Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Cells ; 11(21)2022 10 25.
Article in English | MEDLINE | ID: mdl-36359764

ABSTRACT

BACKGROUND: Intestinal ischemia and reperfusion (IRI) injury induces acute and long-lasting damage to the neuromuscular compartment and dysmotility. This study aims to evaluate the pathogenetic role of hyaluronan (HA), a glycosaminoglycan component of the extracellular matrix, as a modulator of the enteric neuronal and immune function and of the colonic microbiota during in vivo IRI in the rat small intestine. METHODS: mesenteric ischemia was induced in anesthetized adult male rats for 60 min, followed by 24 h reperfusion. Injured, sham-operated and non-injured animals were treated with the HA synthesis inhibitor, 4-methylumbelliferone (4-MU 25 mg/kg). Fecal microbiota composition was evaluated by Next Generation Sequencing. Neutrophil infiltration, HA homeostasis and toll like receptor (TLR2 and TLR4) expression in the small intestine were evaluated by immunohistochemical and biomolecular approaches (qRT-PCR and Western blotting). Neuromuscular responses were studied in vitro, in the absence and presence of the selective TLR2/4 inhibitor, Sparstolonin B (SsnB 10, 30 µM). RESULTS: 4-MU significantly reduced IRI-induced enhancement of potentially harmful Escherichia and Enterococcus bacteria. After IRI, HA levels, neutrophil infiltration, and TLR2 and TLR4 expression were significantly enhanced in the muscularis propria, and were significantly reduced to baseline levels by 4-MU. In the injured, but not in the non-injured and sham-operated groups, SsnB reduced both electrical field-stimulated (EFS, 0.1-40 Hz) contractions and EFS-induced (10 Hz) non-cholinergic non-adrenergic relaxations. CONCLUSIONS: enhanced HA levels after intestinal IRI favors harmful bacteria overgrowth, increases neutrophil infiltration and promotes the upregulation of bacterial target receptors, TLR2 and TLR4, in the muscularis propria, inducing a pro-inflammatory state. TLR2 and TLR4 activation may, however, underlay a provisional benefit on excitatory and inhibitory neuronal pathways underlying peristalsis.


Subject(s)
Microbiota , Reperfusion Injury , Animals , Male , Rats , Hyaluronic Acid/metabolism , Immunity , Intestine, Small/metabolism , Reperfusion Injury/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
2.
Int J Mol Sci ; 22(4)2021 Feb 05.
Article in English | MEDLINE | ID: mdl-33562721

ABSTRACT

The complex bidirectional communication system existing between the gastrointestinal tract and the brain initially termed the "gut-brain axis" and renamed the "microbiota-gut-brain axis", considering the pivotal role of gut microbiota in sustaining local and systemic homeostasis, has a fundamental role in the pathogenesis of Inflammatory Bowel Disease (IBD). The integration of signals deriving from the host neuronal, immune, and endocrine systems with signals deriving from the microbiota may influence the development of the local inflammatory injury and impacts also more distal brain regions, underlying the psychophysiological vulnerability of IBD patients. Mood disorders and increased response to stress are frequently associated with IBD and may affect the disease recurrence and severity, thus requiring an appropriate therapeutic approach in addition to conventional anti-inflammatory treatments. This review highlights the more recent evidence suggesting that alterations of the microbiota-gut-brain bidirectional communication axis may concur to IBD pathogenesis and sustain the development of both local and CNS symptoms. The participation of the main microbial-derived metabolites, also defined as "postbiotics", such as bile acids, short-chain fatty acids, and tryptophan metabolites in the development of IBD-associated gut and brain dysfunction will be discussed. The last section covers a critical evaluation of the main clinical evidence pointing to the microbiome-based therapeutic approaches for the treatment of IBD-related gastrointestinal and neuropsychiatric symptoms.


