1
|
Voss U. Enteric neuroprotection-A matter of balancing redox potentials, limiting inflammation, and boosting resilience. Neurogastroenterol Motil 2024:e14871. [PMID: 39038122 DOI: 10.1111/nmo.14871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/15/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
The enteric nervous system (ENS) orchestrates intricate and autonomous functions throughout the gastrointestinal (GI) tract. Disruptions in ENS function are associated GI disorders. This mini review focuses on the past decade's research, utilizing rodent models, with an emphasis on protecting enteric neurons from loss. The review specifically looks at efforts to reduce oxidative stress, limit inflammation, and enhance neuronal resilience. Protective interventions including administration of antioxidants and compounds targeting cellular redox buffer systems, are evaluated for their effectiveness in preventing loss of enteric neurons in the ischemia-reperfusion model and streptozotocin-induced diabetes model. Interventions such as engrafting mesenchymal stem cells and targeting inflammatory signaling pathways in enteric neurons and glial cells are evaluated in inflammatory bowel disease models including the Winnie mouse, DSS-, and DNBS/TNBS-induced colitis models. The review also touches upon neuronal resilience, particularly in the context of Parkinson's disease models. Including estrogen's neuroprotective role, and the influence of metal ions on enteric neuronal protection. Understanding the dynamic interplay within the ENS and its role in disease pathogenesis holds promise for developing targeted therapies to effectively manage and treat various GI ailments.
Collapse
Affiliation(s)
- Ulrikke Voss
- Department of Clinical Sciences, Lund, Lund University, Lund, Sweden
| |
Collapse
|
2
|
Gałęcka I, Całka J. Oral Exposure to Microplastics Affects the Neurochemical Plasticity of Reactive Neurons in the Porcine Jejunum. Nutrients 2024; 16:2268. [PMID: 39064711 PMCID: PMC11280339 DOI: 10.3390/nu16142268] [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: 06/04/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Plastics are present in almost every aspect of our lives. Polyethylene terephthalate (PET) is commonly used in the food industry. Microparticles can contaminate food and drinks, posing a threat to consumers. The presented study aims to determine the effect of microparticles of PET on the population of neurons positive for selected neurotransmitters in the enteric nervous system of the jejunum and histological structure. An amount of 15 pigs were divided into three groups (control, receiving 0.1 g, and 1 g/day/animal orally). After 28 days, fragments of the jejunum were collected for immunofluorescence and histological examination. The obtained results show that histological changes (injury of the apical parts of the villi, accumulations of cellular debris and mucus, eosinophil infiltration, and hyperaemia) were more pronounced in pigs receiving a higher dose of microparticles. The effect on neuronal nitric oxide synthase-, and substance P-positive neurons, depends on the examined plexus and the dose of microparticles. An increase in the percentage of galanin-positive neurons and a decrease in cocaine and amphetamine-regulated transcript-, vesicular acetylcholine transporter-, and vasoactive intestinal peptide-positive neurons do not show such relationships. The present study shows that microparticles can potentially have neurotoxic and pro-inflammatory effects, but there is a need for further research to determine the mechanism of this process and possible further effects.
Collapse
Affiliation(s)
- Ismena Gałęcka
- Department of Epizootiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13, 10-719 Olsztyn, Poland
| |
Collapse
|
3
|
Pasricha TS, Guerrero-Lopez IL, Kuo B. Management of Gastrointestinal Symptoms in Parkinson's Disease: A Comprehensive Review of Clinical Presentation, Workup, and Treatment. J Clin Gastroenterol 2024; 58:211-220. [PMID: 38260966 PMCID: PMC10855995 DOI: 10.1097/mcg.0000000000001961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024]
Abstract
Gastrointestinal symptoms in Parkinson's disease (PD) are among the most prevalent and debilitating of complications and present unique diagnostic and management challenges. Patients with PD commonly experience dysphagia, nausea, bloating, and constipation related to pathologic involvement of the enteric nervous system. In turn, gastrointestinal complications may impact motor fluctuations and the efficacy of levodopa therapy. This review will explore the common gastrointestinal manifestations of PD with an emphasis on clinical presentation, workup, and treatment strategies.
Collapse
Affiliation(s)
- Trisha S. Pasricha
- Division of Gastroenterology, Massachusetts General Hospital
- Harvard Medical School, Boston, MA
| | | | - Braden Kuo
- Division of Gastroenterology, Massachusetts General Hospital
- Harvard Medical School, Boston, MA
| |
Collapse
|
4
|
Zhang X, Tang B, Guo J. Parkinson's disease and gut microbiota: from clinical to mechanistic and therapeutic studies. Transl Neurodegener 2023; 12:59. [PMID: 38098067 PMCID: PMC10722742 DOI: 10.1186/s40035-023-00392-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases. The typical symptomatology of PD includes motor symptoms; however, a range of nonmotor symptoms, such as intestinal issues, usually occur before the motor symptoms. Various microorganisms inhabiting the gastrointestinal tract can profoundly influence the physiopathology of the central nervous system through neurological, endocrine, and immune system pathways involved in the microbiota-gut-brain axis. In addition, extensive evidence suggests that the gut microbiota is strongly associated with PD. This review summarizes the latest findings on microbial changes in PD and their clinical relevance, describes the underlying mechanisms through which intestinal bacteria may mediate PD, and discusses the correlations between gut microbes and anti-PD drugs. In addition, this review outlines the status of research on microbial therapies for PD and the future directions of PD-gut microbiota research.
Collapse
Affiliation(s)
- Xuxiang Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Beisha Tang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jifeng Guo
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, 410008, China.
- Hunan International Scientific and Technological Cooperation Base of Neurodegenerative and Neurogenetic Diseases, Changsha, 410008, China.
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, 410008, China.
- Engineering Research Center of Hunan Province in Cognitive Impairment Disorders, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
5
|
Xing T, Nanni G, Burkholder CR, Browning KN, Travagli RA. The substantia nigra modulates proximal colon tone and motility in a vagally-dependent manner in the rat. J Physiol 2023; 601:4751-4766. [PMID: 37772988 PMCID: PMC10873099 DOI: 10.1113/jp284238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 09/08/2023] [Indexed: 09/30/2023] Open
Abstract
A monosynaptic pathway connects the substantia nigra pars compacta (SNpc) to neurons of the dorsal motor nucleus of the vagus (DMV). This monosynaptic pathway modulates the vagal control of gastric motility. It is not known, however, whether this nigro-vagal pathway also modulates the tone and motility of the proximal colon. In rats, microinjection of retrograde tracers in the proximal colon and of anterograde tracers in SNpc showed that bilaterally labelled colonic-projecting neurons in the DMV received inputs from SNpc neurons. Microinjections of the ionotropic glutamate receptor agonist, NMDA, in the SNpc increased proximal colonic motility and tone, as measured via a strain gauge aligned with the colonic circular smooth muscle; the motility increase was inhibited by acute subdiaphragmatic vagotomy. Upon transfection of SNpc with pAAV-hSyn-hM3D(Gq)-mCherry, chemogenetic activation of nigro-vagal nerve terminals by brainstem application of clozapine-N-oxide increased the firing rate of DMV neurons and proximal colon motility; both responses were abolished by brainstem pretreatment with the dopaminergic D1-like antagonist SCH23390. Chemogenetic inhibition of nigro-vagal nerve terminals following SNpc transfection with pAAV-hSyn-hM4D(Gi)-mCherry decreased the firing rate of DMV neurons and inhibited proximal colon motility. These data suggest that a nigro-vagal pathway modulates activity of the proximal colon motility tonically via a discrete dopaminergic synapse in a manner dependent on vagal efferent nerve activity. Impairment of this nigro-vagal pathway may contribute to the severely reduced colonic transit and prominent constipation observed in both patients and animal models of parkinsonism. KEY POINTS: Substantia nigra pars compacta (SNpc) neurons are connected to the dorsal motor nucleus of the vagus (DMV) neurons via a presumed direct pathway. Brainstem neurons in the lateral DMV innervate the proximal colon. Colonic-projecting DMV neurons receive inputs from neurons of the SNpc. The nigro-vagal pathway modulates tone and motility of the proximal colon via D1-like receptors in the DMV. The present study provides the mechanistic basis for explaining how SNpc alterations may lead to a high rate of constipation in patients with Parkinson's Disease.
Collapse
Affiliation(s)
| | | | | | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA and Neurobiology Research, Newport, NC
| | | |
Collapse
|
6
|
Abo-Shaban T, Lee CYQ, Hosie S, Balasuriya GK, Mohsenipour M, Johnston LA, Hill-Yardin EL. GutMap: A New Interface for Analysing Regional Motility Patterns in ex vivo Mouse Gastrointestinal Preparations. Bio Protoc 2023; 13:e4831. [PMID: 37817909 PMCID: PMC10560633 DOI: 10.21769/bioprotoc.4831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 10/12/2023] Open
Abstract
Different regions of the gastrointestinal tract have specific functions and thus distinct motility patterns. Motility is primarily regulated by the enteric nervous system (ENS), an intrinsic network of neurons located within the gut wall. Under physiological conditions, the ENS is influenced by the central nervous system (CNS). However, by using ex vivo organ bath experiments, ENS regulation of gut motility can also be studied in the absence of CNS influences. The current technique enables the characterisation of small intestinal, caecal, and colonic motility patterns using an ex vivo organ bath and video imaging protocol. This approach is combined with the novel edge detection script GutMap, available in MATLAB, that functions across Windows and Mac platforms. Dissected intestinal segments are cannulated in an organ bath containing physiological saline with a camera mounted overhead. Video recordings of gut contractions are then converted to spatiotemporal heatmaps and analysed using the GutMap software interface. Using data analysed from the heatmaps, parameters of contractile patterns (including contraction propagation frequency and velocity as well as gut diameter) at baseline and in the presence of drugs/treatments/genetic mutations can be compared. Here, we studied motility patterns of female mice at baseline and in the presence of a nitric oxide synthase inhibitor (Nω-Nitro-L-arginine; NOLA) (nitric oxide being the main inhibitory neurotransmitter of gut motility) to showcase the application of GutMap. This technique is suitable for application to a broad range of animal models of clinical disorders to understand underlying biological pathways contributing to gastrointestinal dysfunction. Key features • Enhanced video imaging analysis of gut contractility in rodents using a novel software interface. • New edge detection algorithm to accurately contour curvatures of the gastrointestinal tract. • Allows for output of high-resolution spatiotemporal heatmaps across Windows and Mac platforms. • Edge detection and analysis method makes motility measurements accessible in different gut regions including the caecum and stomach.
