Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Neurosci ; 35(20): 8021-34, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995484

ABSTRACT

Under physiological conditions, the voltage-gated sodium channel Nav1.8 is expressed almost exclusively in primary sensory neurons. The mechanism restricting Nav1.8 expression is not entirely clear, but we have previously described a 3.7 kb fragment of the Scn10a promoter capable of recapitulating the tissue-specific expression of Nav1.8 in transfected neurons and cell lines (Puhl and Ikeda, 2008). To validate these studies in vivo, a transgenic mouse encoding EGFP under the control of this putative sensory neuron specific promoter was generated and characterized in this study. Approximately 45% of dorsal root ganglion neurons of transgenic mice were EGFP-positive (mean diameter = 26.5 Āµm). The majority of EGFP-positive neurons bound isolectin B4, although a small percentage (Ć¢ĀˆĀ¼10%) colabeled with markers of A-fiber neurons. EGFP expression correlated well with the presence of Nav1.8 transcript (95%), Nav1.8-immunoreactivity (70%), and TTX-R INa (100%), although not all Nav1.8-expressing neurons expressed EGFP. Several cranial sensory ganglia originating from neurogenic placodes, such as the nodose ganglion, failed to express EGFP, suggesting that additional regulatory elements dictate Scn10a expression in placodal-derived sensory neurons. EGFP was also detected in discrete brain regions of transgenic mice. Quantitative PCR and Nav1.8-immunoreactivity confirmed Nav1.8 expression in the amygdala, brainstem, globus pallidus, lateral and paraventricular hypothalamus, and olfactory tubercle. TTX-R INa recorded from EGFP-positive hypothalamic neurons demonstrate the usefulness of this transgenic line to study novel roles of Nav1.8 beyond sensory neurons. Overall, Scn10a-EGFP transgenic mice recapitulate the majority of the Nav1.8 expression pattern in neural crest-derived sensory neurons.


Subject(s)
Cell Lineage , Green Fluorescent Proteins/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , Neural Crest/metabolism , Promoter Regions, Genetic , Animals , Female , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/genetics , Male , Mice , Mice, Transgenic , NAV1.8 Voltage-Gated Sodium Channel/genetics , Nerve Fibers, Myelinated/metabolism , Neural Crest/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Organ Specificity , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism
2.
J Neurosci ; 33(49): 19314-25, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305827

ABSTRACT

Free fatty acids receptor 3 (FFA3, GPR41) and 2 (FFA2, GPR43), for which the short-chain fatty acids (SCFAs) acetate and propionate are agonist, have emerged as important G-protein-coupled receptors influenced by diet and gut flora composition. A recent study (Kimura et al., 2011) demonstrated functional expression of FFA3 in the rodent sympathetic nervous system (SNS) providing a potential link between nutritional status and autonomic function. However, little is known of the source of endogenous ligands, signaling pathways, or effectors in sympathetic neurons. In this study, we found that FFA3 and FFA2 are unevenly expressed in the rat SNS with higher transcript levels in prevertebral (e.g., celiac-superior mesenteric and major pelvic) versus paravertebral (e.g., superior cervical and stellate) ganglia. FFA3, whether heterologously or natively expressed, coupled via PTX-sensitive G-proteins to produce voltage-dependent inhibition of N-type Ca(2+) channels (Cav2.2) in sympathetic neurons. In addition to acetate and propionate, we show that Ɵ-hydroxybutyrate (BHB), a metabolite produced during ketogenic conditions, is also an FFA3 agonist. This contrasts with previous interpretations of BHB as an antagonist at FFA3. Together, these results indicate that endogenous BHB levels, especially when elevated under certain conditions, such as starvation, diabetic ketoacidosis, and ketogenic diets, play a potentially important role in regulating the activity of the SNS through FFA3.


Subject(s)
3-Hydroxybutyric Acid/pharmacology , Calcium Channels, N-Type/drug effects , Neurons/physiology , Receptors, G-Protein-Coupled/agonists , Sympathetic Nervous System/physiology , Animals , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Electrophysiological Phenomena/physiology , Fluorescence Resonance Energy Transfer , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/physiology , HeLa Cells , Humans , In Situ Hybridization , Ketone Bodies/pharmacology , Ligands , Male , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Sympathetic Nervous System/cytology , Transfection
3.
Mol Pharmacol ; 83(1): 267-82, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23104136