Subject(s)
Bacteria/chemistry , Brain/metabolism , Gastrointestinal Tract/microbiology , Inflammatory Bowel Diseases/drug therapy , Anti-Inflammatory Agents/therapeutic use , Bacteria/immunology , Bile Acids and Salts/metabolism , Dysbiosis , Fatty Acids, Volatile/metabolism , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/psychology , Severity of Illness Index , Tryptophan/metabolism
3.
Cells ; 11(1)2021 12 31.
Article in English | MEDLINE | ID: mdl-35011688

ABSTRACT

The commensal microbiota plays a fundamental role in maintaining host gut homeostasis by controlling several metabolic, neuronal and immune functions. Conversely, changes in the gut microenvironment may alter the saprophytic microbial community and function, hampering the positive relationship with the host. In this bidirectional interplay between the gut microbiota and the host, hyaluronan (HA), an unbranched glycosaminoglycan component of the extracellular matrix, has a multifaceted role. HA is fundamental for bacterial metabolism and influences bacterial adhesiveness to the mucosal layer and diffusion across the epithelial barrier. In the host, HA may be produced and distributed in different cellular components within the gut microenvironment, playing a role in the modulation of immune and neuronal responses. This review covers the more recent studies highlighting the relevance of HA as a putative modulator of the communication between luminal bacteria and the host gut neuro-immune axis both in health and disease conditions, such as inflammatory bowel disease and ischemia/reperfusion injury.


Subject(s)
Gastrointestinal Microbiome , Hyaluronic Acid/metabolism , Neuroimmunomodulation , Animals , Extracellular Matrix/metabolism , Homeostasis , Humans , Intestines/pathology
4.
Sci Rep ; 10(1): 11521, 2020 07 13.
Article in English | MEDLINE | ID: mdl-32661417

ABSTRACT

Intestinal ischemia/reperfusion (I/R) injury has severe consequences on myenteric neurons, which can be irreversibly compromised resulting in slowing of transit and hindered food digestion. Myenteric neurons synthesize hyaluronan (HA) to form a well-structured perineuronal net, which undergoes derangement when myenteric ganglia homeostasis is perturbed, i.e. during inflammation. In this study we evaluated HA involvement in rat small intestine myenteric plexus after in vivo I/R injury induced by clamping a branch of the superior mesenteric artery for 60 min, followed by 24 h of reperfusion. In some experiments, 4-methylumbelliferone (4-MU, 25 mg/kg), a HA synthesis inhibitor, was intraperitoneally administered to normal (CTR), sham-operated (SH) and I/R animals for 24 h. In longitudinal muscle myenteric plexus (LMMP) whole-mount preparations, HA binding protein staining as well as HA levels were significantly higher in the I/R group, and were reduced after 4-MU treatment. HA synthase 1 and 2 (HAS1 and HAS2) labelled myenteric neurons and mRNA levels in LMMPs increased in the I/R group with respect to CTR, and were reduced by 4-MU. The efficiency of the gastrointestinal transit was significantly reduced in I/R and 4-MU-treated I/R groups with respect to CTR and SH groups. In the 4-MU-treated I/R group gastric emptying was reduced with respect to the CTR, SH and I/R groups. Carbachol (CCh) and electrical field (EFS, 0.1-40 Hz) stimulated contractions and EFS-induced (10 Hz) NANC relaxations were reduced in the I/R group with respect to both CTR and SH groups. After I/R, 4-MU treatment increased EFS contractions towards control values, but did not affect CCh-induced contractions. NANC on-relaxations after I/R were not influenced by 4-MU treatment. Main alterations in the neurochemical coding of both excitatory (tachykinergic) and inhibitory pathways (iNOS, VIPergic) were also observed after I/R, and were influenced by 4-MU administration. Overall, our data suggest that, after an intestinal I/R damage, changes of HA homeostasis in specific myenteric neuron populations may influence the efficiency of the gastrointestinal transit. We cannot exclude that modulation of HA synthesis in these conditions may ameliorate derangement of the enteric motor function preventing, at least in part, the development of dysmotility.