Collapse
Affiliation(s)
- Tanya Abo-Shaban
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC, Australia
| | - Chalystha Y. Q. Lee
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC, Australia
| | - Suzanne Hosie
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC, Australia
| | - Gayathri K. Balasuriya
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC, Australia
- Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Mitra Mohsenipour
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC, Australia
| | - Leigh A. Johnston
- Department of Biomedical Engineering and Melbourne Brain Centre Imaging Unit, The University of Melbourne, Melbourne, VIC, Australia
| | - Elisa L. Hill-Yardin
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, VIC, Australia
| |
Collapse
|
7
|
Zhou X, Mao B, Tang X, Zhang Q, Zhao J, Zhang H, Cui S. Exploring the Dose–Effect Relationship of Bifidobacterium longum in Relieving Loperamide Hydrochloride-Induced Constipation in Rats through Colon-Released Capsules. Int J Mol Sci 2023; 24:ijms24076585. [PMID: 37047557 PMCID: PMC10095166 DOI: 10.3390/ijms24076585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/19/2023] [Accepted: 03/22/2023] [Indexed: 04/05/2023] Open
Abstract
Constipation is a common disease affecting humans. Bifidobacterium longum is reportedly effective in relieving constipation. Current studies generally focus on the dose–response relationship of oral doses; however, the dose–effect relationship of B. longum in the colon, which is the primary site where B. longum exerts constipation-relieving effects, to treat constipation has not been studied. Herein, three strains of B. longum (FGSZY6M4, FJSWXJ10M2, and FSDJN6M3) were packaged in colon-released capsules to explore the dose–effect relationship in the colon. For each strain, three groups of capsules (104, 106, and 108 CFU/capsule, respectively) and one group of free probiotics (108 CFU/mL) were used to explore the colonic dose effect of B. longum. The results showed that the three strains of B. longum improved fecal water content and promoted intestinal motility by regulating gastrointestinal peptide (MTL, GAS, and VIP), aquaporin-3, and 5-hydroxytryptamine levels while promoting gastrointestinal motility and relieving constipation by regulating the intestinal flora composition of constipated rats and changing their metabolite content (short-chain fatty acids). Among the three free bacterial solution groups (108 CFU/mL), FGSZY6M4 was the most effective in relieving constipation caused by loperamide hydrochloride in rats. The optimal effective dose of each strain was 6M4 (104 CFU/day), 10M2 (106 CFU/day), and S3 (108 CFU/day) of the colon-released capsules. Therefore, for some effective strains, the dose of oral probiotics can be reduced by colon-released capsules, and constipation can be relieved without administering a great number of bacterial solutions. Therefore, investigating the most effective dose of B. longum at the colon site can help to improve the efficiency of relieving constipation.
Collapse
Affiliation(s)
- Xin Zhou
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xin Tang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
8
|
Wang Y, Jiang H, Wang L, Gan H, Xiao X, Huang L, Li W, Li Z. Luteolin ameliorates loperamide-induced functional constipation in mice. Braz J Med Biol Res 2023; 56:e12466. [PMID: 36722660 PMCID: PMC9883005 DOI: 10.1590/1414-431x2023e12466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/29/2022] [Indexed: 02/02/2023] Open
Abstract
Functional constipation (FC) is one of the most common gastrointestinal disorders characterized by hard stools and infrequent bowel movements, which is associated with dysfunction of the enteric nervous system and intestinal motility. Luteolin, a naturally occurring flavone, was reported to possess potential pharmacological activities on intestinal inflammation and nerve injury. This study aimed to explore the role of luteolin and its functional mechanism in loperamide-induced FC mice. Our results showed that luteolin treatment reversed the reduction in defecation frequency, fecal water content, and intestinal transit ratio, and the elevation in transit time of FC models. Consistently, luteolin increased the thickness of the muscular layer and lessened colonic histopathological injury induced by loperamide. Furthermore, we revealed that luteolin treatment increased the expression of neuronal protein HuC/D and the levels of intestinal motility-related biomarkers, including substance P (SP), vasoactive intestinal polypeptide (VIP), and acetylcholine (ACh), as well as interstitial cells of Cajal (ICC) biomarker KIT proto-oncogene, receptor tyrosine kinase (C-Kit), and anoctamin-1 (ANO1), implying that luteolin mediated enhancement of colonic function and contributed to the anti-intestinal dysmotility against loperamide-induced FC. Additionally, luteolin decreased the upregulation of aquaporin (AQP)-3, AQP-4, and AQP-8 in the colon of FC mice. In summary, our data showed that luteolin might be an attractive option for developing FC-relieving medications.
Collapse
Affiliation(s)
- Yujin Wang
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Hua Jiang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Lijun Wang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Huiping Gan
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Xinchun Xiao
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Liangwu Huang
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Wenxin Li
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Zongrun Li
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| |
Collapse
|
9
|
Yu L, Yang X, Guan W, Zhang D, Ren S, Xing Y, An D, Zhang J, Zhu Y, Zhu A. Analysis of Key Genes for Slow Transit Constipation Based on RNA Sequencing. Int J Gen Med 2022; 15:7569-7579. [PMID: 36199586 PMCID: PMC9528044 DOI: 10.2147/ijgm.s380208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/16/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose This study aims to identify key genes in slow transit constipation (STC). We also sought to explore the potential link between STC and colorectal cancer. Patients and Methods mRNA expression profiles were obtained by RNA sequencing, and differentially expressed genes were identified. Functional enrichment analysis and a protein–protein interaction (PPI) network was explored, and differentially expressed genes common to STC and colorectal cancer were examined. Analysis of the effect of constipation and colorectal cancer common genes on the overall survival of colorectal cancer patients based on GEPIA database. Results Functional enrichment showed that significantly different genes are related to lymphocyte chemotaxis, positive regulation of inflammatory response, cellular response to tumor necrosis factor, extracellular region, extracellular space and chemokine activity. The hub gene for STC was found in the PPI network. In addition, AQP8 and CFD were common differential genes for STC and colorectal cancer. AQP8 affects overall survival in patients with colorectal cancer. Conclusion Our findings will contribute to understanding the pathology of STC at the molecular level, with the first discovery that AQP8 may be a hub gene in the transition from STC to colorectal cancer.
Collapse
Affiliation(s)
- Linfeng Yu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xiuding Yang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Wenlong Guan
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Dongxu Zhang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Shuo Ren
- Department of Gastrointestinal Surgery, Sichuan Cancer Hospital, Chengdu, People’s Republic of China
| | - Yanwei Xing
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Da An
- Department of Anesthesiology, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jian Zhang
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yuekun Zhu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Anlong Zhu
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
- Correspondence: Anlong Zhu, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China, Tel/Fax +86 13504848555, Email
| |
Collapse
|
10
|
Poirier AA, Côté M, Jarras H, Litim N, Lamontagne-Proulx J, Al-Sweidi S, Morissette M, Lachhab A, Pelletier M, Di Paolo T, Soulet D. Peripheral Neuroprotective and Immunomodulatory Effects of 5α-Reductase Inhibitors in Parkinson's Disease Models. Front Pharmacol 2022; 13:898067. [PMID: 35935876 PMCID: PMC9355275 DOI: 10.3389/fphar.2022.898067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/27/2022] [Indexed: 11/21/2022] Open
Abstract
Gastrointestinal disorders in Parkinson's disease (PD) have been associated with neuronal alteration in the plexus of the gut. We previously demonstrated the immunomodulatory effect of female hormones to treat enteric neurodegeneration in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. This study made the hypothesis of obtaining similar neuroprotection as with hormone treatments by affecting steroidogenesis with two 5α-reductase inhibitors, finasteride and dutasteride. These drugs are approved to treat benign prostatic hyperplasia and alopecia and display mitochondrial effects. In MPTP-treated mice, the dopaminergic and vasoactive intestinal peptide (VIP) neurons alteration was prevented by finasteride and dutasteride, while the increase in proinflammatory macrophages density was inhibited by dutasteride treatment but not finasteride. NF-κB response, oxidative stress, and nitric oxide and proinflammatory cytokines production in vitro were only prevented by dutasteride. In addition, mitochondrial production of free radicals, membrane depolarization, decreased basal respiration, and ATP production were inhibited by dutasteride, while finasteride had no effect. In conclusion, the present results indicate that dutasteride treatment prevents enteric neuronal damages in the MPTP mouse model, at least in part through anti-inflammatory and mitochondrial effects. This suggests that drug repurposing of dutasteride might be a promising avenue to treat enteric neuroinflammation in early PD.
Collapse
Affiliation(s)
- Andrée-Anne Poirier
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec City, QC, Canada
| | - Mélissa Côté
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Hend Jarras
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec City, QC, Canada
| | - Nadhir Litim
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec City, QC, Canada
| | - Jérôme Lamontagne-Proulx
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec City, QC, Canada
| | - Sara Al-Sweidi
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec City, QC, Canada
| | - Marc Morissette
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Asmaa Lachhab
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Médecine, Université Laval, Québec City, QC, Canada
| | - Martin Pelletier
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Médecine, Université Laval, Québec City, QC, Canada
| | - Thérèse Di Paolo
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec City, QC, Canada
| | - Denis Soulet
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec City, QC, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Québec City, QC, Canada
| |
Collapse
|
11
|
Xu J, Wang L, Chen X, Le W. New Understanding on the Pathophysiology and Treatment of Constipation in Parkinson’s Disease. Front Aging Neurosci 2022; 14:917499. [PMID: 35813960 PMCID: PMC9257174 DOI: 10.3389/fnagi.2022.917499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Constipation, one of the most common prodromal non-motor symptoms of Parkinson’s disease (PD), usually occurs several years earlier than the onset of motor symptoms. Previous studies have shown that constipation occurrence increases as the disease progresses. However, the mechanism underlying this pathologic disorder is not clear yet. Moreover, chronic constipation causes slowness in gastric emptying and, therefore, may lead to a delay in the absorption of medications for PD, including levodopa and dopamine agonists. Accordingly, it is necessary to understand how the pathophysiological factors contribute to constipation during PD as well as pursue precise and effective treatment strategies. In this review, we encapsulate the molecular mechanism of constipation underlying PD and update the progress in the treatments of PD-associated constipation.