ABSTRACT

Recent studies propose that N-arachidonyl glycine (NAGly), a carboxylic analogue of anandamide, is an endogenous ligand of the Gα(i/o) protein-coupled receptor 18 (GPR18). However, a high-throughput Ɵ-arrestin-based screen failed to detect activation of GPR18 by NAGly (Yin et al., 2009; JBC, 18:12328). To address this inconsistency, this study investigated GPR18 coupling in a native neuronal system with endogenous signaling pathways and effectors. GPR18 was heterologously expressed in rat sympathetic neurons, and the modulation of N-type (Ca(v)2.2) calcium channels was examined. Proper expression and trafficking of receptor were confirmed by the "rim-like" fluorescence of fluorescently tagged receptor and the positive staining of external hemagglutinin-tagged GPR18-expressing cells. Application of NAGly on GPR18-expressing neurons did not inhibit calcium currents but instead potentiated currents in a voltage-dependent manner, similar to what has previously been reported (Guo et al., 2008; J Neurophysiol, 100:1147). Other proposed agonists of GPR18, including anandamide and abnormal cannabidiol, also failed to induce inhibition of calcium currents. Mutants of GPR18, designed to constitutively activate receptors, did not tonically inhibit calcium currents, indicating a lack of GPR18 activation or coupling to endogenous G proteins. Other downstream effectors of Gα(i/o)-coupled receptors, G protein-coupled inwardly rectifying potassium channels and adenylate cyclase, were not modulated by GPR18 signaling. Furthermore, GPR18 did not couple to other G proteins tested: Gα(s), Gα(z), and Gα(15). These results suggest NAGly is not an agonist for GPR18 or that GPR18 signaling involves noncanonical pathways not examined in these studies.


Subject(s)
Arachidonic Acids/pharmacology , Glycine/analogs & derivatives , Receptors, G-Protein-Coupled/agonists , Animals , Calcium Channels, N-Type/physiology , Cell Membrane/metabolism , Cyclic AMP/metabolism , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , GTP-Binding Protein alpha Subunits/metabolism , Glycine/pharmacology , HEK293 Cells , HeLa Cells , Humans , In Vitro Techniques , Male , Mutation , Neurons/drug effects , Neurons/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Superior Cervical Ganglion/cytology
4.
Front Endocrinol (Lausanne) ; 14: 1169624, 2023.
Article in English | MEDLINE | ID: mdl-37560311

ABSTRACT

The gastrointestinal tract hosts the largest ecosystem of microorganisms in the body. The metabolism of ingested nutrients by gut bacteria produces novel chemical mediators that can influence chemosensory cells lining the gastrointestinal tract. Specifically, hormone-releasing enteroendocrine cells which express a host of receptors activated by these bacterial metabolites. This review will focus on the activation mechanisms of glucagon-like peptide-1 releasing enteroendocrine cells by the three main bacterial metabolites produced in the gut: short-chain fatty acids, secondary bile acids and indoles. Given the importance of enteroendocrine cells in regulating glucose homeostasis and food intake, we will also discuss therapies based on these bacterial metabolites used in the treatment of metabolic diseases such as diabetes and obesity. Elucidating the mechanisms gut bacteria can influence cellular function in the host will advance our understanding of this fundamental symbiotic relationship and unlock the potential of harnessing these pathways to improve human health.


Subject(s)
Gastrointestinal Microbiome , Metabolic Diseases , Humans , Indoles , Bile Acids and Salts/metabolism , Ecosystem , Enteroendocrine Cells/metabolism , Fatty Acids, Volatile/metabolism , Bacteria/metabolism , Metabolic Diseases/therapy , Metabolic Diseases/metabolism
5.
J Neurophysiol ; 108(2): 441-52, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22496528

ABSTRACT

Peripheral nerve injury promotes the release of brain-derived neurotrophic factor (BDNF) from spinal microglial cells and primary afferent terminals. This induces an increase in dorsal horn excitability that contributes to "central sensitization" and to the onset of neuropathic pain. Although it is accepted that impairment of GABAergic and/or glycinergic inhibition contributes to this process, certain lines of evidence suggest that GABA release in the dorsal horn may increase after nerve injury. To resolve these contradictory findings, we exposed rat spinal cord neurons in defined-medium organotypic culture to 200 ng/ml BDNF for 6 days to mimic the change in spinal BDNF levels that accompanies peripheral nerve injury. Morphological and electrophysiological criteria and glutamic acid decarboxylase (GAD) immunohistochemistry were used to distinguish putative inhibitory tonic-islet-central neurons from putative excitatory delay-radial neurons. Whole cell recording in the presence of 1 ĀµM tetrodotoxin showed that BDNF increased the amplitude of GABAergic and glycinergic miniature inhibitory postsynaptic currents (mIPSCs) in both cell types. It also increased the amplitude and frequency of spontaneous, action potential-dependent IPSCs (sIPSCs) in putative excitatory neurons. By contrast, BDNF reduced sIPSC amplitude in inhibitory neurons but frequency was unchanged. This increase in inhibitory drive to excitatory neurons and decreased inhibitory drive to inhibitory neurons seems inconsistent with the observation that BDNF increases overall dorsal horn excitability. One of several explanations for this discrepancy is that the action of BDNF in the substantia gelatinosa is dominated by previously documented increases in excitatory synaptic transmission rather than by impediment of inhibitory transmission.