Subject(s)
Gastrointestinal Transit/physiology , Hyaluronic Acid/metabolism , Intestine, Small/metabolism , Reperfusion Injury/metabolism , Animals , Disease Models, Animal , Ganglia/metabolism , Gastrointestinal Motility/genetics , Gastrointestinal Motility/physiology , Gastrointestinal Transit/genetics , Humans , Hyaluronan Synthases/genetics , Ileum/metabolism , Ileum/physiology , Intestine, Small/pathology , Myenteric Plexus/metabolism , Nervous System Physiological Phenomena , Neurons/metabolism , Neurons/pathology , Rats , Reperfusion Injury/genetics , Reperfusion Injury/pathology
5.
Am J Physiol Heart Circ Physiol ; 319(2): H507-H518, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32706268

ABSTRACT

The lymphatic system drains and propels lymph by extrinsic and intrinsic mechanisms. Intrinsic propulsion depends upon spontaneous rhythmic contractions of lymphatic muscles in the vessel walls and is critically affected by changes in the surrounding tissue like osmolarity and temperature. Lymphatics of the diaphragm display a steep change in contraction frequency in response to changes in temperature, and this, in turn, affects lymph flow. In the present work, we demonstrated in an ex vivo diaphragmatic tissue rat model that diaphragmatic lymphatics express transient receptor potential channels of the vanilloid 4 subfamily (TRPV4) and that their blockade by both the nonselective antagonist Ruthenium Red and the selective antagonist HC-067047 abolished the response of lymphatics to temperature changes. Moreover, the selective activation of TRPV4 channels by means of GSK1016790A mirrored the behavior of vessels exposed to increasing temperatures, pointing out the critical role played by these channels in sensing the temperature of the lymphatic vessels' environment and thus inducing a change in contraction frequency and lymph flow.NEW & NOTEWORTHY The present work addresses the putative receptor system that enables diaphragmatic lymphatics to change intrinsic contraction frequency and thus lymph flow according to the changes in temperature of the surrounding environment, showing that this role can be sustained by TRPV4 channels alone.


Subject(s)
Lymph/physiology , Lymphatic Vessels/metabolism , Muscle Contraction , Muscle, Smooth/metabolism , TRPV Cation Channels/metabolism , Temperature , Animals , Diaphragm , Female , In Vitro Techniques , Lymphatic Vessels/drug effects , Male , Morpholines/pharmacology , Muscle, Smooth/drug effects , Periodicity , Pyrroles/pharmacology , Rats , Rats, Wistar , Ruthenium Red/pharmacology , Signal Transduction , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , Time Factors
6.
Int J Tryptophan Res ; 13: 1178646920928984, 2020.
Article in English | MEDLINE | ID: mdl-32577079

ABSTRACT

The 'microbiota-gut-brain axis' plays a fundamental role in maintaining host homeostasis, and different immune, hormonal, and neuronal signals participate to this interkingdom communication system between eukaryota and prokaryota. The essential aminoacid tryptophan, as a precursor of several molecules acting at the interface between the host and the microbiota, is fundamental in the modulation of this bidirectional communication axis. In the gut, tryptophan undergoes 3 major metabolic pathways, the 5-HT, kynurenine, and AhR ligand pathways, which may be directly or indirectly controlled by the saprophytic flora. The importance of tryptophan metabolites in the modulation of the gastrointestinal tract is suggested by several preclinical and clinical studies; however, a thorough revision of the available literature has not been accomplished yet. Thus, this review attempts to cover the major aspects on the role of tryptophan metabolites in host-microbiota cross-talk underlaying regulation of gut functions in health conditions and during disease states, with particular attention to 2 major gastrointestinal diseases, such as irritable bowel syndrome (IBS) and inflammatory bowel disease (IBD), both characterized by psychiatric disorders. Research in this area opens the possibility to target tryptophan metabolism to ameliorate the knowledge on the pathogenesis of both diseases, as well as to discover new therapeutic strategies based either on conventional pharmacological approaches or on the use of pre- and probiotics to manipulate the microbial flora.

