Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 481
Filter
1.
Biomed Pharmacother ; 179: 117290, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153433

ABSTRACT

Hydrogen sulfide (H2S) is a gaseous signaling molecule that influences digestive and nervous system functions. Enteric glial cells (EGCs) are integral to the enteric nervous system and play a role in regulating gastrointestinal motility. This study explored the dual effects of exogenous H2S on EGCs and the influence of apoptosis-related pathways and ion channels in EGCs. We also administered honokiol for further interventional studies. The results revealed that low-concentration H2S increased the mitochondrial membrane potential (MMP) of EGCs, decreased the whole-cell membrane potential, downregulated BAX and caspase-3, upregulated Bcl2 expression, reduced apoptosis, and promoted cell proliferation. The Ca2+ concentration, Cx43 mRNA, and protein expression were also increased. A high concentration of H2S had the opposite effect. In addition, GFAP mRNA expression was upregulated in the test-low group, downregulated in the test-high group, and upregulated in the test-high + Hon group. Honokiol treatment increased MMP, reduced whole-cell membrane potential, inhibited BAX and caspase-3 expression, increased Bcl2 expression, decreased cell apoptosis, and increased cell proliferation. The Ca2+ concentration, Cx43 mRNA, and protein expression were also upregulated. In conclusion, our study showed that exogenous H2S can bidirectionally regulate EGC proliferation and apoptosis by affecting MMP and cell membrane potential via the Bcl2/BAX/caspase-3 pathway and modulate Cx43-mediated Ca2+ responses in EGCs to regulate colonic motility bidirectionally. Honokiol can ameliorate the damage to EGCs induced by high H2S concentrations through the Bcl2/BAX/caspase-3 pathway and improve colon motility by increasing Cx43 expression and Ca2+ concentration.


Subject(s)
Apoptosis , Biphenyl Compounds , Calcium Signaling , Cell Proliferation , Connexin 43 , Hydrogen Sulfide , Lignans , Neuroglia , Rats, Sprague-Dawley , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Biphenyl Compounds/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , Lignans/pharmacology , Calcium Signaling/drug effects , Rats , Connexin 43/metabolism , Connexin 43/genetics , Membrane Potential, Mitochondrial/drug effects , Calcium/metabolism , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Cells, Cultured , Allyl Compounds , Phenols
2.
Int J Mol Sci ; 25(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39201268

ABSTRACT

2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), a persistent organic pollutant and a potent aryl hydrocarbon receptor (AHR) ligand, causes delayed intestinal motility and affects the survival of enteric neurons. In this study, we investigated the specific signaling pathways and molecular targets involved in TCDD-induced enteric neurotoxicity. Immortalized fetal enteric neuronal (IM-FEN) cells treated with 10 nM TCDD exhibited cytotoxicity and caspase 3/7 activation, indicating apoptosis. Increased cleaved caspase-3 expression with TCDD treatment, as assessed by immunostaining in enteric neuronal cells isolated from WT mice but not in neural crest cell-specific Ahr deletion mutant mice (Wnt1Cre+/-/Ahrb(fl/fl)), emphasized the pivotal role of AHR in this process. Importantly, the apoptosis in IM-FEN cells treated with TCDD was mediated through a ceramide-dependent pathway, independent of endoplasmic reticulum stress, as evidenced by increased ceramide synthesis and the reversal of cytotoxic effects with myriocin, a potent inhibitor of ceramide biosynthesis. We identified Sptlc2 and Smpd2 as potential gene targets of AHR in ceramide regulation by a chromatin immunoprecipitation (ChIP) assay in IM-FEN cells. Additionally, TCDD downregulated phosphorylated Akt and phosphorylated Ser9-GSK-3ß levels, implicating the PI3 kinase/AKT pathway in TCDD-induced neurotoxicity. Overall, this study provides important insights into the mechanisms underlying TCDD-induced enteric neurotoxicity and identifies potential targets for the development of therapeutic interventions.


Subject(s)
Apoptosis , Ceramides , Endoplasmic Reticulum Stress , Neurons , Polychlorinated Dibenzodioxins , Receptors, Aryl Hydrocarbon , Signal Transduction , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Animals , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , Signal Transduction/drug effects , Polychlorinated Dibenzodioxins/toxicity , Neurons/metabolism , Neurons/drug effects , Ceramides/metabolism , Enteric Nervous System/metabolism , Enteric Nervous System/drug effects
3.
An Acad Bras Cienc ; 96(suppl 1): e20230244, 2024.
Article in English | MEDLINE | ID: mdl-39140520