Subject(s)
Action Potentials/physiology , Brain-Derived Neurotrophic Factor/pharmacology , Long-Term Potentiation/physiology , Neural Inhibition/physiology , Neurons/physiology , Substantia Gelatinosa/physiology , Synaptic Transmission/physiology , Action Potentials/drug effects , Animals , Long-Term Potentiation/drug effects , Neural Inhibition/drug effects , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Substantia Gelatinosa/drug effects , Synaptic Transmission/drug effects
6.
Can J Physiol Pharmacol ; 90(6): 783-90, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22506885

ABSTRACT

Changes in central neural processing are thought to contribute to the development of chronic osteoarthritis pain. This may be reflected as the presence of inflammatory mediators in the cerebral spinal fluid (CSF). We therefore exposed organotypically cultured slices of rat spinal cord to CSF from human subjects with osteoarthritis (OACSF) at a ratio of 1 part CSF in 9 parts culture medium for 5-6 days, and measured changes in neuronal electrophysiological properties by means of whole-cell recording. Although OACSF had no effect on the membrane properties and excitability of neurons in the substantia gelatinosa, synaptic transmission was clearly altered. The frequency of spontaneous excitatory postsynaptic currents (sEPSC) in delay-firing putative excitatory neurons was increased, as was sEPSC amplitude and frequency in tonic-firing inhibitory neurons. These changes could affect sensory processing in the dorsal horn, and may affect the transfer of nociceptive information. Although OACSF also affected inhibitory synaptic transmission (frequency of spontaneous inhibitory synaptic currents; sIPSC), this may have little bearing on sensory processing by substantia gelatinosa neurons, as sEPSC frequency is >3Ɨ greater than sIPSC frequency in this predominantly excitatory network. These results support the clinical notion that changes in nociceptive processing at the spinal level contribute to the generation of chronic osteoarthritis pain.


Subject(s)
Cerebrospinal Fluid/physiology , Osteoarthritis, Spine/cerebrospinal fluid , Posterior Horn Cells/physiology , Action Potentials/physiology , Adult , Aged , Animals , Chronic Pain/physiopathology , Excitatory Postsynaptic Potentials/physiology , Female , Humans , Male , Middle Aged , Nociception/physiology , Osteoarthritis, Spine/physiopathology , Patch-Clamp Techniques/methods , Posterior Horn Cells/physiopathology , Rats , Rats, Sprague-Dawley , Spinal Cord/pathology , Spinal Cord/physiopathology , Substantia Gelatinosa/physiology , Substantia Gelatinosa/physiopathology , Synaptic Transmission/physiology
7.
Nutrients ; 13(3)2021 Mar 09.
Article in English | MEDLINE | ID: mdl-33803183

ABSTRACT

The gastrointestinal tract can assess the nutrient composition of ingested food. The nutrient-sensing mechanisms in specialised epithelial cells lining the gastrointestinal tract, the enteroendocrine cells, trigger the release of gut hormones that provide important local and central feedback signals to regulate nutrient utilisation and feeding behaviour. The evidence for nutrient-stimulated secretion of two of the most studied gut hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), along with the known cellular mechanisms in enteroendocrine cells recruited by nutrients, will be the focus of this review. The mechanisms involved range from electrogenic transporters, ion channel modulation and nutrient-activated G-protein coupled receptors that converge on the release machinery controlling hormone secretion. Elucidation of these mechanisms will provide much needed insight into postprandial physiology and identify tractable dietary approaches to potentially manage nutrition and satiety by altering the secreted gut hormone profile.