7.
Cells ; 9(4)2020 03 31.
Article in English | MEDLINE | ID: mdl-32244316

ABSTRACT

Enteric glial cells (EGCs) influence nitric oxide (NO)- and adenosine diphosphate (ADP)- mediated signaling in the enteric nervous system (ENS). Since Toll-like receptor 4 (TLR4) participates to EGC homoeostasis, this study aimed to evaluate the possible involvement of EGCs in the alterations of the inhibitory neurotransmission in TLR4-/- mice. Ileal segments from male TLR4-/- and wild-type (WT) C57BL/6J mice were incubated with the gliotoxin fluoroacetate (FA). Alterations in ENS morphology and neurochemical coding were investigated by immunohistochemistry whereas neuromuscular responses were determined by recording non-adrenergic non-cholinergic (NANC) relaxations in isometrically suspended isolated ileal preparations. TLR4-/- ileal segments showed increased iNOS immunoreactivity associated with enhanced NANC relaxation, mediated by iNOS-derived NO and sensitive to P2Y1 inhibition. Treatment with FA diminished iNOS immunoreactivity and partially abolished NO- and ADP- mediated relaxation in the TLR4-/- mouse ileum, with no changes of P2Y1 and connexin-43 immunofluorescence distribution in the ENS. After FA treatment, S100ß and GFAP immunoreactivity in TLR4-/- myenteric plexus was reduced to levels comparable to those observed in WT. Our findings show the involvement of EGCs in the alterations of ENS architecture and in the increased purinergic and nitrergic-mediated relaxation, determining gut dysmotility in TLR4-/- mice.


Subject(s)
Enteric Nervous System/physiopathology , Intestine, Small/physiopathology , Neuroglia/metabolism , Neuromuscular Junction/physiopathology , Toll-Like Receptor 4/deficiency , Animals , Enteric Nervous System/drug effects , Fluoroacetates/pharmacology , Gliosis/complications , Gliosis/pathology , Gliosis/physiopathology , Ileum/drug effects , Ileum/pathology , Ileum/physiopathology , Intestine, Small/drug effects , Male , Mice, Inbred C57BL , Models, Biological , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neuroglia/drug effects , Neuromuscular Junction/drug effects , Phenotype , Receptors, Purinergic/metabolism , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Toll-Like Receptor 4/metabolism
8.
PeerJ ; 8: e8442, 2020.
Article in English | MEDLINE | ID: mdl-32095330

ABSTRACT

BACKGROUND: Inflammatory bowel diseases are associated with remodeling of neuronal circuitries within the enteric nervous system, occurring also at sites distant from the acute site of inflammation and underlying disturbed intestinal functions. Homeoproteins orthodenticle OTX1 and OTX2 are neuronal transcription factors participating to adaptation during inflammation and underlying tumor growth both in the central nervous system and in the periphery. In this study, we evaluated OTX1 and OTX2 expression in the rat small intestine and distal colon myenteric plexus after intrarectal dinitro-benzene sulfonic (DNBS) acid-induced colitis. METHODS: OTX1 and OTX2 distribution was immunohistochemically investigated in longitudinal muscle myenteric plexus (LMMP)-whole mount preparations. mRNAs and protein levels of both OTX1 and OTX2 were evaluated by qRT-PCR and Western blotting in LMMPs. RESULTS: DNBS-treatment induced major gross morphology and histological alterations in the distal colon, while the number of myenteric neurons was significantly reduced both in the small intestine and colon. mRNA levels of the inflammatory markers, TNFα, pro-IL1ß, IL6, HIF1α and VEGFα and myeloperoxidase activity raised in both regions. In both small intestine and colon, an anti-OTX1 antibody labeled a small percentage of myenteric neurons, and prevalently enteric glial cells, as evidenced by co-staining with the glial marker S100ß. OTX2 immunoreactivity was present only in myenteric neurons and was highly co-localized with neuronal nitric oxide synthase. Both in the small intestine and distal colon, the number of OTX1- and OTX2-immunoreactive myenteric neurons significantly increased after DNBS treatment. In these conditions, OTX1 immunostaining was highly superimposable with inducible nitric oxide synthase in both regions. OTX1 and OTX2 mRNA and protein levels significantly enhanced in LMMP preparations of both regions after DNBS treatment. CONCLUSIONS: These data suggest that colitis up-regulates OTX1 and OTX2 in myenteric plexus both on site and distantly from the injury, potentially participating to inflammatory-related myenteric ganglia remodeling processes involving nitrergic transmission.

10.
Toxins (Basel) ; 11(8)2019 07 31.
Article in English | MEDLINE | ID: mdl-31370176

ABSTRACT

Visceral pain, of which the pathogenic basis is currently largely unknown, is a hallmark symptom of both functional disorders, such as irritable bowel syndrome, and inflammatory bowel disease. Intrinsic sensory neurons in the enteric nervous system and afferent sensory neurons of the dorsal root ganglia, connecting with the central nervous system, represent the primary neuronal pathways transducing gut visceral pain. Current pharmacological therapies have several limitations, owing to their partial efficacy and the generation of severe adverse effects. Numerous cellular targets of visceral nociception have been recognized, including, among others, channels (i.e., voltage-gated sodium channels, VGSCs, voltage-gated calcium channels, VGCCs, Transient Receptor Potential, TRP, and Acid-sensing ion channels, ASICs) and neurotransmitter pathways (i.e., GABAergic pathways), which represent attractive targets for the discovery of novel drugs. Natural biologically active compounds, such as marine toxins, able to bind with high affinity and selectivity to different visceral pain molecular mediators, may represent a useful tool (1) to improve our knowledge of the physiological and pathological relevance of each nociceptive target, and (2) to discover therapeutically valuable molecules. In this review we report the most recent literature describing the effects of marine toxin on gastrointestinal visceral pain pathways and the possible clinical implications in the treatment of chronic pain associated with gut diseases.


Subject(s)
Gastrointestinal Diseases/drug therapy , Marine Toxins/therapeutic use , Visceral Pain/drug therapy , Animals , Gastrointestinal Diseases/physiopathology , Humans , Nociception , Visceral Pain/physiopathology
11.
Int J Mol Sci ; 20(6)2019 Mar 25.
Article in English | MEDLINE | ID: mdl-30934533

ABSTRACT

A complex bidirectional communication system exists between the gastrointestinal tract and the brain. Initially termed the "gut-brain axis" it is now renamed the "microbiota-gut-brain axis" considering the pivotal role of gut microbiota in maintaining local and systemic homeostasis. Different cellular and molecular pathways act along this axis and strong attention is paid to neuroactive molecules (neurotransmitters, i.e., noradrenaline, dopamine, serotonin, gamma aminobutyric acid and glutamate and metabolites, i.e., tryptophan metabolites), sustaining a possible interkingdom communication system between eukaryota and prokaryota. This review provides a description of the most up-to-date evidence on glutamate as a neurotransmitter/neuromodulator in this bidirectional communication axis. Modulation of glutamatergic receptor activity along the microbiota-gut-brain axis may influence gut (i.e., taste, visceral sensitivity and motility) and brain functions (stress response, mood and behavior) and alterations of glutamatergic transmission may participate to the pathogenesis of local and brain disorders. In this latter context, we will focus on two major gut disorders, such as irritable bowel syndrome and inflammatory bowel disease, both characterized by psychiatric co-morbidity. Research in this area opens the possibility to target glutamatergic neurotransmission, either pharmacologically or by the use of probiotics producing neuroactive molecules, as a therapeutic approach for the treatment of gastrointestinal and related psychiatric disorders.


Subject(s)
Brain/metabolism , Gastrointestinal Microbiome , Gastrointestinal Tract/metabolism , Glutamic Acid/metabolism , Signal Transduction , Animals , Humans , Neurotransmitter Agents/metabolism
13.
Methods Mol Biol ; 1952: 117-125, 2019.
Article in English | MEDLINE | ID: mdl-30825171

ABSTRACT

The cellular components of the enteric nervous system (ENS), namely enteric neurons and glia, display plasticity and respond to environmental cues deriving from growth factors, extracellular matrix (ECM) molecules, and cell-surface molecules, both in physiological and pathological conditions. ECM, in particular, provides an important framework for the enteric microenvironment and influences the homeostasis of myenteric neuronal circuitries. Isolation of pure myenteric plexus preparations from adult tissue permits to investigate changes in the ENS involving specific ECM, such as hyaluronan. This approach is based upon the possibility to isolate myenteric ganglia from the intestinal wall of either adult animals or humans, after microdissection and subsequent enzymatic digestion of the tissue. Enteric ganglia are free of connective tissue, extracellular collagen, and blood vessels, and thus treatment of intact intestinal segments with highly purified collagenases permits ganglia isolation from the surrounding smooth muscle cells. In this chapter, we describe methods for visualizing HA in isolated primary cultures of adult rat small intestine myenteric ganglia.


Subject(s)
Fluorescent Antibody Technique/methods , Ganglia/chemistry , Hyaluronic Acid/analysis , Intestine, Small/innervation , Microscopy, Confocal/methods , Myenteric Plexus/chemistry , Animals , Cells, Cultured , Ganglia/cytology , Ganglia/ultrastructure , Myenteric Plexus/cytology , Myenteric Plexus/ultrastructure , Rats , Tissue Culture Techniques/methods
14.
PLoS One ; 14(2): e0212856, 2019.
Article in English | MEDLINE | ID: mdl-30794676

ABSTRACT

Antibiotic use during adolescence may result in dysbiosis-induced neuronal vulnerability both in the enteric nervous system (ENS) and central nervous system (CNS) contributing to the onset of chronic gastrointestinal disorders, such as irritable bowel syndrome (IBS), showing significant psychiatric comorbidity. Intestinal microbiota alterations during adolescence influence the expression of molecular factors involved in neuronal development in both the ENS and CNS. In this study, we have evaluated the expression of brain-derived neurotrophic factor (BDNF) and its high-affinity receptor tropomyosin-related kinase B (TrkB) in juvenile mice ENS and CNS, after a 2-week antibiotic (ABX) treatment. In both mucosa and mucosa-deprived whole-wall small intestine segments of ABX-treated animals, BDNF and TrKB mRNA and protein levels significantly increased. In longitudinal muscle-myenteric plexus preparations of ABX-treated mice the percentage of myenteric neurons staining for BDNF and TrkB was significantly higher than in controls. After ABX treatment, a consistent population of BDNF- and TrkB-immunoreactive neurons costained with SP and CGRP, suggesting up-regulation of BDNF signaling in both motor and sensory myenteric neurons. BDNF and TrkB protein levels were downregulated in the hippocampus and remained unchanged in the prefrontal cortex of ABX-treated animals. Immunostaining for BDNF and TrkB decreased in the hippocampus CA3 and dentate gyrus subregions, respectively, and remained unchanged in the prefrontal cortex. These data suggest that dysbiosis differentially influences the expression of BDNF-TrkB in the juvenile mice ENS and CNS. Such changes may potentially contribute later to the development of functional gut disorders, such as IBS, showing psychiatric comorbidity.


Subject(s)
Anti-Bacterial Agents/adverse effects , Brain-Derived Neurotrophic Factor/biosynthesis , Brain/metabolism , Dysbiosis/metabolism , Enteric Nervous System/metabolism , Gene Expression Regulation/drug effects , Membrane Glycoproteins/biosynthesis , Protein-Tyrosine Kinases/biosynthesis , Animals , Anti-Bacterial Agents/pharmacology , Brain/pathology , Dysbiosis/chemically induced , Dysbiosis/pathology , Enteric Nervous System/pathology , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/pathology , Mice , Neurons/metabolism , Neurons/pathology , Signal Transduction/drug effects
15.
Purinergic Signal ; 14(4): 409-421, 2018 12.
Article in English | MEDLINE | ID: mdl-30269308

ABSTRACT

Adenosine is a versatile signaling molecule recognized to physiologically influence gut motor functions. Both the duration and magnitude of adenosine signaling in enteric neuromuscular function depend on its availability, which is regulated by the ecto-enzymes ecto-5'-nucleotidase (CD73), alkaline phosphatase (AP), and ecto-adenosine deaminase (ADA) and by dipyridamole-sensitive equilibrative transporters (ENTs). Our purpose was to assess the involvement of CD73, APs, ecto-ADA in the formation of AMP-derived adenosine in primary cultures of ileal myofibroblasts (IMFs). IMFs were isolated from rat ileum longitudinal muscle segments by means of primary explant technique and identified by immunofluorescence staining for vimentin and α-smooth muscle actin. IMFs confluent monolayers were exposed to exogenous 5'-AMP in the presence or absence of CD73, APs, ecto-ADA, or ENTs inhibitors. The formation of adenosine and its metabolites in the IMFs medium was monitored by high-performance liquid chromatography. The distribution of CD73 and ADA in IMFs was detected by confocal immunocytochemistry and qRT-PCR. Exogenous 5'-AMP was rapidly cleared being almost undetectable after 60-min incubation, while adenosine levels significantly increased. Treatment of IMFs with CD73 inhibitors markedly reduced 5'-AMP clearance whereas ADA blockade or inhibition of both ADA and ENTs prevented adenosine catabolism. By contrast, inhibition of APs did not affect 5'-AMP metabolism. Immunofluorescence staining and qRT-PCR analysis confirmed the expression of CD73 and ADA in IMFs. Overall, our data show that in IMFs an extracellular AMP-adenosine pathway is functionally active and among the different enzymatic pathways regulating extracellular adenosine levels, CD73 and ecto-ADA represent the critical catabolic pathway.


Subject(s)
5'-Nucleotidase/metabolism , Adenosine Deaminase/metabolism , Adenosine/metabolism , Intestine, Small/metabolism , Myofibroblasts/metabolism , Adenosine Monophosphate/metabolism , Alkaline Phosphatase/metabolism , Animals , Male , Rats, Wistar
16.
PLoS One ; 13(8): e0201760, 2018.
Article in English | MEDLINE | ID: mdl-30075006

ABSTRACT

We evaluated the chemical coding of the myenteric plexus in the proximal and distal intestine of gilthead sea bream (Sparus aurata), which represents one of the most farmed fish in the Mediterranean area. The presence of nitric oxide (NO), acetylcholine (ACh), serotonin (5-HT), calcitonin-gene-related peptide (CGRP), substance P (SP) and vasoactive intestinal peptide (VIP) containing neurons, was investigated in intestinal whole mount preparations of the longitudinal muscle with attached the myenteric plexus (LMMP) by means of immunohistochemical fluorescence staining. The main excitatory and inhibitory neurochemicals identified in intestinal smooth muscle were ACh, SP, 5HT, and NO, VIP, CGRP. Some neurons displayed morphological features of ascending and descending interneurons and of putative sensory neurons. The expression of these pathways in the two intestinal regions is largely superimposable, although some differences emerged, which may be relevant to the morphological properties of each region. The most important variances are the higher neuronal density and soma size in the proximal intestine, which may depend on the volume of the target tissue. Since in the fish gut the submucosal plexus is less developed, myenteric neurons substantially innervate also the submucosal and epithelial layers, which display a major thickness and surface in the proximal intestine. In addition, myenteric neurons containing ACh and SP, which mainly represent excitatory motor neurons and interneurons innervating the smooth muscle were more numerous in the distal intestine, possibly to sustain motility in the thicker smooth muscle coat. Overall, this study expands our knowledge of the intrinsic innervation that regulates intestinal secretion, absorption and motility in gilthead sea bream and provides useful background information for rational design of functional feeds aimed at improving fish gut health.


Subject(s)
Myenteric Plexus/cytology , Myenteric Plexus/metabolism , Neurons/cytology , Neurons/metabolism , Sea Bream/anatomy & histology , Sea Bream/metabolism , Animals , Cell Size , Immunohistochemistry , Muscle, Smooth/cytology , Muscle, Smooth/growth & development , Muscle, Smooth/innervation , Muscle, Smooth/metabolism , Myenteric Plexus/growth & development , Neurotransmitter Agents/metabolism , Sea Bream/growth & development
17.
Sci Rep ; 7(1): 17644, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29247178

ABSTRACT

Myenteric plexus alterations hamper gastrointestinal motor function during intestinal inflammation. Hyaluronan (HA), an extracellular matrix glycosaminoglycan involved in inflammatory responses, may play a role in this process. In the colon of control rats, HA-binding protein (HABP), was detected in myenteric neuron soma, perineuronal space and ganglia surfaces. Prominent hyaluronan synthase 2 (HAS2) staining was found in myenteric neuron cytoplasm, suggesting that myenteric neurons produce HA. In the myenteric plexus of rats with 2, 4-dinitrobenzene sulfonic (DNBS)-induced colitis HABP staining was altered in the perineuronal space, while both HABP staining and HA levels increased in the muscularis propria. HAS2 immunopositive myenteric neurons and HAS2 mRNA and protein levels also increased. Overall, these observations suggest that inflammation alters HA distribution and levels in the gut neuromuscular compartment. Such changes may contribute to alterations in the myenteric plexus.


Subject(s)
Colitis/metabolism , Hyaluronan Synthases/metabolism , Hyaluronic Acid/metabolism , Myenteric Plexus/metabolism , Neurons/metabolism , Animals , Benzenesulfonates , Cells, Cultured , Colitis/chemically induced , Colon/metabolism , Colon/pathology , Disease Models, Animal , Gastrointestinal Motility , Gene Expression Regulation , Humans , Male , Mitochondrial Proteins/metabolism , Protein Transport , Rats , Rats, Sprague-Dawley
18.
Br J Pharmacol ; 174(20): 3623-3639, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28755521

ABSTRACT

BACKGROUND AND PURPOSE: Gut microbiota is essential for the development of the gastrointestinal system, including the enteric nervous system (ENS). Perturbations of gut microbiota in early life have the potential to alter neurodevelopment leading to functional bowel disorders later in life. We examined the hypothesis that gut dysbiosis impairs the structural and functional integrity of the ENS, leading to gut dysmotility in juvenile mice. EXPERIMENTAL APPROACH: To induce gut dysbiosis, broad-spectrum antibiotics were administered by gavage to juvenile (3weeks old) male C57Bl/6 mice for 14 days. Bile acid composition in the intestinal lumen was analysed by liquid chromatography-mass spectrometry. Changes in intestinal motility were evaluated by stool frequency, transit of a fluorescent-labelled marker and isometric muscle responses of ileal full-thickness preparations to receptor and non-receptor-mediated stimuli. Alterations in ENS integrity were assessed by immunohistochemistry and Western blot analysis. KEY RESULTS: Antibiotic treatment altered gastrointestinal transit, luminal bile acid metabolism and bowel architecture. Gut dysbiosis resulted in distorted glial network, loss of myenteric plexus neurons, altered cholinergic, tachykininergic and nitrergic neurotransmission associated with reduced number of nNOS neurons and different ileal distribution of the toll-like receptor TLR2. Functional defects were partly reversed by activation of TLR2 signalling. CONCLUSIONS AND IMPLICATIONS: Gut dysbiosis caused complex morpho-functional neuromuscular rearrangements, characterized by structural defects of the ENS and increased tachykininergic neurotransmission. Altogether, our findings support the beneficial role of enteric microbiota for ENS homeostasis instrumental in ensuring proper gut neuromuscular function during critical stages of development.


Subject(s)
Anti-Bacterial Agents , Dysbiosis/physiopathology , Gastrointestinal Microbiome , Animals , Cecum/drug effects , Cecum/pathology , Dysbiosis/chemically induced , Dysbiosis/pathology , Gastrointestinal Motility/drug effects , Ileum/drug effects , Ileum/innervation , Ileum/pathology , Ileum/physiology , Male , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Myenteric Plexus/drug effects , Myenteric Plexus/pathology , Stomach/drug effects , Stomach/pathology , Synaptic Transmission/drug effects
19.
Microbiology (Reading) ; 162(9): 1554-1562, 2016 09.
Article in English | MEDLINE | ID: mdl-27516083

ABSTRACT

The antimicrobial power of honey seems to be ascribable to several factors, including oxidative and osmotic stress. The aim of this study was to find genetic determinants involved in the response to honey stress in the opportunistic pathogen Pseudomonas aeruginosa, chosen as model micro-organism. A library of transposon mutants of P. aeruginosa PAO1 was constructed and only four mutants unable to grow in presence of fir honeydew honey were selected. All four mutants were impaired in the major H2O2-scavenging enzyme catalase A (KatA). The knockout of katA gene caused sensitivity, as expected, not only to hydrogen peroxide but also to different types of honey including Manuka GMO 220 honey. Genetic complementation, as well as the addition of PAO1 supernatant containing extracellular catalase, restored tolerance to honey stress in all the mutants. As P. aeruginosa PAO1 catalase KatA copes with H2O2 stress, it is conceivable that the antimicrobial activity of honey is, at least partially, due to the presence of hydrogen peroxide in honey or the ability of honey to induce production of hydrogen peroxide. The katA-deficient mutants could be used as tester micro-organisms to compare the power of different types of natural and curative honeys in eliciting oxidative stress mediated by hydrogen peroxide.


Subject(s)
Bacterial Proteins/metabolism , Catalase/metabolism , Honey/analysis , Pseudomonas aeruginosa/enzymology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Catalase/genetics , Hydrogen Peroxide/pharmacology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...