Collapse
Affiliation(s)
- Jianli Xu
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Lei Wang
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
| | - Xi Chen
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- Xi Chen Weidong Le
| | - Weidong Le
- Institute of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, China
- Xi Chen Weidong Le
| |
Collapse
|
12
|
Characterization of Neurochemical Signature Alterations in the Enteric Nervous System in Autoimmune Encephalomyelitis. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
To date, it has remained unclear whether gastrointestinal symptoms, which are frequently observed in patients with multiple sclerosis (MS), are accompanied by pathology of the enteric nervous system (ENS). Here, the neurotransmitter signature of ENS neurons and morphological alterations of interstitial cells of Cajal (ICCs) were studied in patients with MS and mice with experimental autoimmune encephalomyelitis (EAE), which is an animal model of MS. Immunohistochemical analysis was performed on colonic whole mounts from mice with EAE and on paraffin-embedded sections of intestinal tissue from patients with MS. Antibodies against neurotransmitters or their enzymes (including vasoactive intestinal peptide (VIP), neuronal nitric oxide synthase (nNOS), and choline acetyltransferase (ChAT)) were used in conjunction with pan-neuronal markers. In addition, the presence of anoctamin 1 (ANO1)-expressing ICCs was studied. ENS changes were observed in the myenteric plexus, but they were absent in the submucosal plexus of both EAE mice and patients with MS. There was a significant decrease in the percentage of ChAT-positive neurons in EAE mice as opposed to a trend toward an increase in patients with MS. Moreover, while ANO1 expression was decreased in EAE mice, patients with MS displayed a significant increase. Although additional studies are necessary to accomplish an in-depth characterization of ENS alterations in MS, our results imply that such alterations exist and may reveal novel insights into the pathophysiology of MS.
Collapse
|
13
|
Han MN, Finkelstein DI, McQuade RM, Diwakarla S. Gastrointestinal Dysfunction in Parkinson’s Disease: Current and Potential Therapeutics. J Pers Med 2022; 12:jpm12020144. [PMID: 35207632 PMCID: PMC8875119 DOI: 10.3390/jpm12020144] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/04/2023] Open
Abstract
Abnormalities in the gastrointestinal (GI) tract of Parkinson’s disease (PD) sufferers were first reported over 200 years ago; however, the extent and role of GI dysfunction in PD disease progression is still unknown. GI dysfunctions, including dysphagia, gastroparesis, and constipation, are amongst the most prevalent non-motor symptoms in PD. These symptoms not only impact patient quality of life, but also complicate disease management. Conventional treatment pathways for GI dysfunctions (i.e., constipation), such as increasing fibre and fluid intake, and the use of over-the-counter laxatives, are generally ineffective in PD patients, and approved compounds such as guanylate cyclase C agonists and selective 5-hyroxytryptamine 4 receptor agonists have demonstrated limited efficacy. Thus, identification of potential targets for novel therapies to alleviate PD-induced GI dysfunctions are essential to improve clinical outcomes and quality of life in people with PD. Unlike the central nervous system (CNS), where PD pathology and the mechanisms involved in CNS damage are relatively well characterised, the effect of PD at the cellular and tissue level in the enteric nervous system (ENS) remains unclear, making it difficult to alleviate or reverse GI symptoms. However, the resurgence of interest in understanding how the GI tract is involved in various disease states, such as PD, has resulted in the identification of novel therapeutic avenues. This review focuses on common PD-related GI symptoms, and summarizes the current treatments available and their limitations. We propose that by targeting the intestinal barrier, ENS, and/or the gut microbiome, may prove successful in alleviating PD-related GI symptoms, and discuss emerging therapies and potential drugs that could be repurposed to target these areas.
Collapse
Affiliation(s)
- Myat Noe Han
- Gut-Axis Injury and Repair Laboratory, Department of Medicine Western Health, University of Melbourne, Melbourne, VIC 3021, Australia; (M.N.H.); (S.D.)
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - David I. Finkelstein
- Parkinson’s Disease Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia;
| | - Rachel M. McQuade
- Gut-Axis Injury and Repair Laboratory, Department of Medicine Western Health, University of Melbourne, Melbourne, VIC 3021, Australia; (M.N.H.); (S.D.)
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
- Correspondence: ; Tel.: +61-3-8395-8114
| | - Shanti Diwakarla
- Gut-Axis Injury and Repair Laboratory, Department of Medicine Western Health, University of Melbourne, Melbourne, VIC 3021, Australia; (M.N.H.); (S.D.)
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC 3010, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| |
Collapse
|
14
|
Zhang XL, Zhang XH, Yu X, Zheng LF, Feng XY, Liu CZ, Quan ZS, Zhang Y, Zhu JX. Enhanced Contractive Tension and Upregulated Muscarinic Receptor 2/3 in Colorectum Contribute to Constipation in 6-Hydroxydopamine-Induced Parkinson's Disease Rats. Front Aging Neurosci 2022; 13:770841. [PMID: 35002677 PMCID: PMC8733788 DOI: 10.3389/fnagi.2021.770841] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 12/02/2021] [Indexed: 11/30/2022] Open
Abstract
Constipation and defecatory dysfunctions are frequent symptoms in patients with Parkinson’s disease (PD). The pathology of Lewy bodies in colonic and rectal cholinergic neurons suggests that cholinergic pathways are involved in colorectal dysmotility in PD. However, the underlying mechanism is unclear. The aim of the present study is to examine the effect of central dopaminergic denervation in rats, induced by injection 6-hydroxydopamine into the bilateral substania nigra (6-OHDA rats), on colorectal contractive activity, content of acetylcholine (ACh), vasoactive intestinal peptide (VIP) and expression of neural nitric oxide synthase (nNOS) and muscarinic receptor (MR). Strain gauge force transducers combined with electrical field stimulation (EFS), gut transit time, immunohistochemistry, ELISA, western blot and ultraperformance liquid chromatography tandem mass spectrometry were used in this study. The 6-OHDA rats exhibited outlet obstruction constipation characterized by prolonged transit time, enhanced contractive tension and fecal retention in colorectum. Pretreatment with tetrodotoxin significantly increased the colorectal motility. EFS-induced cholinergic contractions were diminished in the colorectum. Bethanechol chloride promoted colorectal motility in a dose-dependent manner, and much stronger reactivity of bethanechol chloride was observed in 6-OHDA rats. The ACh, VIP and protein expression of nNOS was decreased, but M2R and M3R were notably upregulated in colorectal muscularis externa. Moreover, the number of cholinergic neurons was reduced in sacral parasympathetic nucleus (SPN) of 6-OHDA rats. In conclusion, central nigrostriatal dopaminergic denervation is associated with decreased cholinergic neurons in SPN, decreased ACh, VIP content, and nNOS expression and upregulated M2R and M3R in colorectum, resulting in colorectal dysmotility, which contributes to outlet obstruction constipation. The study provides new insights into the mechanism of constipation and potential therapeutic targets for constipation in PD patients.
Collapse
Affiliation(s)
- Xiao-Li Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Hui Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.,Artificial Liver Treatment Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiao Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li-Fei Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Yan Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chen-Zhe Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhu-Sheng Quan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yue Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jin-Xia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
O’Day C, Finkelstein DI, Diwakarla S, McQuade RM. A Critical Analysis of Intestinal Enteric Neuron Loss and Constipation in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1841-1861. [PMID: 35848035 PMCID: PMC9535602 DOI: 10.3233/jpd-223262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 06/06/2023]
Abstract
Constipation afflicts many patients with Parkinson's disease (PD) and significantly impacts on patient quality of life. PD-related constipation is caused by intestinal dysfunction, but the etiology of this dysfunction in patients is unknown. One possible cause is neuron loss within the enteric nervous system (ENS) of the intestine. This review aims to 1) Critically evaluate the evidence for and against intestinal enteric neuron loss in PD patients, 2) Justify why PD-related constipation must be objectively measured, 3) Explore the potential link between loss of enteric neurons in the intestine and constipation in PD, 4) Provide potential explanations for disparities in the literature, and 5) Outline data and study design considerations to improve future research. Before the connection between intestinal enteric neuron loss and PD-related constipation can be confidently described, future research must use sufficiently large samples representative of the patient population (majority diagnosed with idiopathic PD for at least 5 years), implement a consistent neuronal quantification method and study design, including standardized patient recruitment criteria, objectively quantify intestinal dysfunctions, publish with a high degree of data transparency and account for potential PD heterogeneity. Further investigation into other potential influencers of PD-related constipation is also required, including changes in the function, connectivity, mitochondria and/or α-synuclein proteins of enteric neurons and their extrinsic innervation. The connection between enteric neuron loss and other PD-related gastrointestinal (GI) issues, including gastroparesis and dysphagia, as well as changes in nutrient absorption and the microbiome, should be explored in future research.