Subject(s)
Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/metabolism , Gastrointestinal Tract/metabolism , Glucagon-Like Peptide 1/metabolism , Nutritional Physiological Phenomena/physiology , Bodily Secretions , Gastrointestinal Hormones/metabolism , Humans , Postprandial Period
8.
Sci Rep ; 11(1): 2249, 2021 01 26.
Article in English | MEDLINE | ID: mdl-33500423

ABSTRACT

Brain-derived neurotrophic factor (BDNF) is critically involved in the pathophysiology of chronic pain. However, the mechanisms of BDNF action on specific neuronal populations in the spinal superficial dorsal horn (SDH) requires further study. We used chronic BDNF treatment (200Ā ng/ml, 5-6Ā days) of defined-medium, serum-free spinal organotypic cultures to study intracellular calcium ([Ca2+]i) fluctuations. A detailed quantitative analysis of these fluctuations using the Frequency-independent biological signal identification (FIBSI) program revealed that BDNF simultaneously depressed activity in some SDH neurons while it unmasked a particular subpopulation of 'silent' neurons causing them to become spontaneously active. Blockade of gap junctions disinhibited a subpopulation of SDH neurons and reduced BDNF-induced synchrony in BDNF-treated cultures. BDNF reduced neuronal excitability assessed by measuring spontaneous excitatory postsynaptic currents. This was similar to the depressive effect of BDNF on the [Ca2+]i fluctuations. This study reveals novel regulatory mechanisms of SDH neuronal excitability in response to BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Posterior Horn Cells/physiology , 1-Octanol/pharmacology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Calcium/metabolism , Cluster Analysis , Excitatory Postsynaptic Potentials/drug effects , Gap Junctions/drug effects , Gap Junctions/metabolism , Models, Neurological , Posterior Horn Cells/drug effects , Rats
9.
Mol Metab ; 54: 101356, 2021 12.
Article in English | MEDLINE | ID: mdl-34662713

ABSTRACT

OBJECTIVE: Motilin is a proximal small intestinal hormone with roles in gastrointestinal motility, gallbladder emptying, and hunger initiation. InĀ vivo motilin release is stimulated by fats, bile, and duodenal acidification but the underlying molecular mechanisms of motilin secretion remain poorly understood. This study aimed to establish the key signaling pathways involved in the regulation of secretion from human motilin-expressing M-cells. METHODS: Human duodenal organoids were CRISPR-Cas9 modified to express the fluorescent protein Venus or the Ca2+ sensor GCaMP7s under control of the endogenous motilin promoter. This enabled the identification and purification of M-cells for bulk RNA sequencing, peptidomics, calcium imaging, and electrophysiology. Motilin secretion from 2D organoid-derived cultures was measured by liquid chromatography-tandem mass spectrometry (LC-MS/MS), in parallel with other gut hormones. RESULTS: Human duodenal M-cells synthesize active forms of motilin and acyl-ghrelin in organoid culture, and also co-express cholecystokinin (CCK). Activation of the bile acid receptor GPBAR1 stimulated a 3.4-fold increase in motilin secretion and increased action potential firing. Agonists of the long-chain fatty acid receptor FFA1 and monoacylglycerol receptor GPR119 stimulated secretion by 2.4-fold and 1.5-fold, respectively. Acidification (pH 5.0) was a potent stimulus of M-cell calcium elevation and electrical activity, an effect attributable to acid-sensing ion channels, and a modest inducer of motilin release. CONCLUSIONS: This study presents the first in-depth transcriptomic and functional characterization of human duodenal motilin-expressing cells. We identify several receptors important for the postprandial and interdigestive regulation of motilin release.


Subject(s)
Bile/metabolism , Duodenum/metabolism , Fatty Acids, Nonesterified/metabolism , Motilin/metabolism , Organoids/metabolism , Cells, Cultured , Humans , Hydrogen-Ion Concentration
10.
Peptides ; 140: 170532, 2021 06.
Article in English | MEDLINE | ID: mdl-33744371