ABSTRACT

This study aimed to investigate the antioxidant and anti-inflammatory properties of quercetin on the cellular components of the Enteric Nervous System in the ileum of rats with arthritis. Rats were distributed into five groups: control (C), arthritic (AIA), arthritic treated with ibuprofen (AI), arthritic treated with quercetin (AQ) and arthritic treated with both ibuprofen and quercetin (AIQ). The ileum was processed for immunohistochemical techniques for HuC/D, calcitonin gene-related peptide, and vasoactive intestinal polypeptide. Measurements in histological sections, chemiluminescence assays, and total antioxidant capacity were also performed. Rheumatoid arthritis resulted in a decrease in neuronal density, yet neuroplasticity mechanisms were evident through observed changes in varicosities size and neuronal area compared to the control group. Reduced paw edema and neuroprotective effects were predominantly noted in both plexuses, as evidenced by the increased density preservation of HuC/D-IR neurons in the AIQ group. The increase of lipoperoxidation levels and paw edema volume in the AQ group was observed compared to the arthritic, whereas the AIQ group mainly showed similar results to those observed in the control. The enteropathy associated with arthritis proved to be significant in the field of gastroenterology, and the combination of quercetin and ibuprofen demonstrated promising anti-inflammatory and neuroprotective effects.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Ibuprofen , Quercetin , Rats, Wistar , Animals , Quercetin/pharmacology , Quercetin/therapeutic use , Ibuprofen/pharmacology , Ibuprofen/therapeutic use , Rats , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Antioxidants/pharmacology , Antioxidants/therapeutic use , Neurons/drug effects , Neurons/pathology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Enteric Nervous System/drug effects , Enteric Nervous System/pathology , Immunohistochemistry , Ileum/drug effects , Ileum/pathology
4.
Mol Med ; 30(1): 113, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095693

ABSTRACT

BACKGROUND: To explore whether nobiletin has a protective effect on high-fat diet (HFD)-induced enteric nerve injury and its underlying mechanism. METHODS: An obesity model was induced by a HFD. Nobiletin (100 mg/kg and 200 mg/kg) and vehicle were administered by gastric gavage for 4 weeks. Lee's index, body weight, OGTT and intestinal propulsion assays were performed before sacrifice. After sampling, lipids were detected using Bodipy 493/503; lipid peroxidation was detected using MDA and SOD kits and the expression of PGP 9.5, Trem2, GFAP, ß-tubulin 3, Bax, Bcl2, Nestin, P75 NTR, SOX10 and EDU was detected using immunofluorescence. The GDNF, p-AKT, AKT, p-FOXO3a, FOXO3a and P21 proteins were detected using western blotting. The relative mRNA expression levels of NOS2 were detected via qPCR. Primary enteric neural stem cells (ENSCs) were cultured. After ENSCs were treated with palmitic acid (PA) and nobiletin, CCK-8 and caspase-3/7 activity assays were performed to evaluate proliferation and apoptosis. RESULTS: HFD consumption caused colon lipid accumulation and peroxidation, induced enteric nerve damage and caused intestinal motor dysfunction. However, nobiletin reduced lipid accumulation and peroxidation in the colon; promoted Trem2, ß-tubulin 3, Nestin, P75NTR, SOX10 and Bcl2 expression; inhibited Bax and GFAP expression; reduced NOS2 mRNA transcription; and regulated the GDNF/AKT/FOXO3a/P21 pathway. Nobiletin also promoted PA-induced impairment of ENSCs. CONCLUSIONS: Nobiletin restored HFD-induced enteric nerve injury, which may be associated with inhibiting enteric nerve apoptosis, promoting enteric nerve survival and regulating the GDNF/AKT/FOXO3a/P21 pathway.


Subject(s)
Diet, High-Fat , Enteric Nervous System , Flavones , Forkhead Box Protein O3 , Glial Cell Line-Derived Neurotrophic Factor , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Forkhead Box Protein O3/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Diet, High-Fat/adverse effects , Signal Transduction/drug effects , Male , Flavones/pharmacology , Flavones/therapeutic use , Enteric Nervous System/metabolism , Enteric Nervous System/drug effects , Neuroglia/metabolism , Neuroglia/drug effects , Mice , Disease Models, Animal , Rats , Obesity/metabolism , Obesity/drug therapy , Apoptosis/drug effects
5.
Exp Physiol ; 109(9): 1545-1556, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38979869

ABSTRACT

Gut motility undergoes a switch from myogenic to neurogenic control in late embryonic development. Here, we report on the electrical events that underlie this transition in the enteric nervous system, using the GCaMP6f reporter in neural crest cell derivatives. We found that spontaneous calcium activity is tetrodotoxin (TTX) resistant at stage E11.5, but not at E18.5. Motility at E18.5 was characterized by periodic, alternating high- and low-frequency contractions of the circular smooth muscle; this frequency modulation was inhibited by TTX. Calcium imaging at the neurogenic-motility stages E18.5-P3 showed that CaV1.2-positive neurons exhibited spontaneous calcium activity, which was inhibited by nicardipine and 2-aminoethoxydiphenyl borate (2-APB). Our protocol locally prevented muscle tone relaxation, arguing for a direct effect of nicardipine on enteric neurons, rather than indirectly by its relaxing effect on muscle. We demonstrated that the ENS was mechanosensitive from early stages on (E14.5) and that this behaviour was TTX and 2-APB resistant. We extended our results on L-type channel-dependent spontaneous activity and TTX-resistant mechanosensitivity to the adult colon. Our results shed light on the critical transition from myogenic to neurogenic motility in the developing gut, as well as on the intriguing pathways mediating electro-mechanical sensitivity in the enteric nervous system. HIGHLIGHTS: What is the central question of this study? What are the first neural electric events underlying the transition from myogenic to neurogenic motility in the developing gut, what channels do they depend on, and does the enteric nervous system already exhibit mechanosensitivity? What is the main finding and its importance? ENS calcium activity is sensitive to tetrodotoxin at stage E18.5 but not E11.5. Spontaneous electric activity at fetal and adult stages is crucially dependent on L-type calcium channels and IP3R receptors, and the enteric nervous system exhibits a tetrodotoxin-resistant mechanosensitive response. Abstract figure legend Tetrodotoxin-resistant Ca2+ rise induced by mechanical stimulation in the E18.5 mouse duodenum.


Subject(s)
Calcium Channels, L-Type , Calcium , Enteric Nervous System , Gastrointestinal Motility , Neurons , Tetrodotoxin , Animals , Calcium Channels, L-Type/metabolism , Tetrodotoxin/pharmacology , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Enteric Nervous System/physiology , Mice , Neurons/drug effects , Neurons/metabolism , Neurons/physiology , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Calcium/metabolism , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Muscle, Smooth/physiology , Mice, Inbred C57BL , Calcium Channel Blockers/pharmacology , Female , Muscle Contraction/drug effects , Muscle Contraction/physiology , Nicardipine/pharmacology , Boron Compounds
6.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000048

ABSTRACT

Bisphenols are dangerous endocrine disruptors that pollute the environment. Due to their chemical properties, they are globally used to produce plastics. Structural similarities to oestrogen allow bisphenols to bind to oestrogen receptors and affect internal body systems. Most commonly used in the plastic industry is bisphenol A (BPA), which also has negative effects on the nervous, immune, endocrine, and cardiovascular systems. A popular analogue of BPA-bisphenol S (BPS) also seems to have harmful effects similar to BPA on living organisms. Therefore, with the use of double immunofluorescence labelling, this study aimed to compare the effect of BPA and BPS on the enteric nervous system (ENS) in mouse jejunum. The study showed that both studied toxins impact the number of nerve cells immunoreactive to substance P (SP), galanin (GAL), vasoactive intestinal polypeptide (VIP), the neuronal isoform of nitric oxide synthase (nNOS), and vesicular acetylcholine transporter (VAChT). The observed changes were similar in the case of both tested bisphenols. However, the influence of BPA showed stronger changes in neurochemical coding. The results also showed that long-term exposure to BPS significantly affects the ENS.


Subject(s)
Benzhydryl Compounds , Enteric Nervous System , Jejunum , Phenols , Sulfones , Animals , Phenols/toxicity , Benzhydryl Compounds/toxicity , Mice , Jejunum/drug effects , Jejunum/metabolism , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Sulfones/pharmacology , Sulfones/toxicity , Substance P/metabolism , Vasoactive Intestinal Peptide/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Male , Galanin/metabolism , Endocrine Disruptors/toxicity , Endocrine Disruptors/pharmacology , Nitric Oxide Synthase Type I/metabolism
7.
Nutrients ; 16(14)2024 Jul 14.
Article in English | MEDLINE | ID: mdl-39064711

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.


Subject(s)
Jejunum , Microplastics , Neurons , Animals , Jejunum/drug effects , Jejunum/metabolism , Swine , Microplastics/toxicity , Neurons/drug effects , Neurons/metabolism , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Substance P/metabolism , Vasoactive Intestinal Peptide/metabolism , Polyethylene Terephthalates , Nitric Oxide Synthase Type I/metabolism , Galanin/metabolism , Neuronal Plasticity/drug effects , Administration, Oral , Neurotransmitter Agents/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism , Male , Nerve Tissue Proteins
8.
Nat Commun ; 15(1): 4400, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782898

ABSTRACT

Digestive Chagas disease (DCD) is an enteric neuropathy caused by Trypanosoma cruzi infection. There is a lack of evidence on the mechanism of pathogenesis and rationales for treatment. We used a female C3H/HeN mouse model that recapitulates key clinical manifestations to study how infection dynamics shape DCD pathology and the impact of treatment with the front-line, anti-parasitic drug benznidazole. Curative treatment 6 weeks post-infection resulted in sustained recovery of gastrointestinal transit function, whereas treatment failure led to infection relapse and gradual return of DCD symptoms. Neuro/immune gene expression patterns shifted from chronic inflammation to a tissue repair profile after cure, accompanied by increased cellular proliferation, glial cell marker expression and recovery of neuronal density in the myenteric plexus. Delaying treatment until 24 weeks post-infection led to partial reversal of DCD, suggesting the accumulation of permanent tissue damage over the course of chronic infection. Our study shows that murine DCD pathogenesis is sustained by chronic T. cruzi infection and is not an inevitable consequence of acute stage denervation. The risk of irreversible enteric neuromuscular tissue damage and dysfunction developing highlights the importance of prompt diagnosis and treatment. These findings support the concept of treating asymptomatic, T. cruzi-infected individuals with benznidazole to prevent DCD development.