Collapse
Affiliation(s)
- Chelsea O’Day
- Gut-Axis Injury & Repair Laboratory, Department of Medicine - Western Centre for Health Research and Education (WCHRE), The University of Melbourne, Sunshine Hospital, St Albans, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE) Level 3 and 4, Sunshine Hospital, St Albans, VIC, Australia
| | - David Isaac Finkelstein
- Parkinson’s Disease Laboratory, The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| | - Shanti Diwakarla
- Gut-Axis Injury & Repair Laboratory, Department of Medicine - Western Centre for Health Research and Education (WCHRE), The University of Melbourne, Sunshine Hospital, St Albans, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE) Level 3 and 4, Sunshine Hospital, St Albans, VIC, Australia
| | - Rachel Mai McQuade
- Gut-Axis Injury & Repair Laboratory, Department of Medicine - Western Centre for Health Research and Education (WCHRE), The University of Melbourne, Sunshine Hospital, St Albans, VIC, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
- Australian Institute of Musculoskeletal Science (AIMSS), Western Centre for Health Research and Education (WCHRE) Level 3 and 4, Sunshine Hospital, St Albans, VIC, Australia
| |
Collapse
|
16
|
Li Y, Chen Y, Jiang L, Zhang J, Tong X, Chen D, Le W. Intestinal Inflammation and Parkinson's Disease. Aging Dis 2021; 12:2052-2068. [PMID: 34881085 PMCID: PMC8612622 DOI: 10.14336/ad.2021.0418] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/18/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease which significantly influences the life quality of patients. The protein α-synuclein plays an important driving role in PD occurrence and development. Braak's hypothesis suggests that α-synuclein is produced in intestine, and then spreads into the central nervous system through the vagus nerve. The abnormal expression of α-synuclein has been found in inflammatory bowel disease (IBD). Intestinal inflammation and intestinal dysbiosis have been involved in the occurrence and development of PD. The present review aimed to summarize recent advancements in studies focusing on intestinal inflammation and PD, especially the mechanisms through which link intestinal inflammation and PD. The intestinal dysfunctions such as constipation have been introduced as non-motor manifestations of PD. The possible linkages between IBD and PD, including genetic overlaps, inflammatory responses, intestinal permeability, and intestinal dysbiosis, are mainly discussed. Although it is not confirmed whether PD starts from intestine, intestinal dysfunction may affect intestinal microenvironment to influence central nervous system, including the α-synuclein pathologies and systematic inflammation. It is expected to develop some new strategies in the diagnosis and treatment of PD from the aspect of intestine. It may also become an exciting direction to find better ways to regulate the composition of gut microorganism to treat PD.
Collapse
Affiliation(s)
- Yu Li
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Yuanyuan Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Lili Jiang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Jingyu Zhang
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Xuhui Tong
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Dapeng Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China.
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, Liaoning, China
- Institute of Neurology, Sichuan Academy of Medical Science-Sichuan Provincial Hospital, Chengdu, Sichuan, China
| |
Collapse
|
17
|
Derkinderen P, Cossais F, de Guilhem de Lataillade A, Leclair-Visonneau L, Neunlist M, Paillusson S, De Giorgio R. Gastrointestinal mucosal biopsies in Parkinson's disease: beyond alpha-synuclein detection. J Neural Transm (Vienna) 2021; 129:1095-1103. [PMID: 34816335 DOI: 10.1007/s00702-021-02445-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 11/17/2021] [Indexed: 11/24/2022]
Abstract
Alpha-synuclein deposits, the pathological hallmarks of Parkinson's disease, are consistently found in the gastrointestinal tract of parkinsonian subjects. These observations have raised the potential that endoscopically obtainable mucosal biopsies can aid to a molecular diagnosis of the disease. The possible usefulness of mucosal biopsies is, however, not limited to the detection of alpha-synuclein, but also extends to other essential aspects underlying pathophysiological mechanisms of gastrointestinal manifestations in Parkinson's disease. The aim of the current review is to provide an appraisal of the existing studies showing that gastrointestinal biopsies can be used for the analysis of enteric neuronal and glial cell morphology, intestinal epithelial barrier function, and gastrointestinal inflammation in Parkinson's disease. A perspective on the generation of organoids with GI biopsies and the potential use of single-cell and spatial transcriptomic technologies will be also addressed.
Collapse
Affiliation(s)
- Pascal Derkinderen
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Inserm U1235 Nantes, Université de Nantes, 1 rue Gaston Veil, 44035, Nantes, France. .,Department of Neurology, CHU Nantes, 44093, Nantes, France.
| | | | - Adrien de Guilhem de Lataillade
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Inserm U1235 Nantes, Université de Nantes, 1 rue Gaston Veil, 44035, Nantes, France.,Department of Neurology, CHU Nantes, 44093, Nantes, France
| | - Laurène Leclair-Visonneau
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Inserm U1235 Nantes, Université de Nantes, 1 rue Gaston Veil, 44035, Nantes, France.,Department of Physiology, CHU Nantes, 44093, Nantes, France
| | - Michel Neunlist
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Inserm U1235 Nantes, Université de Nantes, 1 rue Gaston Veil, 44035, Nantes, France
| | - Sébastien Paillusson
- Inserm, TENS, The Enteric Nervous System in Gut and Brain Diseases, IMAD, Inserm U1235 Nantes, Université de Nantes, 1 rue Gaston Veil, 44035, Nantes, France
| | - Roberto De Giorgio
- Department of Translational Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
18
|
Gorecki AM, Anyaegbu CC, Anderton RS. TLR2 and TLR4 in Parkinson's disease pathogenesis: the environment takes a toll on the gut. Transl Neurodegener 2021; 10:47. [PMID: 34814947 PMCID: PMC8609261 DOI: 10.1186/s40035-021-00271-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/29/2021] [Indexed: 02/08/2023] Open
Abstract
Parkinson's disease (PD) is an incurable, devastating disorder that is characterized by pathological protein aggregation and neurodegeneration in the substantia nigra. In recent years, growing evidence has implicated the gut environment and the gut-brain axis in the pathogenesis and progression of PD, especially in a subset of people who exhibit prodromal gastrointestinal dysfunction. Specifically, perturbations of gut homeostasis are hypothesized to contribute to α-synuclein aggregation in enteric neurons, which may spread to the brain over decades and eventually result in the characteristic central nervous system manifestations of PD, including neurodegeneration and motor impairments. However, the mechanisms linking gut disturbances and α-synuclein aggregation are still unclear. A plethora of research indicates that toll-like receptors (TLRs), especially TLR2 and TLR4, are critical mediators of gut homeostasis. Alongside their established role in innate immunity throughout the body, studies are increasingly demonstrating that TLR2 and TLR4 signalling shapes the development and function of the gut and the enteric nervous system. Notably, TLR2 and TLR4 are dysregulated in patients with PD, and may thus be central to early gut dysfunction in PD. To better understand the putative contribution of intestinal TLR2 and TLR4 dysfunction to early α-synuclein aggregation and PD, we critically discuss the role of TLR2 and TLR4 in normal gut function as well as evidence for altered TLR2 and TLR4 signalling in PD, by reviewing clinical, animal model and in vitro research. Growing evidence on the immunological aetiology of α-synuclein aggregation is also discussed, with a focus on the interactions of α-synuclein with TLR2 and TLR4. We propose a conceptual model of PD pathogenesis in which microbial dysbiosis alters the permeability of the intestinal barrier as well as TLR2 and TLR4 signalling, ultimately leading to a positive feedback loop of chronic gut dysfunction promoting α-synuclein aggregation in enteric and vagal neurons. In turn, α-synuclein aggregates may then migrate to the brain via peripheral nerves, such as the vagal nerve, to contribute to neuroinflammation and neurodegeneration typically associated with PD.
Collapse
Affiliation(s)
- Anastazja M Gorecki
- School of Biological Science, University of Western Australia, Crawley, WA, Australia.
- Neurodegenerative Disorders Research Group, Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia.
| | - Chidozie C Anyaegbu
- Curtin Health Innovation Research Institute, Ralph and Patricia Sarich Neuroscience Research Institute, Curtin University, Nedlands, WA, Australia
| | - Ryan S Anderton
- Faculty of Medicine, Nursing and Midwifery and Faculty of Health Sciences, University of Notre Dame Australia, Fremantle, WA, Australia
- School of Nursing, Midwifery, Health Sciences and Physiotherapy, University of Notre Dame Australia, Fremantle, WA, Australia
| |
Collapse
|
19
|
Reduced acetylcholine and elevated muscarinic receptor 2 in duodenal mucosa contribute to the impairment of mucus secretion in 6-hydroxydopamine-induced Parkinson's disease rats. Cell Tissue Res 2021; 386:249-260. [PMID: 34370080 DOI: 10.1007/s00441-021-03515-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 07/21/2021] [Indexed: 12/27/2022]
Abstract
Patients with Parkinson's disease (PD) have a higher incidence rate of duodenal ulcers. The mucus barrier provides the first line of defense for duodenal mucosal protection. However, it is unknown whether duodenal mucus secretion is affected in PD. In the present study, we used the rats microinjected 6-hydroxydopamine (6-OHDA) into the bilateral substantia nigra to investigate duodenal mucus secretion and potential therapeutic targets in duodenal ulcer in PD. Alcian blue-periodic acid-Schiff, transmission electron microscopy, immunofluorescence, duodenal mucosal incubation, and enzyme-linked immunosorbent assays were used. The 6-OHDA rats exhibited mucin accumulation and retention in duodenal goblet cells. Mucin granules were unable to fuse with the apical membranes of goblet cells, and the exocytosis ratio of goblet cells was significantly reduced. Moreover, decreased acetylcholine and increased muscarinic receptor 2 (M2R) levels were detected in the duodenal mucosa of 6-OHDA rats. Bilateral vagotomy rats were also characterized by defective duodenal mucus secretion and decreased acetylcholine with increased M2R levels in the duodenal mucosa. Application of the cholinomimetic drug carbachol or blocking M2R with methoctramine significantly promoted mucus secretion by goblet cells and increased MUC2 content in duodenal mucosa-incubated solutions from 6-OHDA and vagotomy rats. We conclude that the reduced acetylcholine and increased M2R contribute to the impaired duodenal mucus secretion of 6-OHDA rats. The study provides new insights into the mechanism of duodenal mucus secretion and potential therapeutic targets for the treatment of duodenal ulcers in PD patients.