ABSTRACT

OBJECTIVES: To analyse the peptidomics of mouse enteroendocrine cells (EECs) and human gastrointestinal (GI) tissue and identify novel gut derived peptides. METHODS: High resolution nano-flow liquid chromatography mass spectrometry (LC-MS/MS) was performed on (i) flow-cytometry purified NeuroD1 positive cells from mouse and homogenised human intestinal biopsies, (ii) supernatants from primary murine intestinal cultures, (iii) intestinal homogenates from mice fed high fat diet. Candidate bioactive peptides were selected on the basis of species conservation, high expression/biosynthesis in EECs and evidence of regulated secretionin vitro. Candidate novel gut-derived peptides were chronically administered to mice to assess effects on food intake and glucose tolerance. RESULTS: A large number of peptide fragments were identified from human and mouse, including known full-length gut hormones and enzymatic degradation products. EEC-specific peptides were largely from vesicular proteins, particularly prohormones, granins and processing enzymes, of which several exhibited regulated secretion in vitro. No regulated peptides were identified from previously unknown genes. High fat feeding particularly affected the distal colon, resulting in reduced peptide levels from GCG, PYY and INSL5. Of the two candidate novel peptides tested in vivo, a peptide from Chromogranin A (ChgA 435-462a) had no measurable effect, but a progastrin-derived peptide (Gast p59-79), modestly improved glucose tolerance in lean mice. CONCLUSION: LC-MS/MS peptidomic analysis of murine EECs and human GI tissue identified the spectrum of peptides produced by EECs, including a potential novel gut hormone, Gast p59-79, with minor effects on glucose tolerance.


Subject(s)
Enteroendocrine Cells/metabolism , Gastrins/pharmacology , Gastrointestinal Tract/metabolism , Glucose Tolerance Test/methods , Peptides/metabolism , Protein Precursors/pharmacology , Proteome/metabolism , Thinness/drug therapy , Animals , Cells, Cultured , Glucose/metabolism , Humans , Male , Mice , Models, Animal , Peptides/chemistry , Proteome/analysis , Thinness/metabolism
11.
Mol Pain ; 6: 44, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20653959

ABSTRACT

Peripheral nerve injury activates spinal microglia. This leads to enduring changes in the properties of dorsal horn neurons that initiate central sensitization and the onset of neuropathic pain. Although a variety of neuropeptides, cytokines, chemokines and neurotransmitters have been implicated at various points in this process, it is possible that much of the information transfer between activated microglia and neurons, at least in this context, may be explicable in terms of the actions of brain derived neurotrophic factor (BDNF). Microglial-derived BDNF mediates central sensitization in lamina I by attenuating inhibitory synaptic transmission. This involves an alteration in the chloride equilibrium potential as a result of down regulation of the potassium-chloride exporter, KCC2. In lamina II, BDNF duplicates many aspects of the effects of chronic constriction injury (CCI) of the sciatic nerve on excitatory transmission. It mediates an increase in synaptic drive to putative excitatory neurons whilst reducing that to inhibitory neurons. CCI produces a specific pattern of changes in excitatory synaptic transmission to tonic, delay, phasic, transient and irregular neurons. A very similar 'injury footprint' is seen following long-term exposure to BDNF. This review presents new information on the action of BDNF and CCI on lamina II neurons, including the similarity of their actions on the kinetics and distributions of subpopulations of miniature excitatory postsynaptic currents (mEPSC). These findings raise the possibility that BDNF functions as a final common path for a convergence of perturbations that culminate in the generation of neuropathic pain.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Microglia/physiology , Peripheral Nerve Injuries , Posterior Horn Cells/physiology , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Constriction , Microglia/drug effects , Peripheral Nerves/drug effects , Peripheral Nerves/physiology , Posterior Horn Cells/drug effects , Symporters/metabolism , Synaptic Transmission , K Cl- Cotransporters
12.
Front Neurosci ; 14: 615362, 2020.
Article in English | MEDLINE | ID: mdl-33424545

ABSTRACT

Interoceptive and exteroceptive signals, and the corresponding coordinated control of internal organs and sensory functions, including pain, are received and orchestrated by multiple neurons within the peripheral, central and autonomic nervous systems. A central aim of the present report is to obtain a molecularly informed basis for analgesic drug development aimed at peripheral rather than central targets. We compare three key peripheral ganglia: nodose, sympathetic (superior cervical), and dorsal root ganglia in the rat, and focus on their molecular composition using next-gen RNA-Seq, as well as their neuroanatomy using immunocytochemistry and in situ hybridization. We obtained quantitative and anatomical assessments of transmitters, receptors, enzymes and signaling pathways mediating ganglion-specific functions. Distinct ganglionic patterns of expression were observed spanning ion channels, neurotransmitters, neuropeptides, G-protein coupled receptors (GPCRs), transporters, and biosynthetic enzymes. The relationship between ganglionic transcript levels and the corresponding protein was examined using immunohistochemistry for select, highly expressed, ganglion-specific genes. Transcriptomic analyses of spinal dorsal horn and intermediolateral cell column (IML), which form the termination of primary afferent neurons and the origin of preganglionic innervation to the SCG, respectively, disclosed pre- and post-ganglionic molecular-level circuits. These multimodal investigations provide insight into autonomic regulation, nodose transcripts related to pain and satiety, and DRG-spinal cord and IML-SCG communication. Multiple neurobiological and pharmacological contexts can be addressed, such as discriminating drug targets and predicting potential side effects, in analgesic drug development efforts directed at the peripheral nervous system.