Subject(s)
Chagas Disease , Disease Models, Animal , Enteric Nervous System , Mice, Inbred C3H , Nitroimidazoles , Trypanocidal Agents , Trypanosoma cruzi , Animals , Chagas Disease/drug therapy , Chagas Disease/parasitology , Female , Trypanocidal Agents/pharmacology , Trypanocidal Agents/therapeutic use , Nitroimidazoles/pharmacology , Nitroimidazoles/therapeutic use , Trypanosoma cruzi/drug effects , Mice , Enteric Nervous System/drug effects , Nerve Regeneration/drug effects
9.
Auton Neurosci ; 253: 103176, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38669866

ABSTRACT

Tributyltin (TBT) is a biocide used in the formulation of antifouling paints and it is highly harmful. Despite the ban, the compound persists in the environment, contaminating marine foodstuffs and household products. Therefore, considering the route of exposure to the contaminant, the gastrointestinal tract (GIT) acts as an important barrier against harmful substances and is a potential biomarker for understanding the consequences of these agents. This work aimed to evaluate histological and neuronal alterations in the duodenum of male Wistar rats that received 20 ng/g TBT and 600 ng/g via gavage for 30 consecutive days. After the experimental period, the animals were euthanized, and the duodenum was intended for neuronal histochemistry (total and metabolically active populations) and histological routine (morphometry and histopathology). The results showed more severe changes in neuronal density and intestinal morphometry in rats exposed to 20 ng/g, such as total neuronal density decrease and reduction of intestinal layers. In rats exposed to 600 ng/g of TBT, it was possible to observe only an increase in intraepithelial lymphocytes. We conclude that TBT can be more harmful to intestinal homeostasis when consumed in lower concentrations.


Subject(s)
Duodenum , Neuronal Plasticity , Rats, Wistar , Trialkyltin Compounds , Animals , Trialkyltin Compounds/toxicity , Male , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Duodenum/drug effects , Duodenum/pathology , Neurons/drug effects , Neurons/pathology , Rats , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Enteric Nervous System/drug effects , Enteric Nervous System/pathology
10.
Sci Rep ; 12(1): 22076, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36543902

ABSTRACT

Enteric neuron degeneration has been observed during aging, and in individuals with metabolic dysfunction including obesity and diabetes. Honokiol, a naturally occurring compound, is an activator of Sirtuin-3 (SIRT3) that has antioxidant activity. Its role in modulating enteric neuron-specific neurodegeneration is unknown. We studied the effects of honokiol and its fluorinated analog, hexafluoro-honokiol, on enteric neuronal differentiation and survival. We used a previously established model of mouse primary enteric neuronal cells and an enteric neuronal cell line treated with palmitate (PA) and lipopolysaccharide (LPS) to induce mitochondrial dysfunction and enteric neuronal cell death. The effect of honokiol and hexafluoro-honokiol was assessed on neuronal phenotype, fiber density, differentiation, and pyroptosis. Honokiol and hexafluoro-honokiol significantly increased neuronal networks and fiber density in enteric neurons and increased levels of neuronal nitric oxide synthase and Choline acetyltransferase mRNA. Hexafluoro-honokiol and honokiol also significantly increased SIRT3 mRNA levels and suppressed palmitate and LPS-induced neuronal pyroptosis. SIRT3 knock-down prevented the hexafluoro-honokiol mediated suppression of mitochondrial superoxide release. Our data supports a neuroprotective effect of honokiol and its derivative and these could be used as prophylactic or therapeutic agents for treating enteric neurodegeneration and associated motility disorders.


Subject(s)
Enteric Nervous System , Sirtuin 3 , Animals , Mice , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Cell Differentiation/genetics , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Lipopolysaccharides/pharmacology , Neurons/metabolism , Palmitates/pharmacology , Sirtuin 3/genetics , Sirtuin 3/metabolism
11.
Am J Physiol Gastrointest Liver Physiol ; 322(1): G66-G78, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34755545