Collapse
|
20
|
de Souza FRO, Ribeiro FM, Lima PMD. Implications of VIP and PACAP in Parkinson's Disease: What do we Know So Far? Curr Med Chem 2021; 28:1703-1715. [PMID: 32196442 DOI: 10.2174/0929867327666200320162436] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/30/2020] [Accepted: 02/03/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Parkinson's disease is one of the most common neurodegenerative disorders and although its aetiology is not yet fully understood, neuroinflammation has been identified as a key factor in the progression of the disease. Vasoactive intestinal peptide and pituitary adenylate-cyclase activating polypeptide are two neuropeptides that exhibit anti-inflammatory and neuroprotective properties, modulating the production of cytokines and chemokines and the behaviour of immune cells. However, the role of chemokines and cytokines modulated by the endogenous receptors of the peptides varies according to the stage of the disease. METHODS We present an overview of the relationship between some cytokines and chemokines with vasoactive intestinal peptide, pituitary adenylate cyclase activating polypeptide and their endogenous receptors in the context of Parkinson's disease neuroinflammation and oxidative stress, as well as the modulation of microglial cells by the peptides in this context. RESULTS The two peptides exhibit neuroprotective and anti-inflammatory properties in models of Parkinson's disease, as they ameliorate cognitive functions, decrease the level of neuroinflammation and promote dopaminergic neuronal survival. The peptides have been tested in a variety of in vivo and in vitro models of Parkinson's disease, demonstrating the potential for therapeutic application. CONCLUSION More studies are needed to establish the clinical use of vasoactive intestinal peptide and pituitary adenylate cyclase activating polypeptide as safe candidates for treating Parkinson's disease, as the use of the peptides in different stages of the disease could produce different results concerning effectiveness.
Collapse
Affiliation(s)
- Filipe Resende Oliveira de Souza
- Laboratory of Immunology and Microbiology, Department of Natural Sciences, Federal University of Sao Joao Del Rei, Praca Dom Helvecio, n. 74, Fabricas, 36301160, Sao Joao Del Rei, MG, Brazil
| | - Fabiola Mara Ribeiro
- Laboratory of Neurobiochemistry, Department of Biochemistry and Immunology, Federal University of Minas Gerais, MG, Brazil
| | - Patrícia Maria d'Almeida Lima
- Laboratory of Immunology and Microbiology, Department of Natural Sciences, Federal University of Sao Joao Del Rei, Praca Dom Helvecio, n. 74, Fabricas, 36301160, Sao Joao Del Rei, MG, Brazil
| |
Collapse
|
21
|
Toschi A, Galiazzo G, Piva A, Tagliavia C, Mazzuoli-Weber G, Chiocchetti R, Grilli E. Cannabinoid and Cannabinoid-Related Receptors in the Myenteric Plexus of the Porcine Ileum. Animals (Basel) 2021; 11:263. [PMID: 33494452 PMCID: PMC7912003 DOI: 10.3390/ani11020263] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/16/2021] [Accepted: 01/19/2021] [Indexed: 12/29/2022] Open
Abstract
An important piece of evidence has shown that molecules acting on cannabinoid receptors influence gastrointestinal motility and induce beneficial effects on gastrointestinal inflammation and visceral pain. The aim of this investigation was to immunohistochemically localize the distribution of canonical cannabinoid receptor type 1 (CB1R) and type 2 (CB2R) and the cannabinoid-related receptors transient potential vanilloid receptor 1 (TRPV1), transient potential ankyrin receptor 1 (TRPA1), and serotonin receptor 5-HT1a (5-HT1aR) in the myenteric plexus (MP) of pig ileum. CB1R, TRPV1, TRPA1, and 5-HT1aR were expressed, with different intensities in the cytoplasm of MP neurons. For each receptor, the proportions of the immunoreactive neurons were evaluated using the anti-HuC/HuD antibody. These receptors were also localized on nerve fibers (CB1R, TRPA1), smooth muscle cells of tunica muscularis (CB1R, 5-HT1aR), and endothelial cells of blood vessels (TRPV1, TRPA1, 5-HT1aR). The nerve varicosities were also found to be immunoreactive for both TRPV1 and 5-HT1aR. No immunoreactivity was documented for CB2R. Cannabinoid and cannabinoid-related receptors herein investigated showed a wide distribution in the enteric neurons and nerve fibers of the pig MP. These results could provide an anatomical basis for additional research, supporting the therapeutic use of cannabinoid receptor agonists in relieving motility disorders in porcine enteropathies.
Collapse
Affiliation(s)
- Andrea Toschi
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Via Tolara di Sopra, 50, 40064 Ozzano dell’Emilia, Italy; (A.T.); (G.G.); (A.P.); (C.T.); (E.G.)
| | - Giorgia Galiazzo
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Via Tolara di Sopra, 50, 40064 Ozzano dell’Emilia, Italy; (A.T.); (G.G.); (A.P.); (C.T.); (E.G.)
| | - Andrea Piva
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Via Tolara di Sopra, 50, 40064 Ozzano dell’Emilia, Italy; (A.T.); (G.G.); (A.P.); (C.T.); (E.G.)
- R&D Division, Vetagro S.p.A., via Porro 2, 42124 Reggio Emilia, Italy
| | - Claudio Tagliavia
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Via Tolara di Sopra, 50, 40064 Ozzano dell’Emilia, Italy; (A.T.); (G.G.); (A.P.); (C.T.); (E.G.)
| | - Gemma Mazzuoli-Weber
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Foundation, Bischofsholer Damm 15, 30173 Hannover, Germany;
| | - Roberto Chiocchetti
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Via Tolara di Sopra, 50, 40064 Ozzano dell’Emilia, Italy; (A.T.); (G.G.); (A.P.); (C.T.); (E.G.)
| | - Ester Grilli
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), University of Bologna, Via Tolara di Sopra, 50, 40064 Ozzano dell’Emilia, Italy; (A.T.); (G.G.); (A.P.); (C.T.); (E.G.)
- R&D Division, Vetagro, Inc., 116 W. Jackson Blvd., Suite #320, Chicago, IL 60604, USA
| |
Collapse
|
22
|
Chiocchetti R, Galiazzo G, Fracassi F, Giancola F, Pietra M. ACE2 Expression in the Cat and the Tiger Gastrointestinal Tracts. Front Vet Sci 2020; 7:514. [PMID: 32903561 PMCID: PMC7438561 DOI: 10.3389/fvets.2020.00514] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/06/2020] [Indexed: 01/15/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) has been identified as the functional receptor for Severe Acute Respiratory Syndrome—Coronavirus−2 (SARS-CoV-2). It has been identified in the human gastrointestinal tract (GIT), and SARS-CoV-2 has been isolated in human and animal fecal samples. The aim of the present study was to investigate the expression of ACE2 in the gastrointestinal tract of domestic (cat) and wild (tiger) felines. Samples of the pylorus, duodenum, and distal colon were collected from six cats and one tiger. The tissues were processed for immunofluorescence assay with an anti-human ACE2 antibody. Angiotensin-converting enzyme 2 was widely expressed in the gastrointestinal mucosa of the cats and the tiger. In both the species, ACE2-immunoreactivity (ACE2-IR) was expressed by the mucosal epithelial cells of the GIT and by the enteric neurons. In the cats, ACE2-IR was also expressed by the smooth muscle cells of the blood vessels and the tunica muscularis. The expression of the ACE2 receptor in enteric neurons may support the potential neurotropic properties of SARS-CoV-2. Although the evidence of ACE2-IR in the feline GIT does not necessarily indicate the possibility of viral replication and SARS-CoV-2 spread with stool, the findings in the present study could serve as an anatomical basis for additional studies considering the risk of the SARS-CoV-2 fecal-oral transmission between cats/felids, and between cats/felids and humans.
Collapse
Affiliation(s)
- Roberto Chiocchetti
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giorgia Galiazzo
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Federico Fracassi
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Fiorella Giancola
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Marco Pietra
- Department of Veterinary Medical Sciences (UNI EN ISO 9001:2008), Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
23
|
Li Y, Long S, Liu Q, Ma H, Li J, Xiaoqing W, Yuan J, Li M, Hou B. Gut microbiota is involved in the alleviation of loperamide-induced constipation by honey supplementation in mice. Food Sci Nutr 2020; 8:4388-4398. [PMID: 32884719 PMCID: PMC7455974 DOI: 10.1002/fsn3.1736] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/02/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022] Open
Abstract
Constipation is one of the most common functional gastrointestinal disorders accompanied with intestinal dysbiosis. Laxatives for constipation usually have side effects. Bee honey is a natural food with unique composition, antimicrobial properties, and bifidogenic effect. In order to assess whether honey can ameliorate loperamide-induced constipation in BALB/c mice through the alteration of the gut microbiota, the present study was undertaken. Mice were given Jarrah honey (7.5 g/kg body weight) by gavage once per day for 5 days. Fecal water content, intestinal transit rate together with the colon concentrations of substance P (SP), vasoactive intestinal peptide (VIP), and serotonin (5-hydroxytryptamine; 5-HT) were evaluated. Furthermore, we determined the effect of honey treatment on gut microbiota in mice using stool genomic 16S rRNA sequencing. As a result, honey showed an obvious improvement in fecal water content and alleviated constipation by modulating the microbial composition of the microbiota, and this was highly associated with a proportional decrease in gut Desulfovibrio. In addition, we found that the colon level of neurotransmitters SP and VIP was significantly related to microbial variations. Our results indicate that gut microbiota is involved in the alleviation of loperamide-induced constipation by honey supplementation in mice, and it could be considered as an evaluating parameter in constipation therapy strategies.
Collapse
Affiliation(s)
- Yuyuan Li
- Advanced Institute for Medical SciencesDalian Medical UniversityDalianChina
| | - Shangqin Long
- Department of MicroecologyCollege of Basic Medical ScienceDalian Medical UniversityDalianChina
| | - Qiaochu Liu
- Department of MicroecologyCollege of Basic Medical ScienceDalian Medical UniversityDalianChina
| | - Hong Ma
- Department of MicroecologyCollege of Basic Medical ScienceDalian Medical UniversityDalianChina
| | - Jianxin Li
- Department of MicroecologyCollege of Basic Medical ScienceDalian Medical UniversityDalianChina
| | - Wei Xiaoqing
- The Core Laboratory of Medical Molecular Biology of Liaoning ProvinceDalian Medical UniversityDalianChina
| | - Jieli Yuan
- Department of MicroecologyCollege of Basic Medical ScienceDalian Medical UniversityDalianChina
| | - Ming Li
- Department of MicroecologyCollege of Basic Medical ScienceDalian Medical UniversityDalianChina
| | - Binbin Hou
- The Second Hospital of Dalian Medical UniversityDalianChina
| |
Collapse
|
24
|
Lavin KM, Ge Y, Sealfon SC, Nair VD, Wilk K, McAdam JS, Windham ST, Kumar PL, McDonald MLN, Bamman MM. Rehabilitative Impact of Exercise Training on Human Skeletal Muscle Transcriptional Programs in Parkinson's Disease. Front Physiol 2020; 11:653. [PMID: 32625117 PMCID: PMC7311784 DOI: 10.3389/fphys.2020.00653] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/22/2020] [Indexed: 12/18/2022] Open
Abstract
Parkinson's disease (PD) is the most common motor neurodegenerative disease, and neuromuscular function deficits associated with PD contribute to disability. Targeting these symptoms, our laboratory has previously evaluated 16-week high-intensity resistance exercise as rehabilitative training (RT) in individuals with PD. We reported significant improvements in muscle mass, neuromuscular function (strength, power, and motor unit activation), indices of neuromuscular junction integrity, total and motor scores on the unified Parkinson's disease rating scale (UPDRS), and total and sub-scores on the 39-item PD Quality of Life Questionnaire (PDQ-39), supporting the use of RT to reverse symptoms. Our objective was to identify transcriptional networks that may contribute to RT-induced neuromuscular remodeling in PD. We generated transcriptome-wide skeletal muscle RNA-sequencing in 5 participants with PD [4M/1F, 67 ± 2 years, Hoehn and Yahr stages 2 (n = 3) and 3 (n = 2)] before and after 16-week high intensity RT to identify transcriptional networks that may in part underpin RT-induced neuromuscular remodeling in PD. Following RT, 304 genes were significantly upregulated, notably related to remodeling and nervous system/muscle development. Additionally, 402 genes, primarily negative regulators of muscle adaptation, were downregulated. We applied the recently developed Pathway-Level Information ExtractoR (PLIER) method to reveal coordinated gene programs (as latent variables, LVs) that differed in skeletal muscle among young (YA) and old (OA) healthy adults and PD (n = 12 per cohort) at baseline and in PD pre- vs. post-RT. Notably, one LV associated with angiogenesis, axon guidance, and muscle remodeling was significantly lower in PD than YA at baseline and was significantly increased by exercise. A different LV annotated to denervation, autophagy, and apoptosis was increased in both PD and OA relative to YA and was also reduced by 16-week RT in PD. Thus, this analysis identified two novel skeletal muscle transcriptional programs that are dysregulated by PD and aging, respectively. Notably, RT has a normalizing effect on both programs in individuals with PD. These results identify potential molecular transducers of the RT-induced improvements in neuromuscular remodeling and motor function that may aid in optimizing exercise rehabilitation strategies for individuals with PD.
Collapse
Affiliation(s)
- Kaleen M. Lavin
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Center for Exercise Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Yongchao Ge
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Stuart C. Sealfon
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Venugopalan D. Nair
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Katarzyna Wilk
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Advanced Research on Diagnostic Assays, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jeremy S. McAdam
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Center for Exercise Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Samuel T. Windham
- UAB Center for Exercise Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Surgery, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Preeti Lakshman Kumar
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Merry-Lynn N. McDonald
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Marcas M. Bamman
- Department of Cell, Developmental and Integrative Biology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Center for Exercise Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Birmingham/Atlanta VA Geriatric Research, Education, and Clinical Center, Birmingham, AL, United States
- Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
25
|
Giancola F, Volta U, Repossi R, Latorre R, Beeckmans D, Carbone F, Van den Houte K, Bianco F, Bonora E, Gori A, Costanzini A, Boschetti E, Caio G, Vanuytsel T, Stanghellini V, Tack J, De Giorgio R. Mast cell-nerve interactions correlate with bloating and abdominal pain severity in patients with non-celiac gluten / wheat sensitivity. Neurogastroenterol Motil 2020; 32:e13814. [PMID: 32022388 DOI: 10.1111/nmo.13814] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/15/2020] [Accepted: 01/17/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gastrointestinal (GI) and extra-GI symptoms/manifestations represent key clinical features of patients with non-celiac gluten/wheat sensitivity (NCG/WS). This study aimed to investigate neuro-immune (focusing on mast cells, MCs) interactions in the duodenal submucosa of patients with NCG/WS. METHODS Submucosal whole mounts from duodenal biopsies of 34 patients with self-reported NCG/WS, 28 with celiac disease (CD), 13 with functional dyspepsia (FD), and 24 healthy controls (HC) were analyzed by immunohistochemistry. Quantitative data on neuronal and MCs density and the percentage of MCs in close vicinity to nerves were obtained, and correlations among neurons, MC density and MC-nerve distance (D), and symptoms were assessed in the three groups. KEY RESULTS The number of submucosal neurons was not different among groups. In NCG/WS, MC density was not different from HC, while it was slightly increased vs. CD (P = .07) and significantly decreased vs. FD (P < .05). The percentage of MCs close to nerves (D < 15 µm) was similarly increased in all three pathological groups vs. HC (P < .001). In NCG/WS, MC infiltration correlated with bloating (P = .001) and abdominal pain severity (P = .03) and the percentage of MCs in proximity to neurons correlated with the number of GI symptoms (D < 5 µm; P = .05), bloating and abdominal pain severity (D < 15um; P = .01). CONCLUSIONS AND INFERENCES Submucosal MC infiltration and the close (within 15 µm) MC-to-nerve proximity in the duodenum of NCG/WS patients are features providing a histopathological basis to better understand GI symptoms in this condition.
Collapse
Affiliation(s)
- Fiorella Giancola
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Umberto Volta
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Roberta Repossi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Rocco Latorre
- Department Basic Science & Craniofacial Biology, New York University, New York City, New York
| | - Dorien Beeckmans
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Florencia Carbone
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Karen Van den Houte
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | - Francesca Bianco
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Elena Bonora
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Gori
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Anna Costanzini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Elisa Boschetti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Giacomo Caio
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Tim Vanuytsel
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | | | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders (TARGID), KU Leuven, Leuven, Belgium
| | | |
Collapse
|
26
|
Atrophic Myenteric and Submucosal Neurons Are Observed in Parkinson's Disease. PARKINSONS DISEASE 2019; 2019:7935820. [PMID: 31321021 PMCID: PMC6607708 DOI: 10.1155/2019/7935820] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/27/2019] [Accepted: 06/12/2019] [Indexed: 01/20/2023]
Abstract
Aim Parkinson's disease is often accompanied by gastrointestinal symptoms, especially constipation. Microscopic studies of the enteric nervous system and enteric neuropathy have often been performed by immunostaining in the myenteric plexa. The aim of the present study was to examine whether pathologic changes could be identified by conventional hematoxylin and eosin (H&E) staining and could also be seen in the submucosal plexa. Materials and Methods In 20 deceased cases (11 male/9 female) of Parkinson's disease, the intestinal tract was investigated for potential neuroganglionic disease. Ten cases (7 male/3 female) of non-Parkinson, intestinally asymptomatic individuals were used as controls. Specimens from the jejunum and colon were sampled. The material was treated with standard histopathological procedures, i.e., fixed in formaldehyde solution, dehydrated and embedded in paraffin, sectioned at 5 μm thickness, and stained with H&E and immunostaining for α-synuclein. Results In 15 cases (7 male/8 female) of Parkinson's disease, atrophic/pycnotic nerve plexus cells were present, i.e., signs of ganglionic degeneration in the submucosal and/or myenteric plexa, mostly identified in both loci, by H&E staining. In some cases, the degenerative signs were mild, however, corroborated by findings of α-synuclein deposits in the ganglion cells. The remaining 5 cases showed no signs of degeneration in the H&E staining, but immunostaining revealed minimal α-synuclein deposits in 3 cases. None of the controls showed any ganglionic degeneration/α-synuclein deposits. Conclusion It seems possible to identify a morphologic intestinal disease substrate in Parkinson's disease by H&E staining, showing ganglion cell pycnosis and degeneration in both plexa. This finding may indicate a potential to diagnose enteric neuropathy in highly accessible sites.
Collapse
|
27
|
Bellier JP, Yuan PQ, Mukaisho K, Tooyama I, Taché Y, Kimura H. A Novel Antiserum Against a Predicted Human Peripheral Choline Acetyltransferase (hpChAT) for Labeling Neuronal Structures in Human Colon. Front Neuroanat 2019; 13:37. [PMID: 31040770 PMCID: PMC6476985 DOI: 10.3389/fnana.2019.00037] [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] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/08/2019] [Indexed: 12/13/2022] Open
Abstract
Choline acetyltransferase (ChAT), the enzyme synthesizing acetylcholine (ACh), has an exon-skipping splice variant which is expressed preferentially in the peripheral nervous system (PNS) and thus termed peripheral ChAT (pChAT). A rabbit antiserum previously produced against rat pChAT (rpChAT) has been used for immunohistochemistry (IHC) to study peripheral cholinergic structures in various animals. The present study was undertaken to develop a specific antiserum against a predicted human pChAT (hpChAT) protein. A novel mouse antiserum has been successfully raised against a unique 14-amino acid sequence of hpChAT protein. Our Western blot using this antiserum (termed here anti-hpChAT serum) on human colon extracts revealed only a single band of 47 kDa, matching the deduced size of hpChAT protein. By IHC, the antiserum gave intense staining in many neuronal cells and fibers of human colon but not brain, and such a pattern of staining seemed identical with that reported in colon of various animals using anti-rpChAT serum. In the antibody-absorption test, hpChAT-immunoreactive staining in human colon was completely blocked by using the antiserum pre-absorbed with the antigen peptide. Double immunofluorescence in human colon moreover indicated that structures stained with anti-hpChAT were also stained with anti-rpChAT, and vice versa. hpChAT antiserum allowed the identification of cell types, as Dogiel type cells in intramural plexuses, and fiber innervation of colon muscles and mucosae. The present results demonstrate the specificity and reliability of the hpChAT antiserum as a novel tool for immunohistochemical studies in human colon, opening venues to map cholinergic innervation in other human PNS tissues.
Collapse
Affiliation(s)
- Jean-Pierre Bellier
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Pu-Qing Yuan
- CURE/Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, United States.,VA Greater Los Angeles Health System, Los Angeles, CA, United States
| | - Kenichi Mukaisho
- Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Tooyama
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| | - Yvette Taché
- CURE/Digestive Diseases Research Center, Vatche and Tamar Manoukian Digestive Diseases Division, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, United States.,VA Greater Los Angeles Health System, Los Angeles, CA, United States
| | - Hiroshi Kimura
- Molecular Neuroscience Research Center, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
28
|
Doi H, Sakakibara R, Masuda M, Tateno F, Aiba Y, Kishi M, Yamanishi T, Yamamoto T, Matsuoka K. Gastrointestinal function in dementia with Lewy bodies: a comparison with Parkinson disease. Clin Auton Res 2019; 29:633-638. [PMID: 30741396 DOI: 10.1007/s10286-019-00597-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 02/04/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE To investigate gastrointestinal function in dementia with Lewy bodies and Parkinson disease. METHODS We examined gastric emptying and colonic transit time in 19 dementia with Lewy bodies and 46 Parkinson disease patients. RESULTS Gastric emptying was longer in dementia with Lewy bodies than in Parkinson disease (p = 0.014). Colonic transit time tended to be longer in dementia with Lewy bodies than in Parkinson disease. There was no relationship between gastric emptying and colonic transit time, nor between gastric emptying, colonic transit time and age. CONCLUSION Gastric emptying was prolonged in dementia with Lewy bodies compared to Parkinson disease.
Collapse
Affiliation(s)
- Hirokazu Doi
- Pharmaceutical Unit, Sakura Medical Center, Toho University, Sakura, Japan
| | - Ryuji Sakakibara
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, 564-1 Shimoshizu, Sakura, 285-8741, Japan.
| | - Masayuki Masuda
- Pharmaceutical Unit, Sakura Medical Center, Toho University, Sakura, Japan
| | - Fuyuki Tateno
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, 564-1 Shimoshizu, Sakura, 285-8741, Japan
| | - Yosuke Aiba
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, 564-1 Shimoshizu, Sakura, 285-8741, Japan
| | - Masahiko Kishi
- Neurology, Internal Medicine, Sakura Medical Center, Toho University, 564-1 Shimoshizu, Sakura, 285-8741, Japan
| | | | | | - Katsuyoshi Matsuoka
- Gastroenterology, Internal Medicine, Sakura Medical Center, Toho University, Sakura, Japan
| |
Collapse
|
29
|
Liu J, Chang R, Zhang X, Wang Z, Wen J, Zhou T. Non-isoflavones Diet Incurred Metabolic Modifications Induced by Constipation in Rats via Targeting Gut Microbiota. Front Microbiol 2018; 9:3002. [PMID: 30564225 PMCID: PMC6288237 DOI: 10.3389/fmicb.2018.03002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/20/2018] [Indexed: 01/03/2023] Open
Abstract
Isoflavones, presenting in leguminous plants and the normal chow diet, are known to alter intestinal microbiota, yet their deficiency has not been widely studied for its effect on constipation in biochemical state of rats. Our previous study discovered the differences in pharmacokinetic traits of isoflavones from Semen sojae praeparatum fed with normal chow diet (ISO) and non-isoflavones diet (NISO). To gain insight into the key role of intestinal microbiota in constipation and metabolic differences caused by isoflavones deficiency, we observed a significant decrease in fecal pellet numbers, fecal water content, intestinal transit rate together with the serum concentrations of substance P (SP) and vasoactive intestinal peptide (VIP) in NISO group, compared with those in the ISO group. Following 16S rRNA compositional sequencing, results excluded the changes in intestinal microbiota over time and highlighted that a total of 5 phyla and 21 genera changed significantly, among which Firmicutes, Bacteroidetes, Blautia, Prevotella, Lactobacillus and Bifidobacterium were closely related to constipation. In addition, Lactobacillus, produceing β-glucosidase which contribute to biotransform glycosides into aglycons and exert the bioactivities consequently, was decreased after non-isoflavones diet intake. Meanwhile, predicted metagenomics indicated that the pathway of glycan biosynthesis and metabolism was markedly down-regulated after non-isoflavones diet intake. Taken together, the findings suggested that the changes in the dietary components could alter the biochemical state of rats, which may be triggered by the abnormal modifications facilitated by β-glucosidase-producing bacteria. Our study shed a new strategy to explore the relationship among disease phenotypes (D), intestinal microbiota (I), enzymes (E) and traits of metabolism (T) named as "DIET," which can provide a reference for further study of the mechanism in regulation of intestinal bacteria-mediated diet on diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Tingting Zhou
- School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
30
|
Chalazonitis A, Rao M. Enteric nervous system manifestations of neurodegenerative disease. Brain Res 2018; 1693:207-213. [PMID: 29360466 PMCID: PMC6003851 DOI: 10.1016/j.brainres.2018.01.011] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/26/2022]
Abstract
Neurological disorders cause gastrointestinal (GI) symptoms that are debilitating and markedly diminish quality of life in patients. The enteric nervous system (ENS), the intrinsic nervous system of the GI tract that is often referred to as "the second brain", shares many features with the central nervous system. The ENS plays an essential role in regulating many GI functions including motility and fluid secretion. Enteric neuronal degeneration could therefore be responsible for the GI symptoms commonly observed in neurological conditions. Here we describe the organization and functions of the ENS and then review the evidence for ENS involvement in two common neurodegenerative disorders, Parkinson's disease (PD) and Alzheimer's disease (AD). Data from patients as well as animal models suggest that PD affects distinct subsets of neurons and glia in the ENS, and that the ENS may participate in the pathogenesis of this disorder. While there has been great enthusiasm for the possibility of sampling the ENS for diagnosis or therapeutic monitoring of PD, further work is needed to determine which enteric neurons are most affected and how ENS function could be modulated to ameliorate GI symptoms in patients. Although AD is far more common than PD and AD patients also experience GI symptoms, understanding of ENS dysfunction in AD is in its infancy. Much work remains to be done in both of these fields to determine how the ENS contributes to and/or is altered by these disorders, and how to target the ENS for more effective treatment of GI comorbidities.
Collapse
Affiliation(s)
- Alcmène Chalazonitis
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, P&S 11-511, New York, NY, USA.
| | - Meenakshi Rao
- Department of Pediatrics, Columbia University, 622 West 168th Street, PH 17, New York, NY, USA.
| |
Collapse
|
31
|
Lionnet A, Wade MA, Corbillé AG, Prigent A, Paillusson S, Tasselli M, Gonzales J, Durieu E, Rolli-Derkinderen M, Coron E, Duchalais E, Neunlist M, Perkinton MS, Hanger DP, Noble W, Derkinderen P. Characterisation of tau in the human and rodent enteric nervous system under physiological conditions and in tauopathy. Acta Neuropathol Commun 2018; 6:65. [PMID: 30037345 PMCID: PMC6055332 DOI: 10.1186/s40478-018-0568-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022] Open
Abstract
Tau is normally a highly soluble phosphoprotein found predominantly in neurons. Six different isoforms of tau are expressed in the adult human CNS. Under pathological conditions, phosphorylated tau aggregates are a defining feature of neurodegenerative disorders called tauopathies. Recent findings have suggested a potential role of the gut-brain axis in CNS homeostasis, and therefore we set out to examine the isoform profile and phosphorylation state of tau in the enteric nervous system (ENS) under physiological conditions and in tauopathies. Surgical specimens of human colon from controls, Parkinson's disease (PD) and progressive supranuclear palsy (PSP) patients were analyzed by Western Blot and immunohistochemistry using a panel of anti-tau antibodies. We found that adult human ENS primarily expresses two tau isoforms, localized in the cell bodies and neuronal processes. We did not observe any difference in the enteric tau isoform profile and phosphorylation state between PSP, PD and control subjects. The htau mouse model of tauopathy also expressed two main isoforms of human tau in the ENS, and there were no apparent differences in ENS tau localization or phosphorylation between wild-type and htau mice. Tau in both human and mouse ENS was found to be phosphorylated but poorly susceptible to dephosphorylation with lambda phosphatase. To investigate ENS tau phosphorylation further, primary cultures from rat enteric neurons, which express four isoforms of tau, were pharmacologically manipulated to show that ENS tau phosphorylation state can be regulated, at least in vitro. Our study is the first to characterize tau in the rodent and human ENS. As a whole, our findings provide a basis to unravel the functions of tau in the ENS and to further investigate the possibility of pathological changes in enteric neuropathies and tauopathies.
Collapse
|
32
|
Shannon K, Vanden Berghe P. The enteric nervous system in PD: gateway, bystander victim, or source of solutions. Cell Tissue Res 2018; 373:313-326. [PMID: 29936550 DOI: 10.1007/s00441-018-2856-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 05/10/2018] [Indexed: 12/19/2022]
Abstract
Apart from the characteristic and progressive motor- and movement-related problems, Parkinson's disease (PD) patients also suffer from several non-motor symptoms, including gastrointestinal dysfunction. The fact that the enteric nervous system (ENS) controls motility and that one of the typical PD hallmarks, α-synuclein-positive deposits, has also been found in the intestinal wall have rendered the ENS and the gut a popular subject of study in the context of PD. The possibility that these deposits could serve as an early biomarker is obviously of tremendous medical benefit but also the idea that the gut may possibly be a gateway via which the disease is initiated and progressively makes its way via the peripheral nerves to the central nervous system has increased the interest in the ENS-PD link. Furthermore, the fact that gastrointestinal symptoms are present in PD suggests that the ENS might be affected as well. However, despite a large body of literature on the topic, the actual role or the magnitude of involvement of the ENS in PD remains elusive. The multitudes of experimental approaches and animal models have complicated the interpretation of results and the outcome of different studies does not necessarily align well. In this review, we chose to highlight some elements of interest and some items of confusion, particularly those where research should be focusing. We also list a number of open questions in the field that could serve as a guideline for future, preferably concerted research.
Collapse
Affiliation(s)
| | - Pieter Vanden Berghe
- Lab. for Enteric NeuroScience (LENS), Translational Research of Gastrointestinal Disorder (TARGID), CHROMETA, University of Leuven, Leuven, Belgium.
| |
Collapse
|
33
|
Peripheral and central autonomic nervous system: does the sympathetic or parasympathetic nervous system bear the brunt of the pathology during the course of sporadic PD? Cell Tissue Res 2018; 373:267-286. [PMID: 29869180 DOI: 10.1007/s00441-018-2851-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 05/03/2018] [Indexed: 01/24/2023]
Abstract
It is a well-established fact that the sympathetic, parasympathetic and enteric nervous systems are affected at early stages in Parkinson's disease (PD). However, it is not yet clarified whether the earliest pathological events preferentially occur in any of these three divisions of the autonomic nervous system (ANS). Significant involvement of the peripheral autonomic nervous system of the heart and gastrointestinal tract has been documented in PD. Accumulating evidence suggests that the PD pathology spreads centripetally from the peripheral to central nervous system through autonomic nerve fibers, implicating the ANS as a major culprit in PD pathogenesis and a potential target for therapy. This study begins with a brief overview of the structures of the central and peripheral autonomic nervous system and then outlines the major clinicopathological manifestations of cardiovascular and gastrointestinal disturbances in PD.
Collapse
|
34
|
Knudsen K, Fedorova TD, Bekker AC, Iversen P, Østergaard K, Krogh K, Borghammer P. Objective Colonic Dysfunction is Far more Prevalent than Subjective Constipation in Parkinson's Disease: A Colon Transit and Volume Study. JOURNAL OF PARKINSONS DISEASE 2018; 7:359-367. [PMID: 28157109 DOI: 10.3233/jpd-161050] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gastrointestinal function has received increased interest in the context of Parkinson's disease (PD). Constipation is among the most frequent non-motor symptoms, but our understanding of the underlying pathology is limited. Subjective constipation correlates poorly with objective markers. OBJECTIVE The aims were to evaluate colonic transit time and volume in PD and to correlate these measures with subjective symptoms and gastric emptying. METHODS Thirty-two PD patients and 26 controls were included. Colonic transit time, computed tomography-based volume estimation, and gastric emptying were performed as objective markers of gastrointestinal function. Subjective gastrointestinal symptoms were evaluated by three different questionnaires. RESULTS Seventy-nine percent of PD patients displayed prolonged colonic transit time (p < 0.0001) and 66% of patients had significantly increased colonic volume (p = 0.0002). Particularly the transverse and rectosigmoid segments were affected. There was no difference in gastric emptying time between groups. The prevalence of subjective constipation in PD patients was significantly lower and ranged from 3% to 38% depending on the type of questionnaire. CONCLUSIONS Significantly delayed colonic transit time and increased volume were frequent findings in PD patients, and objective dysfunction was considerably more prevalent than subjective constipation symptoms. Also, the prevalence of subjective constipation varied widely depending upon which questionnaire was employed. These findings highlight the need for more research on how to define constipation in PD and also the need for improved understanding of the relationship between subjective symptoms and objective dysfunction.
Collapse
Affiliation(s)
- Karoline Knudsen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Denmark
| | - Tatyana D Fedorova
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Denmark
| | - Anne C Bekker
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Denmark
| | - Peter Iversen
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Denmark
| | | | - Klaus Krogh
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Denmark
| | - Per Borghammer
- Department of Nuclear Medicine and PET Centre, Aarhus University Hospital, Denmark
| |
Collapse
|
35
|
Mikołajczyk A, Gonkowski S, Złotkowska D. Modulation of the main porcine enteric neuropeptides by a single low-dose of lipopolysaccharide (LPS) Salmonella Enteritidis. Gut Pathog 2017; 9:73. [PMID: 29255488 PMCID: PMC5727943 DOI: 10.1186/s13099-017-0225-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 12/06/2017] [Indexed: 12/31/2022] Open
Abstract
Background The present research was conducted to investigate the influence of a low, single dose of LPS, which does not result in any clinical symptoms of intoxication on the expression of selected neuropeptides within the intestines of the domestic pig. Methods This experiment was conducted on immature female pigs of the Pitrain × Duroc breed (n = five per group). Seven days after the intravenous injection of 10 mL saline solution for control animals and 5 μg/kg b.w. (in 10 mL saline solution) LPS Salmonella Enteritidis for the experimental group, the excised segments of duodenum, jejunum, ileum, ileocecal valve, caecum, descending colon, transverse colon, ascending colon and rectum were prepared to extract the main enteric neuropeptides, including GAL, NPY, SOM, SP, VIP. Results The results of this research indicate that single low-dose LPS S. Enteritidis produced changes in the content of the selected neuropeptides of the porcine intestine. The most visible changes were observed in the transverse colon, where LPS induced the increase of GAL expression from 19.41 ± 7.121 to 92.92 ± 11.447 ng/g tissue. Conclusion The exact functions of the substances studied and mechanisms of responses to LPS action depend on the sections of the intestines. The mechanisms of observed changes are not fully understood, but fluctuations in neuronal active substance levels may be connected with neurodegenerative and/or pro-inflammatory activity of LPS.
Collapse
Affiliation(s)
- Anita Mikołajczyk
- Department of Public Health, Epidemiology and Microbiology, Faculty of Health Sciences, University of Warmia and Mazury in Olsztyn, ul. Warszawska 30 Str., 10-082 Olsztyn, Poland
| | - Sławomir Gonkowski
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury in Olsztyn, Oczapowskiego 13 Str., 10-718 Olsztyn, Poland
| | - Dagmara Złotkowska
- Department of Food Immunology and Microbiology, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences in Olsztyn, Tuwima 10 Str., 10-748 Olsztyn, Poland
| |
Collapse
|
36
|
Ren X, Liu L, Gamallat Y, Zhang B, Xin Y. Enteromorpha and polysaccharides from enteromorpha ameliorate loperamide-induced constipation in mice. Biomed Pharmacother 2017; 96:1075-1081. [PMID: 29198923 DOI: 10.1016/j.biopha.2017.11.119] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Revised: 11/15/2017] [Accepted: 11/27/2017] [Indexed: 02/06/2023] Open
Abstract
Slow-transit constipation(STC)is a disease characterized by functional gastrointestinal disorder. Common laxatives used in clinical practice against constipation such as Senna have side effects. Enteromorpha(EP)is a common marine alga, and the polysaccharide extracted from EP has been reported possessing anti-cancer and anti-inflammation effects. The aim of this study is to investigate the effects of EP and Polysaccharides from Enteromorpha (PEP) on loperamide induced constipated mice model and illustrating mechanism of action. We investigated the effect of EP and PEP on fecal water content, defecation frequency and gastrointestinal transit (GI) time of loperamide-induced STC mice. In addition, serum Nitric Oxide (NO) content and vasoactive intestinal peptide receptor1 (VIPR1) as well as serotonin receptor (5-HT4) expression in the distal colon were analyzed. Furthermore, we determined the role of EP and PEP on microbiota distribution using stool genomic 16S rRNA sequencing. EP and PEP significantly enhanced intestinal motility function, and alleviated constipation associated intestinal inflammation. Moreover, EP and PEP significantly decreased serum NO concentration, down-regulated VIPR1 expression and up-regulated 5-HT4 expression in distal colon. Genomic stool DNA MiSeq Sequencing Analysis of microbiota community structures and distribution revealed that intestinal microecological changes caused by constipation recovered after both EP and PEP treatment. Our results indicate that EP and PEP are potent natural products which could be suggested in constipation therapy strategies.
Collapse
Affiliation(s)
- Xinxiu Ren
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Lei Liu
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yaser Gamallat
- Department of Biotechnology, Dalian Medical University, Dalian, China
| | - Bingqiang Zhang
- Key Laboratory of Cell Transplantation, Ministry of Health, Qingdao, China
| | - Yi Xin
- Department of Biotechnology, Dalian Medical University, Dalian, China.
| |
Collapse
|
37
|
Nolano M, Provitera V, Manganelli F, Iodice R, Stancanelli A, Caporaso G, Saltalamacchia A, Califano F, Lanzillo B, Picillo M, Barone P, Santoro L. Loss of cutaneous large and small fibers in naive and l-dopa-treated PD patients. Neurology 2017; 89:776-784. [PMID: 28747449 DOI: 10.1212/wnl.0000000000004274] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/30/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To study small and large fiber pathology in drug-naive and l-dopa-treated patients affected by Parkinson disease (PD) in early phases, before the occurrence of neuropathic electrophysiologic abnormalities. METHODS We enrolled 85 patients with idiopathic PD (male/female 49/36, age 61.3 ± 9.7 years) without electrophysiologic signs of neuropathy, including 48 participants naive to l-dopa treatment. All patients underwent clinical, functional, and morphologic assessment of sensory and autonomic nerves through dedicated questionnaires, quantitative sensory testing, sympathetic skin response, dynamic sweat test, and punch biopsies from glabrous and hairy skin. Sensory and autonomic innervation was visualized with specific antibodies and analyzed by confocal microscopy. Data were compared with those obtained from sex- and age-comparable healthy controls. In 35 patients, skin biopsies were performed bilaterally to evaluate side-to-side differences. RESULTS Intraepidermal nerve fiber density was lower in patients compared to controls in all the examined sites (p < 0.001). The loss was higher in the more affected side (p < 0.01). A loss of autonomic nerves to vessels, sweat glands, and arrector pili muscles and of Meissner corpuscles and their myelinated endings in glabrous skin was found (p < 0.001). Patients showed increased tactile and thermal thresholds, impairment of mechanical pain perception, and reduced sweat output (p < 0.001). The naive and l-dopa-treated groups differed only for Meissner corpuscle density (p < 0.001). CONCLUSIONS Both large and small fiber pathology occurs in the early stages of PD and may account for the sensory and autonomic impairment. l-Dopa affects the 2 populations of fibers differently.
Collapse
Affiliation(s)
- Maria Nolano
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy.
| | - Vincenzo Provitera
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Fiore Manganelli
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Rosa Iodice
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Annamaria Stancanelli
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Giuseppe Caporaso
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Annamaria Saltalamacchia
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Francesca Califano
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Bernardo Lanzillo
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Marina Picillo
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Paolo Barone
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| | - Lucio Santoro
- From the Neurology Department (M.N., V.P., A.S., G.C., A.S., F.C., B.L.), Istituti Clinici Scientifici Maugeri SpA Società Benefit, IRCCS Telese Terme, Benevento; Department of Neurosciences, Reproductive Sciences and Odontostomatology (F.M., R.I., L.S.), University Federico II of Naples; and Centre for Neurodegenerative Diseases (M.P., P.B.), Department of Medicine and Surgery, Neuroscience Section, University of Salerno, Italy
| |
Collapse
|