13.
Cell Rep ; 31(13): 107833, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32610134

ABSTRACT

Glucagon-like peptide-1 (GLP-1) from intestinal L-cells stimulates insulin secretion and reduces appetite after food ingestion, and it is the basis for drugs against type-2 diabetes and obesity. Drugs targeting L- and other enteroendocrine cells are under development, with the aim to mimic endocrine effects of gastric bypass surgery, but they are difficult to develop without human L-cell models. Human ileal organoids, engineered by CRISPR-Cas9, express the fluorescent protein Venus in the proglucagon locus, enabling maintenance of live, identifiable human L-cells in culture. Fluorescence-activated cell sorting (FACS)-purified organoid-derived L-cells, analyzed by RNA sequencing (RNA-seq), express hormones, receptors, and ion channels, largely typical of their murine counterparts. L-cells are electrically active and exhibit membrane depolarization and calcium elevations in response to G-protein-coupled receptor ligands. Organoids secrete hormones in response to glucose and other stimuli. The ability to label and maintain human L-cells in organoid culture opens avenues to explore L-cell function and develop drugs targeting the human enteroendocrine system.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Ileum/cytology , Organoids/cytology , Staining and Labeling , Animals , Cells, Cultured , Electrophysiological Phenomena , Glucose/metabolism , Humans , L Cells , Mice , Peptides/metabolism
14.
Cell Rep ; 32(3): 107941, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32697988

ABSTRACT

By studying healthy women who do not request analgesia during their first delivery, we investigate genetic effects on labor pain. Such women have normal sensory and psychometric test results, except for significantly higher cuff pressure pain. We find an excess of heterozygotes carrying the rare allele of SNP rs140124801 in KCNG4. The rare variant KV6.4-Met419 has a dominant-negative effect and cannot modulate the voltage dependence of KV2.1 inactivation because it fails to traffic to the plasma membrane. InĀ vivo, Kcng4 (KV6.4) expression occurs in 40% of retrograde-labeled mouse uterine sensory neurons, all of which express KV2.1, and over 90% express the nociceptor genes Trpv1 and Scn10a. In neurons overexpressing KV6.4-Met419, the voltage dependence of inactivation for KV2.1 is more depolarized compared with neurons overexpressing KV6.4. Finally, KV6.4-Met419-overexpressing neurons have a higher action potential threshold. We conclude that KV6.4 can influence human labor pain by modulating the excitability of uterine nociceptors.


Subject(s)
Labor Pain/metabolism , Potassium Channels, Voltage-Gated/metabolism , Protein Subunits/metabolism , Adult , Alleles , Amino Acid Sequence , Analgesics/pharmacology , Animals , Base Sequence , Cell Membrane/metabolism , Cognition , Cohort Studies , Emotions , Female , Ganglia, Spinal/metabolism , Heterozygote , Humans , Ion Channel Gating/genetics , Labor Pain/genetics , Labor Pain/physiopathology , Male , Mice, Inbred C57BL , Models, Biological , Mutation/genetics , Nociceptors/metabolism , Pain Threshold , Polymorphism, Single Nucleotide/genetics , Potassium Channels, Voltage-Gated/chemistry , Potassium Channels, Voltage-Gated/genetics , Pregnancy , Protein Multimerization , Sensory Receptor Cells/metabolism , Shab Potassium Channels/metabolism , Subcellular Fractions/metabolism , Uterus/innervation
15.
Mol Metab ; 35: 100954, 2020 05.
Article in English | MEDLINE | ID: mdl-32244182