ABSTRACT

Allosteric modulators (AMs) are molecules that can fine-tune signaling by G protein-coupled receptors (GPCRs). Although they are a promising therapeutic approach for treating a range of disorders, allosteric modulation of GPCRs in the context of the enteric nervous system (ENS) and digestive dysfunction remains largely unexplored. This study examined allosteric modulation of the delta opioid receptor (DOR) in the ENS and assessed the suitability of DOR AMs for the treatment of irritable bowel syndrome (IBS) symptoms using mouse models. The effects of the positive allosteric modulator (PAM) of DOR, BMS-986187, on neurogenic contractions of the mouse colon and on DOR internalization in enteric neurons were quantified. The ability of BMS-986187 to influence colonic motility was assessed both in vitro and in vivo. BMS-986187 displayed DOR-selective PAM-agonist activity and orthosteric agonist probe dependence in the mouse colon. BMS-986187 augmented the inhibitory effects of DOR agonists on neurogenic contractions and enhanced reflex-evoked DOR internalization in myenteric neurons. BMS-986187 significantly increased DOR endocytosis in myenteric neurons in response to the weakly internalizing agonist ARM390. BMS-986187 reduced the generation of complex motor patterns in the isolated intact colon. BMS-986187 reduced fecal output and diarrhea onset in the novel environment stress and castor oil models of IBS symptoms, respectively. DOR PAMs enhance DOR-mediated signaling in the ENS and have potential benefit for the treatment of dysmotility. This study provides proof of concept to support the use of GPCR AMs for the treatment of gastrointestinal motility disorders.NEW & NOTEWORTHY This study assesses the use of positive allosteric modulation as a pharmacological approach to enhance opioid receptor signaling in the enteric nervous system. We demonstrate that selective modulation of endogenous delta opioid receptor signaling can suppress colonic motility without causing constipation. We propose that allosteric modulation of opioid receptor signaling may be a therapeutic strategy to normalize gastrointestinal motility in conditions such as irritable bowel syndrome.


Subject(s)
Enteric Nervous System/drug effects , Gastrointestinal Motility/drug effects , Receptors, Opioid, delta/drug effects , Xanthones/pharmacology , Analgesics, Opioid/pharmacology , Benzamides/pharmacology , Colon/drug effects , Enteric Nervous System/physiopathology , Gastrointestinal Motility/physiology , Humans , Receptors, Opioid/drug effects , Receptors, Opioid, delta/agonists , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/drug effects , Signal Transduction/drug effects
12.
Nutrients ; 13(12)2021 Nov 24.
Article in English | MEDLINE | ID: mdl-34959768

ABSTRACT

The aggregation of alpha-synuclein protein (αSyn) is a hallmark of Parkinson's disease (PD). Considerable evidence suggests that PD involves an early aggregation of αSyn in the enteric nervous system (ENS), spreading to the brain. While it has previously been reported that omega-3 polyunsaturated fatty acids (ω-3 PUFA) acts as neuroprotective agents in the brain in murine models of PD, their effect in the ENS remains undefined. Here, we studied the effect of dietary supplementation with docosahexaenoic acid (DHA, an ω-3 PUFA), on the ENS, with a particular focus on enteric dopaminergic (DAergic) neurons. Thy1-αSyn mice, which overexpress human αSyn, were fed ad libitum with a control diet, a low ω-3 PUFA diet or a diet supplemented with microencapsulated DHA and then compared with wild-type littermates. Our data indicate that Thy1-αSyn mice showed a lower density of enteric dopaminergic neurons compared with non-transgenic animals. This decrease was prevented by dietary DHA. Although we found that DHA reduced microgliosis in the striatum, we did not observe any evidence of peripheral inflammation. However, we showed that dietary intake of DHA promoted a build-up of ω-3 PUFA-derived endocannabinoid (eCB)-like mediators in plasma and an increase in glucagon-like peptide-1 (GLP-1) and the redox regulator, Nrf2 in the ENS. Taken together, our results suggest that DHA exerts neuroprotection of enteric DAergic neurons in the Thy1-αSyn mice, possibly through alterations in eCB-like mediators, GLP-1 and Nrf2.


Subject(s)
Dietary Supplements , Docosahexaenoic Acids/pharmacology , Enteric Nervous System/drug effects , Neuroprotective Agents/pharmacology , Synucleinopathies/drug therapy , Animals , Diet , Disease Models, Animal , Dopaminergic Neurons/drug effects , Mice , Mice, Transgenic , Thy-1 Antigens/metabolism , alpha-Synuclein/metabolism
13.
Gut Microbes ; 13(1): 1996848, 2021.
Article in English | MEDLINE | ID: mdl-34812107

