0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Fecal supernatants from dogs with idiopathic epilepsy activate enteric neurons

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Introduction

          Alterations in the composition and function of the gut microbiome have been reported in idiopathic epilepsy (IE), however, interactions of gut microbes with the enteric nervous system (ENS) in this context require further study. This pilot study examined how gastrointestinal microbiota (GIM), their metabolites, and nutrients contained in intestinal contents communicate with the ENS.

          Methods

          Fecal supernatants (FS) from healthy dogs and dogs with IE, including drug-naïve, phenobarbital (PB) responsive, and PB non-responsive dogs, were applied to cultured myenteric neurons to test their activation using voltage-sensitive dye neuroimaging. Additionally, the concentrations of short-chain fatty acids (SCFAs) in the FS were quantified.

          Results

          Our findings indicate that FS from all examined groups elicited neuronal activation. Notably, FS from PB non-responsive dogs with IE induced action potential discharge in a higher proportion of enteric neurons compared to healthy controls, which exhibited the lowest burst frequency overall. Furthermore, the highest burst frequency in enteric neurons was observed upon exposure to FS from drug-naïve dogs with IE. This frequency was significantly higher compared to that observed in PB non-responsive dogs with IE and showed a tendency to surpass that of healthy controls.

          Discussion

          Although observed disparities in SCFA concentrations across the various FS samples might be associated with the induced neuronal activity, a direct correlation remains elusive at this point. The obtained results hint at an involvement of the ENS in canine IE and set the basis for future studies.

          Related collections

          Most cited references72

          • Record: found
          • Abstract: found
          • Article: not found

          GPR41/FFAR3 and GPR43/FFAR2 as cosensors for short-chain fatty acids in enteroendocrine cells vs FFAR3 in enteric neurons and FFAR2 in enteric leukocytes.

          The expression of short-chain fatty acid receptors GPR41/FFAR3 and GPR43/ free fatty acid receptor 2 (FFAR2) was studied in the gastrointestinal tract of transgenic monomeric red fluorescent protein (mRFP) reporter mice. In the stomach free fatty acid receptor 3 (FFAR3)-mRFP was expressed in a subpopulation of ghrelin and gastrin cells. In contrast, strong expression of FFAR3-mRFP was observed in all cholecystokinin, glucose-dependent insulinotropic peptide (GIP), and secretin cells of the proximal small intestine and in all glucagon-like peptide-1 (GLP-1), peptide YY, and neurotensin cells of the distal small intestine. Throughout the colon and rectum, FFAR3-mRFP was strongly expressed in the large population of peptide YY and GLP-1 cells and in the neurotensin cells of the proximal colon. A gradient of expression of FFAR3-mRFP was observed in the somatostatin cells from less than 5% in the stomach to more than 95% in the rectum. Substance P-containing enterochromaffin cells displayed a similar gradient of FFAR3-mRFP expression throughout the small intestine. Surprisingly, FFAR3-mRFP was also expressed in the neuronal cells of the submucosal and myenteric ganglia. Quantitative PCR analysis of fluorescence-activated cell sorting (FACS) purified FFAR3-mRFP positive cells confirmed the coexpression with the various peptide hormones as well as key neuronal marker proteins. The FFAR2-mRFP reporter was strongly expressed in a large population of leukocytes in the lamina propria of in particular the small intestine but surprisingly only weakly in a subpopulation of enteroendocrine cells. Nevertheless, synthetic ligands specific for either FFAR3 or FFAR2 each released GLP-1 from colonic crypt cultures and the FFAR2 agonist mobilized intracellular Ca²⁺ in FFAR2 positive enteroendocrine cells. It is concluded that FFAR3-mRFP serves as a useful marker for the majority of enteroendocrine cells of the small and large intestine and that FFAR3 and FFAR2 both act as sensors for short-chain fatty acids in enteroendocrine cells, whereas FFAR3 apparently has this role alone in enteric neurons and FFAR2 in enteric leukocytes.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: found
            Is Open Access

            The role of inflammation in the development of epilepsy

            Epilepsy, a neurological disease characterized by recurrent seizures, is often associated with a history of previous lesions in the nervous system. Impaired regulation of the activation and resolution of inflammatory cells and molecules in the injured neuronal tissue is a critical factor to the development of epilepsy. However, it is still unclear as to how that unbalanced regulation of inflammation contributes to epilepsy. Therefore, one of the goals in epilepsy research is to identify and elucidate the interconnected inflammatory pathways in systemic and neurological disorders that may further develop epilepsy progression. In this paper, inflammatory molecules, in neurological and systemic disorders (rheumatoid arthritis, Crohn’s, Type I Diabetes, etc.) that could contribute to epilepsy development, are reviewed. Understanding the neurobiology of inflammation in epileptogenesis will contribute to the development of new biomarkers for better screening of patients at risk for epilepsy and new therapeutic targets for both prophylaxis and treatment of epilepsy.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Utilization of nutrients by isolated epithelial cells of the rat colon.

              Isolated suspensions of colonocytes from the rat were used to assess utilization, interaction, and fate of metabolic substrates normally obtained from colonic bacteria (acetate, propionate, butyrate) or derived from the blood circulation to the colonic mucosa (D-glucose, acetoacetate, L-glutamine). The short-chain fatty acid n-butyrate (10 mM), on its own, accounted for 86% of the total oxygen consumption and suppressed oxidation of endogenous fuel by 82%. Ths value was not altered by the addition of acetoacetate (5 mM), of L-glutamine (5 mM), or of D-glucose (10 mM). Activation of short-chain fatty acids by colonocytes proceeded in the order of butyrate greater than acetate greater than propionate. D-Glucose on its own accounted for 30% of the oxygen consumption by colonocytes and hardly suppressed utilization of endogenous fuels. Colonocytes utilized ketone bodies (acetoacetate) and produced them (acetoacetate and beta-hydroxybutyrate) from short-chain fatty acids. Considering the interaction of substrates, isolated colonic epithelial cells utilized respiratory fuels in the preferential order of butyrate greater than acetoacetate greater than glutamine greater than glucose. The high rate of CO2 production from butyrate should be a worthwhile means of examining the functional activity of the colonic mucosa clinically and in vivo.
                Bookmark

                Author and article information

                Contributors
                Role: Role: Role: Role: Role:
                URI : https://loop.frontiersin.org/people/1795364/overviewRole: Role: Role: Role: Role:
                Role: Role: Role: URI : https://loop.frontiersin.org/people/2637264/overview
                URI : https://loop.frontiersin.org/people/2146857/overviewRole: Role: Role:
                Role: Role: URI : https://loop.frontiersin.org/people/2637549/overview
                URI : https://loop.frontiersin.org/people/623377/overviewRole: Role:
                URI : https://loop.frontiersin.org/people/1423901/overviewRole: Role: Role:
                URI : https://loop.frontiersin.org/people/202497/overviewRole: Role: Role:
                URI : https://loop.frontiersin.org/people/56183/overviewRole: Role: Role: Role:
                Journal
                Front Neurosci
                Front Neurosci
                Front. Neurosci.
                Frontiers in Neuroscience
                Frontiers Media S.A.
                1662-4548
                1662-453X
                31 January 2024
                2024
                : 18
                : 1281840
                Affiliations
                [1] 1Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover Foundation , Hannover, Germany
                [2] 2Department of Small Animal Medicine and Surgery, University of Veterinary Medicine Hannover Foundation , Hannover, Germany
                [3] 3Center for Systems Neuroscience (ZSN) , Hannover, Germany
                [4] 4Veterinary Research and Academic Service, Faculty of Veterinary Medicine, Kasetsart University , Kamphaeng Saen, Nakhon Pathom, Thailand
                [5] 5Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University , College Station, TX, United States
                Author notes

                Edited by: Michel Neunlist, Institut National de la Santé et de la Recherche Médicale (INSERM), France

                Reviewed by: Elisa L. Hill-Yardin, RMIT University, Australia; Fabio Stabile, Southfields Veterinary Specialists, United Kingdom

                *Correspondence: Antja Watanangura, antja.watanangura@ 123456tiho-hannover.de

                These authors have contributed equally to this work and share first authorship

                Article
                10.3389/fnins.2024.1281840
                10864448
                38356649
                705fa048-ac91-4516-add2-003adb06b8e6
                Copyright © 2024 Elfers, Watanangura, Hoffmann, Suchodolski, Khattab, Pilla, Meller, Volk and Mazzuoli-Weber.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 23 August 2023
                : 15 January 2024
                Page count
                Figures: 3, Tables: 1, Equations: 0, References: 72, Pages: 12, Words: 9533
                Funding
                The author(s) declare financial support was received for the research, authorship, and/or publication of this article. This open access publication was funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)—491094227 “Open Access Publication Funding” and the University of Veterinary Medicine Hannover Foundation. AW is a holder of a scholarship from the Faculty of Veterinary Medicine, Kasetsart University, Thailand.
                Categories
                Neuroscience
                Original Research
                Custom metadata
                Gut-Brain Axis

                Neurosciences
                fecal supernatant,canine idiopathic epilepsy,myenteric neurons,enteric nervous system,microbiota-gut-brain axis,phenobarbital,short-chain fatty acids

                Comments

                Comment on this article