ABSTRACT

OBJECTIVE: Non-alcoholic steatohepatitis (NASH) is characterized by a robust pro-inflammatory component at both hepatic and systemic levels together with a disease-specific gut microbiome signature. Protein tyrosine phosphatase 1Ā B (PTP1B) plays distinct roles in non-immune and immune cells, in the latter inhibiting pro-inflammatory signaling cascades. In this study, we have explored the role of PTP1B in the composition of gut microbiota and gut barrier dynamics in methionine and choline-deficient (MCD) diet-induced NASH in mice. METHODS: Gut features and barrier permeability were characterized in wild-type (PTP1B WT) and PTP1B-deficient knockout (PTP1B KO) mice fed a chow or methionine/choline-deficient (MCD) diet for 4 weeks. The impact of inflammation was studied in intestinal epithelial and enteroendocrine cells. The secretion of GLP-1 was evaluated in primary colonic cultures and plasma of mice. RESULTS: We found that a shift in the gut microbiota shape, disruption of gut barrier function, higher levels of serum bile acids, and decreased circulating glucagon-like peptide (GLP)-1 are features during NASH. Surprisingly, despite the pro-inflammatory phenotype of global PTP1B-deficient mice, they were partly protected against the alterations in gut microbiota composition during NASH and presented better gut barrier integrity and less permeability under this pathological condition. These effects concurred with higher colonic mucosal inflammation, decreased serum bile acids, and protection against the decrease in circulating GLP-1 levels during NASH compared with their WT counterparts together with increased expression of GLP-2-sensitive genes in the gut. At the molecular level, stimulation of enteroendocrine STC-1 cells with a pro-inflammatory conditioned medium (CM) from lipopolysaccharide (LPS)-stimulated macrophages triggered pro-inflammatory signaling cascades that were further exacerbated by a PTP1B inhibitor. Likewise, the pro-inflammatory CM induced GLP-1 secretion in primary colonic cultures, an effect augmented by PTP1B inhibition. CONCLUSION: Altogether our results have unraveled a potential role of PTP1B in the gut-liver axis during NASH, likely mediated by increased sensitivity to GLPs, with potential therapeutic value.


Subject(s)
Gastrointestinal Microbiome/genetics , Intestinal Mucosa/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Animals , Choline Deficiency/complications , Diet/adverse effects , Disease Models, Animal , Gene Expression , Gene Knockout Techniques , Glucagon-Like Peptide 1/blood , Inflammation/metabolism , Liver/metabolism , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Permeability , RAW 264.7 Cells
16.
J Physiol ; 587(Pt 5): 1013-32, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19124536

ABSTRACT

Peripheral nerve injury can promote neuropathic pain. The basis of the 'central sensitization' that underlies this often intractable condition was investigated using 14-20-day chronic constriction injury (CCI) of the sciatic nerve of 20-day-old rats followed by electrophysiological analysis of acutely isolated spinal cord slices. In addition, defined-medium organotypic spinal cord slice cultures were exposed for 5-6 days to brain-derived neurotrophic factor (BDNF, 200 ng ml(-1)) or to medium conditioned with activated microglia (aMCM). Since microglial activation is an early consequence of CCI, the latter manipulation allowed us to model the effect of peripheral nerve injury on the dorsal horn in vitro. Using whole-cell recording from superficial dorsal horn neurons, we found that both BDNF and CCI increased excitatory synaptic drive to putative excitatory 'radial delay' neurons and decreased synaptic excitation of inhibitory 'tonic islet/central' neurons. BDNF also attenuated synaptic excitation of putative GABAergic neurons identified by glutamic acid decarboxylase (GAD) immunoreactivity. Intrinsic neuronal properties (rheobase, input resistance and action potential discharge rates) were unaffected. Exposure of organotypic cultures to either BDNF or aMCM increased overall excitability of the dorsal horn, as seen by increased cytoplasmic Ca(2+) responses to 35 mm K(+) as monitored by confocal Fluo-4AM imaging. The effect of aMCM was attenuated by the recombinant BDNF binding protein TrkBd5 and the effect of BDNF persisted when GABAergic inhibition was blocked with SR95531. These findings suggest that CCI enhances excitatory synaptic drive to excitatory neurons but decreases that to inhibitory neurons. Both effects are mediated by nerve injury-induced release of BDNF from microglia.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Excitatory Postsynaptic Potentials/physiology , Posterior Horn Cells/physiology , Sciatic Neuropathy/physiopathology , Animals , Male , Organ Culture Techniques , Posterior Horn Cells/drug effects , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Time Factors
17.
Nat Commun ; 10(1): 1029, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30833673

ABSTRACT

Enteroendocrine cells are specialised sensory cells located in the intestinal epithelium and generate signals in response to food ingestion. Whilst traditionally considered hormone-producing cells, there is evidence that they also initiate activity in the afferent vagus nerve and thereby signal directly to the brainstem. We investigate whether enteroendocrine L-cells, well known for their production of the incretin hormone glucagon-like peptide-1 (GLP-1), also release other neuro-transmitters/modulators. We demonstrate regulated ATP release by ATP measurements in cell supernatants and by using sniffer patches that generate electrical currents upon ATP exposure. Employing purinergic receptor antagonists, we demonstrate that evoked ATP release from L-cells triggers electrical responses in neighbouring enterocytes through P2Y2 and nodose ganglion neurones in co-cultures through P2X2/3-receptors. We conclude that L-cells co-secrete ATP together with GLP-1 and PYY, and that ATP acts as an additional signal triggering vagal activation and potentially synergising with the actions of locally elevated peptide hormone concentrations.


Subject(s)
Adenosine Triphosphate/metabolism , Enterocytes/metabolism , Glucagon-Like Peptide 1/metabolism , Intestines , Neurons, Afferent/metabolism , Afferent Pathways , Animals , Cell Line , Eating , Enteroendocrine Cells/metabolism , Female , Ganglion Cysts/metabolism , Ganglion Cysts/pathology , Incretins/metabolism , Intestinal Mucosa/innervation , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Nodose Ganglion/metabolism , Nodose Ganglion/pathology , Peptide YY/metabolism , Receptors, Purinergic P2X2/metabolism , Receptors, Purinergic P2X3/metabolism , Vagus Nerve/metabolism
18.
Mol Pain ; 4: 63, 2008 Dec 17.
Article in English | MEDLINE | ID: mdl-19091115

ABSTRACT

BACKGROUND: Cytokines such as interleukin 1beta (IL-1beta) have been implicated in the development of central sensitization that is characteristic of neuropathic pain. To examine its long-term effect on nociceptive processing, defined medium organotypic cultures of rat spinal cord were exposed to 100 pM IL-1beta for 6-8 d. Interleukin effects in the dorsal horn were examined by whole-cell patch-clamp recording and Ca(2+) imaging techniques. RESULTS: Examination of the cultures with confocal Fluo-4 AM imaging showed that IL-1beta increased the change in intracellular Ca(2+) produced by exposure to 35-50 mM K+. This is consistent with a modest increase in overall dorsal horn excitability. Despite this, IL-1beta did not have a direct effect on rheobase or resting membrane potential nor did it selectively destroy any specific neuronal population. All effects were instead confined to changes in synaptic transmission. A variety of pre- and postsynaptic actions of IL-1beta were seen in five different electrophysiologically-defined neuronal phenotypes. In putative excitatory 'delay' neurons, cytokine treatment increased the amplitude of spontaneous EPSC's (sEPSC) and decreased the frequency of spontaneous IPSC's (sIPSC). These effects would be expected to increase dorsal horn excitability and to facilitate the transfer of nociceptive information. However, other actions of IL-1beta included disinhibition of putative inhibitory 'tonic' neurons and an increase in the amplitude of sIPSC's in 'delay' neurons. CONCLUSION: Since spinal microglial activation peaks between 3 and 7 days after the initiation of chronic peripheral nerve injury and these cells release IL-1beta at this time, our findings define some of the neurophysiological mechanisms whereby nerve-injury induced release of IL-1beta may contribute to the central sensitization associated with chronic neuropathic pain.


Subject(s)
Interleukin-1beta/pharmacology , Neurons/drug effects , Posterior Horn Cells/drug effects , Action Potentials , Animals , Calcium/metabolism , Interleukin-1beta/metabolism , Neurons/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Posterior Horn Cells/embryology , Posterior Horn Cells/metabolism , Rats , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/embryology , Spinal Cord/metabolism , Synaptic Transmission
20.
Endocrinology ; 159(7): 2826-2835, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29688303

ABSTRACT

Free fatty acid receptors (FFAs) are highly enriched in enteroendocrine cells providing pathways to link dietary fats and microbially generated short-chain fatty acids (SCFAs) to the secretion of a variety of gut hormones. FFA1 and FFA4 are receptors for long-chain fatty acids that have been linked to the elevation of plasma gut hormones after fat ingestion. FFA2 and FFA3 are receptors for SCFA, which are generated at high concentrations by microbial fermentation of dietary fiber and have also been implicated in enhancement of gut hormone secretion. FFAs are candidate drug targets for increasing the secretion of intestinal hormones such as glucagon-like peptide-1 and peptide YY as potential new treatments for type 2 diabetes and obesity. This review will examine aspects of intestinal physiology and pharmacology related to the function of FFAs in enteroendocrine cells.


Subject(s)
Enteroendocrine Cells/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Glucagon-Like Peptide 1/metabolism , Humans , Peptide YY/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL