluorescent protein (GFP) under the control of the VZV ORF66 gene (VZV(OKA66.GFP)). GFP-expressing enteric neurons were found throughout the bowel within 2 days and continued to be present for greater than 6 weeks. DNA encoding VZV gene products also appeared in enteric and dorsal root ganglion (DRG) neurons following intradermal administration of WT-VZV and in enteric neurons after intradermal injection of VZV(OKA66.GFP); moreover, a small number of guinea pig DRG neurons were found to project both to the skin and the intraperitoneal viscera. Viremia, in which lymphocytes carry VZV, or axonal transport from DRG neurons infected through their epidermal projections are thus each potential routes that enable VZV to gain access to the ENS.
Collapse
Affiliation(s)
- Jason J Chen
- Departments of Pathology and Cell Biology, Columbia University, College of P&S, New York, NY, USA
| | | | | | | | | |
Collapse
|
49
|
Asuzu DT, Hayashi Y, Izbeki F, Popko LN, Young DL, Bardsley MR, Lorincz A, Kuro-o M, Linden DR, Farrugia G, Ordog T. Generalized neuromuscular hypoplasia, reduced smooth muscle myosin and altered gut motility in the klotho model of premature aging. Neurogastroenterol Motil 2011; 23:e309-23. [PMID: 21605285 PMCID: PMC3149585 DOI: 10.1111/j.1365-2982.2011.01730.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Gastrointestinal symptoms, particularly constipation, increase with aging, but their underlying mechanisms are poorly understood due to lack of experimental models. Previously we established the progeric klotho mouse as a model of aging-associated anorexia and gastric dysmotility. We also detected reduced fecal output in these animals; therefore, the aim of this study was to investigate in vivo function and cellular make-up of the small intestinal and colonic neuromuscular apparatus. METHODS Klotho expression was studied by RT-PCR and immunohistochemistry. Motility was assessed by dye transit and bead expulsion. Smooth muscle and neuron-specific gene expression was studied by Western immunoblotting. Interstitial cells of Cajal (ICC) and precursors were analyzed by flow cytometry, confocal microscopy, and three-dimensional reconstruction. HuC/D(+) myenteric neurons were enumerated by fluorescent microscopy. KEY RESULTS Klotho protein was detected in neurons, smooth muscle cells, and some ICC classes. Small intestinal transit was slower but whole-gut transit of klotho mice was accelerated due to faster colonic transit and shorter intestinal lengths, apparent only after weaning. Fecal water content remained normal despite reduced output. Smooth muscle myosin expression was reduced. ICC, ICC precursors, as well as nitrergic and cholinergic neurons maintained their normal proportions in the shorter intestines. CONCLUSIONS & INFERENCES Progeric klotho mice express less contractile proteins and develop generalized intestinal neuromuscular hypoplasia mainly arising from stunted postweaning growth. As reduced fecal output in these mice occurs in the presence of accelerated colonic and whole-gut transit, it likely reflects reduced food intake rather than intestinal dysmotility.
Collapse
Affiliation(s)
- David T. Asuzu
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Yujiro Hayashi
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Ferenc Izbeki
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Laura N. Popko
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - David L. Young
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Michael R. Bardsley
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Andrea Lorincz
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| | - Makoto Kuro-o
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - David R. Linden
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Gianrico Farrugia
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamas Ordog
- Enteric Neuroscience Program and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA,Gastroenterology Research Unit, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
50
|
Kramer K, da Silveira ABM, Jabari S, Kressel M, Raab M, Brehmer A. Quantitative evaluation of neurons in the mucosal plexus of adult human intestines. Histochem Cell Biol 2011; 136:1-9. [DOI: 10.1007/s00418-011-0807-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2011] [Indexed: 10/18/2022]
|