ABSTRACT

Amyotrophic Lateral Sclerosis is a neuromuscular disease characterized by the progressive death of motor neurons and muscle atrophy. The gastrointestinal symptoms in ALS patients were largely ignored or underestimated. The relationship between the enteric neuromuscular system and microbiome in ALS progression is unknown. We performed longitudinal studies on the enteric neuron system (ENS) and microbiome in the ALS human-SOD1G93A (Superoxide Dismutase 1) transgenic mice. We treated age-matched wild-type and ALS mice with butyrate or antibiotics to investigate the microbiome and neuromuscular functions. We examined intestinal mobility, microbiome, an ENS marker GFAP (Glial Fibrillary Acidic Protein), a smooth muscle marker (SMMHC, Smooth Muscle Myosin Heavy Chain), and human colonoids. The distribution of human-G93A-SOD1 protein was tested as an indicator of ALS progression. At 2-month-old before ALS onset, SOD1G93A mice had significantly lower intestinal mobility, decreased grip strength, and reduced time in the rotarod. We observed increased GFAP and decreased SMMHC expression. These changes correlated with consistent increased aggregation of mutated SOD1G93A in the colon, small intestine, and spinal cord. Butyrate or antibiotics treated SOD1G93A mice had a significantly longer latency to fall in the rotarod test, reduced SOD1G93A aggregation, and enhanced enteric neuromuscular function. Feces from 2-month-old SOD1G93A mice significantly enhanced SOD1G93A aggregation in human colonoids transfected with a SOD1G93A-GFP plasmid. Longitudinal studies of microbiome data further showed the altered bacterial community related to autoimmunity (e.g., Clostridium sp. ASF502, Lachnospiraceae bacterium A4), inflammation (e.g., Enterohabdus Muris,), and metabolism (e.g., Desulfovibrio fairfieldensis) at 1- and 2-month-old SOD1G93A mice, suggesting the early microbial contribution to the pathological changes. We have demonstrated a novel link between the microbiome, hSOD1G93A aggregation, and intestinal mobility. Dysbiosis occurred at the early stage of the ALS mice before observed mutated-SOD1 aggregation and dysfunction of ENS. Manipulating the microbiome improves the muscle performance of SOD1G93A mice. We provide insights into the fundamentals of intestinal neuromuscular function and microbiome in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/microbiology , Dysbiosis/microbiology , Enteric Nervous System/physiopathology , Muscle, Smooth/physiopathology , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/physiopathology , Animals , Anti-Bacterial Agents/therapeutic use , Butyrates/therapeutic use , Disease Models, Animal , Dysbiosis/drug therapy , Dysbiosis/physiopathology , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Gastrointestinal Microbiome/drug effects , Gastrointestinal Motility/drug effects , Humans , Intestine, Small/innervation , Intestine, Small/metabolism , Intestine, Small/pathology , Intestine, Small/physiopathology , Longitudinal Studies , Mice , Mice, Transgenic , Muscle Strength/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/metabolism , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/microbiology , Protein Aggregation, Pathological/physiopathology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
14.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34769120

ABSTRACT

Side effects associated with nonsteroidal anti-inflammatory drugs (NSAIDs) treatment are a serious limitation of their use in anti-inflammatory therapy. The negative effects of taking NSAIDs include abdominal pain, indigestion nausea as well as serious complications such as bleeding and perforation. The enteric nervous system is involved in regulation of gastrointestinal functions through the release of neurotransmitters. The present study was designed to determine, for the first time, the changes in pituitary adenylate cyclase-activating polypeptide (PACAP), substance P (SP) and galanin (GAL) expression in porcine jejunum after long-term treatment with aspirin, indomethacin and naproxen. The study was performed on 16 immature pigs. The animals were randomly divided into four experimental groups: control, aspirin, indomethacin and naproxen. Control animals were given empty gelatin capsules, while animals in the test groups received selected NSAIDs for 28 days. Next, animals from each group were euthanized. Frozen sections were prepared from collected jejunum and subjected to double immunofluorescence staining. NSAIDs supplementation caused a significant increase in the population of PACAP-, SP- and GAL-containing enteric neurons in the porcine jejunum. Our results suggest the participation of the selected neurotransmitters in regulatory processes of the gastrointestinal function and may indicate the direct toxic effect of NSAIDs on the ENS neurons.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Enteric Nervous System/drug effects , Galanin/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Substance P/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Chronic Pain/drug therapy , Enteric Nervous System/metabolism , Female , Jejunum/innervation , Jejunum/metabolism , Neurons/drug effects , Neurons/metabolism , Swine
15.
Acta Med Okayama ; 75(5): 549-556, 2021.
Article in English | MEDLINE | ID: mdl-34703037

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. The loss of nigrostriatal dopaminergic neurons produces its characteristic motor symptoms, but PD patients also have non-motor symptoms such as constipation and orthostatic hypotension. The pathological hallmark of PD is the presence of α-synuclein-containing Lewy bodies and neurites in the brain. However, the PD pathology is observed in not only the central nervous system (CNS) but also in parts of the peripheral nervous system such as the enteric nervous system (ENS). Since constipation is a typical prodromal non-motor symptom in PD, often preceding motor symptoms by 10-20 years, it has been hypothesized that PD pathology propagates from the ENS to the CNS via the vagal nerve. Discovery of pharmacological and other methods to halt this progression of neurodegeneration in PD has the potential to improve millions of lives. Astrocytes protect neurons in the CNS by secretion of neurotrophic and antioxidative factors. Similarly, astrocyte-like enteric glial cells (EGCs) are known to secrete neuroprotective factors in the ENS. In this article, we summarize the neuroprotective function of astrocytes and EGCs and discuss therapeutic strategies for the prevention of neurodegeneration in PD targeting neurotrophic and antioxidative molecules in glial cells.


Subject(s)
Antioxidants/metabolism , Central Nervous System/drug effects , Enteric Nervous System/drug effects , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , Central Nervous System/cytology , Enteric Nervous System/cytology , Humans
16.
Am J Physiol Gastrointest Liver Physiol ; 321(6): G617-G627, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34643099

ABSTRACT

Glucagon-like peptide-1 (GLP-1), a well-known insulin secretagogue, is released from enteroendocrine L cells both luminally and basolaterally to exert different effects. Basolaterally released GLP-1 increases epithelial ion transport by activating CGRP-containing enteric afferent neurons. Although bath-applied GLP-1 reduced the contractility of colonic segments, GLP-1-induced stimulation of afferent neurons could also accelerate peristaltic contractions. Here, the roles of endogenous GLP-1 in regulating colonic peristalsis were investigated using isolated colonic segments. Isolated segments of rat proximal colon were placed in an organ bath, serosally perfused with oxygenated physiological salt solution, and luminally perfused with degassed 0.9% saline. Colonic wall motion was recorded using a video camera and converted into spatiotemporal maps. Intraluminal administration of GLP-1 (100 nM) stimulating the secretion of GLP-1 from L cells increased the frequency of oro-aboral propagating peristaltic contractions. The acceleratory effect of GLP-1 was blocked by luminally applied exendin-3 (9-39) (100 nM), a GLP-1 receptor antagonist. GLP-1-induced acceleration of peristaltic contractions was also prevented by bath-applied BIBN4069 (1 µM), a CGRP receptor antagonist. In colonic segments that had been exposed to bath-applied capsaicin (100 nM) that desensitizes extrinsic afferents, GLP-1 was still capable of exerting its prokinetic effect. Stimulation of endogenous GLP-1 secretion with a luminally applied cocktail of short-chain fatty acids (1 mM) increased the frequency of peristaltic waves in an exendin-3 (9-39)-sensitive manner. Thus, GLP-1 activates CGRP-expressing intrinsic afferents to accelerate peristalsis in the proximal colon. Short-chain fatty acids appear to stimulate endogenous GLP-1 secretion from L cells resulting in the acceleration of colonic peristalsis.NEW & NOTEWORTHY Glucagon-like peptide-1 (GLP-1) activates CGRP-containing intrinsic afferent neurons resulting in the acceleration of colonic peristalsis. Short-chain fatty acids stimulate the secretion of endogenous GLP-1 from L cells that accelerates colonic peristalsis. Thus, besides the well-known humoral insulinotropic action, GLP-1 exerts a local action via the activation of the enteric nervous system to accelerate colonic motility. Such a prokinetic action of GLP-1 could underlie the mechanisms causing diarrhea in patients with type-2 diabetes treated with GLP-1 analogs.


Subject(s)
Colon/metabolism , Enteroendocrine Cells/metabolism , Gastrointestinal Motility , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Colon/drug effects , Colon/innervation , Enteric Nervous System/drug effects , Enteric Nervous System/metabolism , Fatty Acids/pharmacology , Gastrointestinal Motility/drug effects , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , In Vitro Techniques , Male , Peptide Fragments/pharmacology , Peptides/pharmacology , Rats, Wistar
17.
Int J Mol Sci ; 22(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34638647

ABSTRACT

Bisphenol A (BPA) is a substance used in the manufacture of plastics which shows multidirectional adverse effects on living organisms. Since the main path of intoxication with BPA is via the gastrointestinal (GI) tract, the stomach and intestine are especially vulnerable to the impact of this substance. One of the main factors participating in the regulation of intestinal functions is the enteric nervous system (ENS), which is characterized by high neurochemical diversity. Neuregulin 1 (NRG1) is one of the lesser-known active substances in the ENS. During the present study (performed using the double immunofluorescence method), the co-localization of NRG1 with other neuronal substances in the ENS of the caecum and the ascending and descending colon has been investigated under physiological conditions and after the administration of BPA. The obtained results indicate that NRG1-positive neurons also contain substance P, vasoactive intestinal polypeptide, a neuronal isoform of nitric oxide synthase and galanin and the degree of each co-localization depend on the type of enteric plexus and the particular fragment of the intestine. Moreover, it has been shown that BPA generally increases the degree of co-localization of NRG1 with other substances.


Subject(s)
Benzhydryl Compounds/adverse effects , Enteric Nervous System/drug effects , Intestine, Large/drug effects , Neuregulin-1/metabolism , Neurons/drug effects , Phenols/adverse effects , Animals , Enteric Nervous System/metabolism , Intestine, Large/metabolism , Neurons/metabolism , Substance P/metabolism , Swine , Vasoactive Intestinal Peptide/metabolism
18.
Mil Med Res ; 8(1): 50, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34503577

ABSTRACT

By its nature, Gulf war illness (GWI) is multisymptomatic and affects several organ systems in the body. Along with other symptoms, veterans who suffer from GWI commonly report chronic gastrointestinal issues such as constipation, pain, indigestion, etc. However, until recently, most attention has been focused on neurological disturbances such as cognitive impairments, chronic fatigue, and chronic pain among affected veterans. With such high prevalence of gastrointestinal problems among Gulf war (GW) veterans, it is surprising that there is little research to investigate the mechanisms behind these issues. This review summarizes all the available works on the mechanisms behind gastrointestinal problems in GWI that have been published to date in various databases. Generally, these studies, which were done in rodent models, in vitro and human cohorts propose that an altered microbiome, a reactive enteric nervous system or a leaky gut among other possible mechanisms are the major drivers of gastrointestinal problems reported in GWI. This review aims to draw attention to the gastrointestinal tract as an important player in GWI disease pathology and a potential therapeutic target.


Subject(s)
Gastrointestinal Diseases/etiology , Persian Gulf Syndrome/complications , Enteric Nervous System/drug effects , Enteric Nervous System/physiopathology , Gastrointestinal Diseases/physiopathology , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Humans , Persian Gulf Syndrome/physiopathology , Veterans/statistics & numerical data
19.
Molecules ; 26(12)2021 Jun 21.
Article in English | MEDLINE | ID: mdl-34205534

ABSTRACT

Until recently, glia were considered to be a structural support for neurons, however further investigations showed that glial cells are equally as important as neurons. Among many different types of glia, enteric glial cells (EGCs) found in the gastrointestinal tract, have been significantly underestimated, but proved to play an essential role in neuroprotection, immune system modulation and many other functions. They are also said to be remarkably altered in different physiopathological conditions. A nutraceutical is defined as any food substance or part of a food that provides medical or health benefits, including prevention and treatment of the disease. Following the description of these interesting peripheral glial cells and highlighting their role in physiological and pathological changes, this article reviews all the studies on the effects of nutraceuticals as modulators of their functions. Currently there are only a few studies available concerning the effects of nutraceuticals on EGCs. Most of them evaluated molecules with antioxidant properties in systemic conditions, whereas only a few studies have been performed using models of gastrointestinal disorders. Despite the scarcity of studies on the topic, all agree that nutraceuticals have the potential to be an interesting alternative in the prevention and/or treatment of enteric gliopathies (of systemic or local etiology) and their associated gastrointestinal conditions.


Subject(s)
Enteric Nervous System/drug effects , Neuroglia/drug effects , Animals , Antioxidants/pharmacology , Dietary Supplements , Gastrointestinal Diseases/drug therapy , Gastrointestinal Tract/drug effects , Humans , Neurons/drug effects
20.
Cells ; 10(7)2021 06 23.
Article in English | MEDLINE | ID: mdl-34201851

ABSTRACT

Recently, the involvement of the nervous system in the pathology of allergic diseases has attracted increasing interest. However, the precise pathophysiological role of enteric neurons in food allergies has not been elucidated. We report the presence of functional high-affinity IgE receptors (FcεRIs) in enteric neurons. FcεRI immunoreactivities were observed in approximately 70% of cholinergic myenteric neurons from choline acetyltransferase-eGFP mice. Furthermore, stimulation by IgE-antigen elevated intracellular Ca2+ concentration in isolated myenteric neurons from normal mice, suggesting that FcεRIs are capable of activating myenteric neurons. Additionally, the morphological investigation revealed that the majority of mucosal mast cells were in close proximity to enteric nerve fibers in the colonic mucosa of food allergy mice. Next, using a newly developed coculture system of isolated myenteric neurons and mucosal-type bone-marrow-derived mast cells (mBMMCs) with a calcium imaging system, we demonstrated that the stimulation of isolated myenteric neurons by veratridine caused the activation of mBMMCs, which was suppressed by the adenosine A3 receptor antagonist MRE 3008F20. Moreover, the expression of the adenosine A3 receptor gene was detected in mBMMCs. Therefore, in conclusion, it is suggested that, through interaction with mucosal mast cells, IgE-antigen-activated myenteric neurons play a pathological role in further exacerbating the pathology of food allergy.


Subject(s)
Cell Communication , Enteric Nervous System/physiopathology , Food Hypersensitivity/immunology , Food Hypersensitivity/physiopathology , Intestinal Mucosa/immunology , Intestinal Mucosa/physiopathology , Mast Cells/immunology , Neurons/pathology , Adenosine/pharmacology , Adenosine A3 Receptor Antagonists/pharmacology , Animals , Antigens/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Communication/drug effects , Cells, Cultured , Enteric Nervous System/drug effects , Enteric Nervous System/immunology , Intestinal Mucosa/drug effects , Intracellular Space/metabolism , Male , Mast Cells/drug effects , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Myenteric Plexus/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Adenosine A3/genetics , Receptor, Adenosine A3/metabolism , Receptors, IgE/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL