Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37662229

ABSTRACT

Dynamic interactions between gut mucosal cells and the external environment are essential to maintain gut homeostasis. Enterochromaffin (EC) cells transduce both chemical and mechanical signals and produce 5-hydroxytryptamine (5-HT) to mediate disparate physiological responses. However, the molecular and cellular basis for functional diversity of ECs remains to be adequately defined. Here, we integrated single-cell transcriptomics with spatial image analysis to identify fourteen EC clusters that are topographically organized along the gut. Subtypes predicted to be sensitive to the chemical environment and mechanical forces were identified that express distinct transcription factors and hormones. A Piezo2+ population in the distal colon was endowed with a distinctive neuronal signature. Using a combination of genetic, chemogenetic and pharmacological approaches, we demonstrated Piezo2+ ECs are required for normal colon motility. Our study constructs a molecular map for ECs and offers a framework for deconvoluting EC cells with pleiotropic functions.

2.
Elife ; 92020 07 15.
Article in English | MEDLINE | ID: mdl-32667883

ABSTRACT

Breast cancer is the most common type of cancer in women and notwithstanding important therapeutic advances, remains the second leading cause of cancer-related death. Despite extensive research relating to the hormone ghrelin, responsible for the stimulation of growth hormone release and appetite, little is known of the effects of its unacylated form, especially in cancer. The present study aimed to characterize effects of unacylated ghrelin on breast cancer cells, define its mechanism of action, and explore the therapeutic potential of unacylated ghrelin or analog AZP-531. We report potent anti-tumor effects of unacylated ghrelin, dependent on cells being cultured in 3D in a biologically-relevant extracellular matrix. The mechanism of unacylated ghrelin-mediated growth inhibition involves activation of Gαi and suppression of MAPK signaling. AZP-531 also suppresses the growth of breast cancer cells in vitro and in xenografts, and may be a novel approach for the safe and effective treatment of breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Ghrelin/pharmacology , Peptide Fragments/pharmacology , Peptides, Cyclic/pharmacology , Spheroids, Cellular/drug effects , Acylation , Animals , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cell Culture Techniques , Cell Line, Tumor , Female , Ghrelin/chemistry , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Xenograft Model Antitumor Assays
3.
Cell Tissue Res ; 379(1): 109-120, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31478137

ABSTRACT

Recent studies reveal substantial species and regional differences in enteroendocrine cell (EEC) populations, including differences in patterns of hormone coexpression, which limit extrapolation between animal models and human. In this study, jejunal samples, with no histologically identifiable pathology, from patients undergoing Whipple's procedure were investigated for the presence of gastrointestinal hormones using double- and triple-labelling immunohistochemistry and high-resolution confocal microscopy. Ten hormones (5-HT, CCK, secretin, proglucagon-derived peptides, PYY, GIP, somatostatin, neurotensin, ghrelin and motilin) were localised in EEC of the human jejunum. If only single staining is considered, the most numerous EEC were those containing 5-HT, CCK, ghrelin, GIP, motilin, secretin and proglucagon-derived peptides. All hormones had some degree of colocalisation with other hormones. This included a population of EEC in which GIP, CCK and proglucagon-derived peptides are costored, and four 5-HT cell populations, 5-HT/GIP, 5-HT/ghrelin, 5-HT/PYY, and 5-HT/secretin cell groups, and a high degree of overlap between motilin and ghrelin. The presence of 5-HT in many secretin cells is consistent across species, whereas lack of 5-HT and CCK colocalisation distinguishes human from mouse. It seems likely that the different subclasses of 5-HT cells subserve different roles. At a subcellular level, we examined the vesicular localisation of secretin and 5-HT, and found these to be separately stored. We conclude that hormone-containing cells in the human jejunum do not comply with a one-cell, one-hormone classification and that colocalisations of hormones are likely to define subtypes of EEC that have different roles.


Subject(s)
Enteroendocrine Cells/metabolism , Jejunum/cytology , Adult , Aged , Aged, 80 and over , Cell Count , Female , Gastrointestinal Hormones/metabolism , Humans , Jejunum/metabolism , Male , Serotonin/metabolism
4.
Cell Tissue Res ; 376(1): 37-49, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30467709

ABSTRACT

Gastric endocrine cell hormones contribute to the control of the stomach and to signalling to the brain. In other gut regions, enteroendocrine cells (EECs) exhibit extensive patterns of colocalisation of hormones. In the current study, we characterise EECs in the human gastric fundus and corpus. We utilise immunohistochemistry to investigate EECs with antibodies to ghrelin, serotonin (5-HT), somatostatin, peptide YY (PYY), glucagon-like peptide 1, calbindin, gastrin and pancreastatin, the latter as a marker of enterochromaffin-like (ECL) cells. EECs were mainly located in regions of the gastric glands populated by parietal cells. Gastrin cells were absent and PYY cells were very rare. Except for about 25% of 5-HT cells being a subpopulation of ECL cells marked by pancreastatin, colocalisation of hormones in gastric EECs was infrequent. Ghrelin cells were distributed throughout the fundus and corpus; most were basally located in the glands, often very close to parietal cells and were closed cells i.e., not in contact with the lumen. A small proportion had long processes located close to the base of the mucosal epithelium. The 5-HT cells were of at least three types: small, round, closed cells; cells with multiple, often very long, processes; and a subgroup of ECL cells. Processes were in contact with their surrounding cells, including parietal cells. Mast cells had very weak or no 5-HT immunoreactivity. Somatostatin cells were a closed type with long processes. In conclusion, four major chemically defined EEC types occurred in the human oxyntic mucosa. Within each group were cells with distinct morphologies and relationships to other mucosal cells.


Subject(s)
Enteroendocrine Cells , Gastric Fundus , Gastrointestinal Hormones/analysis , Enteroendocrine Cells/chemistry , Enteroendocrine Cells/cytology , Female , Gastric Fundus/cytology , Gastric Fundus/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Obesity/surgery
5.
ACS Chem Biol ; 13(6): 1577-1587, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29746088

ABSTRACT

α-Conotoxins are disulfide-bonded peptides from cone snail venoms and are characterized by their affinity for nicotinic acetylcholine receptors (nAChR). Several α-conotoxins with distinct selectivity for nAChR subtypes have been identified as potent analgesics in animal models of chronic pain. However, a number of α-conotoxins have been shown to inhibit N-type calcium channel currents in rodent dissociated dorsal root ganglion (DRG) neurons via activation of G protein-coupled GABAB receptors (GABABR). Therefore, it is unclear whether activation of GABABR or inhibition of α9α10 nAChRs is the analgesic mechanism. To investigate the mechanisms by which α-conotoxins provide analgesia, we synthesized a suite of Vc1.1 analogues where all residues, except the conserved cysteines, in Vc1.1 were individually replaced by alanine (A), lysine (K), and aspartic acid (D). Our results show that the amino acids in the first loop play an important role in binding of the peptide to the receptor, whereas those in the second loop play an important role for the selectivity of the peptide for the GABABR over α9α10 nAChRs. We designed a cVc1.1 analogue that is >8000-fold selective for GABABR-mediated inhibition of high voltage-activated (HVA) calcium channels over α9α10 nAChRs and show that it is analgesic in a mouse model of chronic visceral hypersensitivity (CVH). cVc1.1[D11A,E14A] caused dose-dependent inhibition of colonic nociceptors with greater efficacy in ex vivo CVH colonic nociceptors relative to healthy colonic nociceptors. These findings suggest that selectively targeting GABABR-mediated HVA calcium channel inhibition by α-conotoxins could be effective for the treatment of chronic visceral pain.


Subject(s)
Analgesics/therapeutic use , Calcium Channel Blockers/therapeutic use , Conotoxins/therapeutic use , Pain/drug therapy , Analgesics/chemical synthesis , Analgesics/chemistry , Animals , Calcium Channel Blockers/chemical synthesis , Calcium Channel Blockers/chemistry , Calcium Channels, N-Type/metabolism , Conotoxins/chemical synthesis , Conotoxins/chemistry , Male , Mice, Inbred C57BL , Molecular Structure , Nicotinic Antagonists/chemical synthesis , Nicotinic Antagonists/chemistry , Nicotinic Antagonists/therapeutic use , Rats, Wistar , Receptors, GABA-B/metabolism , Receptors, Nicotinic/metabolism , Structure-Activity Relationship , Xenopus laevis
6.
Spinal Cord ; 56(3): 199-205, 2018 03.
Article in English | MEDLINE | ID: mdl-29142293

ABSTRACT

STUDY DESIGN: Narrative review. OBJECTIVES: The purpose is to review the organisation of the nerve pathways that control defecation and to relate this knowledge to the deficits in colorectal function after SCI. METHODS: A literature review was conducted to identify salient features of defecation control pathways and the functional consequences of damage to these pathways in SCI. RESULTS: The control pathways for defecation have separate pontine centres under cortical control that influence defecation. The pontine centres connect, separately, with autonomic preganglionic neurons of the spinal defecation centres and somatic motor neurons of Onuf's nucleus in the sacral spinal cord. Organised propulsive motor patterns can be generated by stimulation of the spinal defecation centres. Activation of the somatic neurons contracts the external sphincter. The analysis aids in interpreting the consequences of SCI and predicts therapeutic strategies. CONCLUSIONS: Analysis of the bowel control circuits identifies sites at which bowel function may be modulated after SCI. Colokinetic drugs that elicit propulsive contractions of the colorectum may provide valuable augmentation of non-pharmacological bowel management procedures.


Subject(s)
Colonic Diseases , Disease Management , Neural Pathways/physiopathology , Spinal Cord Injuries/complications , Colonic Diseases/etiology , Colonic Diseases/pathology , Colonic Diseases/therapy , Female , Humans , Internet , Male , Pons/physiopathology , PubMed
7.
Physiol Rep ; 5(15)2017 Aug.
Article in English | MEDLINE | ID: mdl-28801520

ABSTRACT

In laboratory animals and in human, centrally penetrant ghrelin receptor agonists, given systemically or orally, cause defecation. Animal studies show that the effect is due to activation of ghrelin receptors in the spinal lumbosacral defecation centers. However, it is not known whether there is a physiological role of ghrelin or the ghrelin receptor in the control of defecation. Using immunohistochemistry and immunoassay, we detected and measured ghrelin in the stomach, but were unable to detect ghrelin by either method in the lumbosacral spinal cord, or other regions of the CNS In rats in which the thoracic spinal cord was transected 5 weeks before, the effects of a ghrelin agonist on colorectal propulsion were significantly enhanced, but defecation caused by water avoidance stress (WAS) was reduced. In knockout rats that expressed no ghrelin and in wild-type rats, WAS-induced defecation was reduced by a ghrelin receptor antagonist, to similar extents. We conclude that the ghrelin receptors of the lumbosacral defecation centers have a physiological role in the control of defecation, but that their role is not dependent on ghrelin. This implies that a transmitter other than ghrelin engages the ghrelin receptor or a ghrelin receptor complex.


Subject(s)
Defecation , Ghrelin/physiology , Receptors, Ghrelin/physiology , Spinal Cord/physiology , Animals , Brain/metabolism , Gastric Mucosa/metabolism , Gastrointestinal Motility/drug effects , Gene Knockout Techniques , Ghrelin/genetics , Ghrelin/metabolism , Lumbosacral Region , Male , Piperidines/administration & dosage , Quinazolinones/administration & dosage , Rats, Sprague-Dawley , Receptors, Ghrelin/agonists , Spinal Cord/metabolism
8.
Endocrinology ; 158(7): 2113-2123, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28430903

ABSTRACT

Recent studies reveal complex patterns of hormone coexpression within enteroendocrine cells (EECs), contrary to the traditional view that gut hormones are expressed individually in EECs. Moreover, different hormones have been found in separate subcellular vesicles. However, detailed analysis of relative expression of multiple hormones has not been made. Subcellular studies have been confined to peptide hormones, and have not included the indolamine 5-hydroxytryptamine (5-HT) or the neuroendocrine protein chromogranin A (CgA). In the present work, coexpression of 5-HT, CgA, secretin, cholecystokinin (CCK), ghrelin, and glucagonlike peptide (GLP)-1 in mouse duodenum was quantified at a cellular and subcellular level by semiautomated cell counting and quantitative vesicle measurements. We investigated whether relative numbers of cells with colocalized hormones analyzed at a cell level matched the numbers revealed by examination of individual storage vesicles within cells. CgA and 5-HT were frequently expressed in EECs that contained combinations of GLP-1, ghrelin, secretin, and CCK. Separate subcellular stores of 5-HT, CgA, secretin, CCK, ghrelin, and GLP-1 were identified. In some cases, high-resolution analysis revealed small numbers of immunoreactive vesicles in cells dominated by a different hormone. Thus the observed incidence of cells with colocalized hormones is greater when analyzed at a subcellular, compared with a cellular, level. Subcellular analysis also showed that relative numbers of vesicles differ considerably between cells. Thus separate packaging of hormones that are colocalized is a general feature of EECs, and EECs exhibit substantial heterogeneity, including the colocalization of hormones that were formerly thought to be in cells of different lineages.


Subject(s)
Cytoplasmic Vesicles/metabolism , Enteroendocrine Cells/cytology , Enteroendocrine Cells/metabolism , Gastrointestinal Hormones/metabolism , Animals , Cholecystokinin/metabolism , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Male , Mice , Mice, Inbred C57BL , Peptide YY/metabolism , Protein Transport , Serotonin/metabolism , Subcellular Fractions , Tissue Distribution
9.
Cell Tissue Res ; 369(2): 245-253, 2017 08.
Article in English | MEDLINE | ID: mdl-28413860

ABSTRACT

There is general consensus that enteroendocrine cells, EEC, containing the enteric hormone cholecystokinin (CCK) are confined to the small intestine and predominate in the duodenum and jejunum. Contrary to this, EEC that express the gene for CCK have been isolated from the large intestine of the mouse and there is evidence for EEC that contain CCK-like immunoreactivity in the mouse colon. However, the human and rat colons do not contain CCK cells. In the current study, we use immunohistochemistry to investigate CCK peptide presence in endocrine cells, PCR to identify cck transcripts and chromatography to identify CCK peptide forms in the mouse small and large intestine. The colocalisation of CCK and 5-HT, hormones that have been hypothesised to derive from cells of different lineages, was also investigated. CCK immunoreactivity was found in EEC throughout the mouse small and large intestine but positive cells were rare in the rectum. Immunoreactive EEC were as common in the caecum and proximal colon as they were in the duodenum and jejunum. CCK gene transcripts were found in the mucosa throughout the intestine but mRNA for gastrin, a hormone that can bind some anti-CCK antibodies, was only found in the stomach and duodenum. Characterisation of CCK peptides of the colon by extraction, chromatographic separation and radioimmunoassay revealed bioactive amidated and sulphated forms, including CCK-8 and CCK-33. Moreover, CCK-containing EEC in the large intestine bound antibodies that target the biologically active sulfated form. Colocalisation of CCK and 5-HT occurred in a proportion of EEC throughout the small intestine and in the caecum but these hormones were not colocalised in the colon, where there was CCK and PYY colocalisation. It is concluded that authentic, biologically active, CCK occurs in EEC of the mouse large intestine.


Subject(s)
Cholecystokinin/metabolism , Enteroendocrine Cells/metabolism , Intestine, Large/cytology , Intestine, Small/cytology , Animals , Cell Count , Cholecystokinin/genetics , Enteroendocrine Cells/cytology , Gastrins/genetics , Gastrins/metabolism , Male , Mice, Inbred C57BL , Peptide YY/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Serotonin/metabolism
10.
Handb Exp Pharmacol ; 239: 379-416, 2017.
Article in English | MEDLINE | ID: mdl-28035532

ABSTRACT

Ghrelin and motilin are released from gastrointestinal endocrine cells during hunger, to act through G protein-coupled receptors that have closely related amino acid sequences. The actions of ghrelin are more complex than motilin because ghrelin also exists outside the GI tract, it is processed to des-acyl ghrelin which has activity, ghrelin can exist in truncated forms and retain activity, the ghrelin receptor can have constitutive activity and is subject to biased agonism and finally additional ghrelin-like and des-acyl ghrelin receptors are proposed. Both ghrelin and motilin can stimulate gastric emptying, acting via different pathways, perhaps influenced by biased agonism at the receptors, but research is revealing additional pathways of activity. For example, it is becoming apparent that reduction of nausea may be a key therapeutic target for ghrelin receptor agonists and perhaps for compounds that modulate the constitutive activity of the ghrelin receptor. Reduction of nausea may be the mechanism through which gastroparesis symptoms are reduced. Intriguingly, a potential ability of motilin to influence nausea is also becoming apparent. Ghrelin interacts with digestive function through its effects on appetite, and ghrelin antagonists may have a place in treating Prader-Willi syndrome. Unlike motilin, ghrelin receptor agonists also have the potential to treat constipation by acting at the lumbosacral defecation centres. In conclusion, agonists of both ghrelin and motilin receptors hold potential as treatments for specific subsets of digestive system disorders.


Subject(s)
Gastrointestinal Diseases/metabolism , Gastrointestinal Motility , Gastrointestinal Tract/metabolism , Ghrelin/metabolism , Motilin/metabolism , Signal Transduction , Animals , Appetite Regulation , Enteric Nervous System/metabolism , Enteric Nervous System/physiopathology , Gastrointestinal Agents/therapeutic use , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/physiopathology , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiopathology , Humans , Neural Pathways/metabolism , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Ghrelin/agonists , Receptors, Ghrelin/metabolism , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/metabolism , Signal Transduction/drug effects
11.
Cell Tissue Res ; 367(2): 161-168, 2017 02.
Article in English | MEDLINE | ID: mdl-27844204

ABSTRACT

Recent studies have shown that patterns of colocalisation of hormones in enteroendocrine cells are more complex than previously appreciated and that the patterns differ substantially between species. In this study, the human sigmoid colon is investigated by immunohistochemistry for the presence of gastrointestinal hormones and their colocalisation. The segments of colon were distant from the pathology that led to colectomy and appeared structurally normal. Only four hormones, 5-hydroxytryptamine (5-HT), glucagon-like peptide 1 (GLP-1), peptide YY (PYY) and somatostatin, were common in enteroendocrine cells of the human colon. Cholecystokinin, present in the colon of some species, was absent, as were glucose-dependent insulinotropic peptide, ghrelin and motilin. Neurotensin cells were extremely rare. The most numerous cells were 5-HT cells, some of which also contained PYY or somatostatin and very rarely GLP-1. Almost all GLP-1 cells contained PYY. It is concluded that enteroendocrine cells of the human colon, like those of other regions and species, exhibit overlapping patterns of hormone colocalisation and that the hormones and their patterns of expression differ between human and other species.


Subject(s)
Colon/cytology , Enteroendocrine Cells/cytology , Cell Count , Hormones/metabolism , Humans , Jejunum/cytology , Staining and Labeling
12.
Nutrients ; 8(10)2016 Oct 10.
Article in English | MEDLINE | ID: mdl-27735854

ABSTRACT

TRPA1 is a ligand-activated cation channel found in the intestine and other tissues. Components of food that stimulate TRPA1 receptors (phytonutrients) include allyl isothiocyanate, cinnamaldehyde and linalool, but these may also act at other receptors. Cells lining the intestinal mucosa are immunoreactive for TRPA1 and Trpa1 mRNA occurs in mucosal extracts, suggesting that the TRPA1 receptor is the target for these agonists. However, in situ hybridisation reveals Trpa1 expression in 5-HT containing enteroendocrine cells, not enterocytes. TRPA1 agonists evoke mucosal secretion, which may be indirect (through release of 5-HT) or direct by activation of enterocytes. We investigated effects of the phytonutrients on transmucosal ion currents in mouse duodenum and colon, and the specificity of the phytonutrients in cells transfected with Trpa1, and in Trpa1-deficient mice. The phytonutrients increased currents in the duodenum with the relative potencies: allyl isothiocyanate (AITC) > cinnamaldehyde > linalool (0.1 to 300 µM). The rank order was similar in the colon, but linalool was ineffective. Responses to AITC were reduced by the TRPA1 antagonist HC-030031 (100 µM), and were greatly diminished in Trpa1-/- duodenum and colon. Responses were not reduced by tetrodotoxin, 5-HT receptor antagonists, or atropine, but inhibition of prostaglandin synthesis reduced responses. Thus, functional TRPA1 channels are expressed by enterocytes of the duodenum and colon. Activation of enterocyte TRPA1 by food components has the potential to facilitate nutrient absorption.


Subject(s)
Intestinal Mucosa/physiology , Phytochemicals/pharmacology , Transient Receptor Potential Channels/drug effects , Acrolein/analogs & derivatives , Acrolein/pharmacology , Acyclic Monoterpenes , Animals , Calcium/metabolism , Colon/physiology , Duodenum/physiology , Electrophysiological Phenomena , Enterocytes/drug effects , Enterocytes/metabolism , Food , Gene Expression , HEK293 Cells , Humans , Intestinal Mucosa/chemistry , Isothiocyanates/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monoterpenes/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , TRPA1 Cation Channel , Transfection , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/genetics
13.
Cell Tissue Res ; 364(3): 489-497, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26803512

ABSTRACT

The majority of 5-HT (serotonin) in the body is contained in enteroendocrine cells of the gastrointestinal mucosa. From the time of their discovery over 80 years ago, the 5-HT-containing cells have been regarded as a class of cell that is distinct from enteroendocrine cells that contain peptide hormones. However, recent studies have cast doubt on the concept of there being distinct classes of enteroendocrine cells, each containing a single hormone or occasionally more than one hormone. Instead, data are rapidly accumulating that there are complex patterns of colocalisation of hormones that identify multiple subclasses of enteroendocrine cells. In the present work, multiple labelling immunohistochemistry is used to investigate patterns of colocalisation of 5-HT with enteric peptide hormones. Over 95 % of 5-HT cells in the duodenum also contained cholecystokinin and about 40 % of them also contained secretin. In the jejunum, about 75 % of 5-HT cells contained cholecystokinin but not secretin and 25 % contained 5-HT plus both cholecystokinin and secretin. Small proportions of 5-HT cells contained gastrin or somatostatin in the stomach, PYY or GLP-1 in the small intestine and GLP-1 or somatostatin in the large intestine. Rare or very rare 5-HT cells contained ghrelin (stomach), neurotensin (small and large intestines), somatostatin (small intestine) and PYY (in the large intestine). It is concluded that 5-HT-containing enteroendocrine cells are heterogeneous in their chemical coding and by implication in their functions.


Subject(s)
Enteroendocrine Cells/metabolism , Gastrointestinal Tract/cytology , Serotonin/metabolism , Animals , Cholecystokinin/metabolism , Gastric Mucosa/metabolism , Gastrins/metabolism , Ghrelin/metabolism , Glucagon-Like Peptide 1/metabolism , Immunohistochemistry , Mice, Inbred C57BL , Neurotensin/metabolism , Peptide YY/metabolism , Secretin/metabolism , Somatostatin/metabolism
14.
Eur J Pharmacol ; 761: 11-8, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25934568

ABSTRACT

Angiotensin II (Ang II) increases sympathetic nerve-evoked contractions of arterial vessels. Here the mechanisms underlying this effect were investigated in mouse tail artery. Isometrically mounted segments of mouse distal tail artery were used to investigate the effects of endothelium denudation, blocking Ca(2+) channels and inhibiting superoxide signalling on Ang II-induced facilitation of nerve-evoked contractions. In addition, in situ amperometry was used to assess effects of Ang II on noradrenaline release. Ang II (0.1-1nM) increased nerve-evoked contractions but did not change noradrenaline release. Losartan (Ang II type 1 receptor antagonist), but not PD 123319 (Ang II type 2 receptor antagonist), blocked the facilitatory effect of Ang II on nerve-evoked contractions. Ang II increased vascular muscle reactivity to phenylephrine and UK-14304 (α1- and α2-adrenoceptor agonists, respectively). Endothelial denudation increased nerve-evoked contractions and reduced the facilitatory effect of Ang II on these responses. Efonidipine (L- and T-type Ca(2+) channel blocker) and NNC 55-0396 (T-type Ca(2+) channel blocker) also attenuated this effect of Ang II, while nifedipine (L-type Ca(2+) channel blocker) did not. Blockers of superoxide generation/signalling did not change the facilitatory effect of Ang II on nerve-evoked contractions. The findings indicate that Ang II increases the contribution of T-type Ca(2+) channels to neural activation of the vascular muscle. In addition, Ang II appears to reduce the inhibitory influence of the endothelium on nerve-evoked contractions.


Subject(s)
Angiotensin II/metabolism , Calcium Channels, T-Type/drug effects , Calcium Signaling/drug effects , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Sympathetic Nervous System/drug effects , Tail/blood supply , Adrenergic alpha-Agonists/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Arteries/drug effects , Arteries/innervation , Arteries/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels, T-Type/metabolism , Dose-Response Relationship, Drug , Electric Stimulation , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , In Vitro Techniques , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/innervation , Muscle, Smooth, Vascular/metabolism , Norepinephrine/metabolism , Receptor, Angiotensin, Type 1/drug effects , Receptor, Angiotensin, Type 1/metabolism , Superoxides/metabolism , Sympathetic Nervous System/metabolism
15.
Eur J Pharmacol ; 752: 34-9, 2015 Apr 05.
Article in English | MEDLINE | ID: mdl-25687251

ABSTRACT

Ulimorelin (TZP101) is a ghrelin receptor agonist that stimulates intestinal motility, but also reduces blood pressure in rodents and humans and dilates blood vessels. It has been proposed as a treatment for intestinal motility disorders. Here we investigated the mechanisms through which ulimorelin affects vascular diameter. Actions of ulimorelin on wall tension of rodent arteries were investigated and compared with other ghrelin receptor agonists. Saphenous, mesenteric and basilar arteries were obtained from Sprague-Dawley rats (male, 8 weeks) and saphenous arteries were obtained from wild type or ghrelin receptor null mice. These were mounted in myography chambers to record artery wall tension. Ulimorelin (0.03-30µM) inhibited phenylephrine-induced contractions of rat saphenous (IC50=0.6µM; Imax=66±5%; n=3-6) and mesenteric arteries (IC50=5µM, Imax=113±16%; n=3-4), but not those contracted by U46619, ET-1 or 60mM [K(+)]. Relaxation of phenylephrine-constricted arteries was not observed with ghrelin receptor agonists TZP102, capromorelin or AZP-531. In rat saphenous and basilar arteries, ulimorelin (10-100µM) and TZP102 (10-100µM) constricted arteries (EC50=9.9µM; Emax=50±7% and EC50=8µM; Emax=99±16% respectively), an effect not attenuated by the ghrelin receptor antagonist YIL 781 3µM or mimicked by capromorelin or AZP-531. In mesenteric arteries, ulimorelin, 1-10µM, caused a surmountable rightward shift in the response to phenylephrine (0.01-1000µM; pA2=5.7; n=3-4). Ulimorelin had similar actions in mouse saphenous artery from both wild type and ghrelin receptor null mice. We conclude that ulimorelin causes vasorelaxation through competitive antagonist action at α1-adrenoceptors and a constrictor action not mediated via the ghrelin receptor.


Subject(s)
Arteries/drug effects , Macrocyclic Compounds/pharmacology , Receptors, Ghrelin/metabolism , Animals , Arteries/physiology , Male , Mice , Rats , Receptors, Ghrelin/agonists , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/genetics , Vasoconstriction/drug effects
16.
Cell Tissue Res ; 359(2): 693-698, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25378285

ABSTRACT

This study has investigated the patterns of colocalisation of the conventional K cell marker, glucagon-like insulinotropic peptide (GIP), and the L cell markers, glucagon like peptide-1 (GLP-1) and peptide YY (PYY), in enteroendocrine cells (EEC) of the small intestine and colon of mouse and pig. All combinations of the hormones, 3 in a cell, 2 in a cell and 1 at a time, were encountered. In both species, the three most common EEC types contained (1) both GLP-1 and PYY but not GIP, (2) GLP-1 alone or (3) GIP plus GLP-1 without PYY. Few GIP plus PYY cells and rare cells containing all 3 hormones were encountered. Gradients of cell types occurred along the intestine. For example, in mouse, there were no PYY cells in the duodenum and few in the jejunum, but >50% of labelled EEC in the distal ileum and colon were PYY immunoreactive. By contrast, over 40% of EEC in the pig duodenum contained PYY, and most also contained either GLP-1 or GIP. The gradient in pig was less pronounced. It is concluded that the traditional classification of K and L cells requires revision, and that there are major inter-species differences in the patterns of colocalisation of hormones that have been used to characterise K and L cells.


Subject(s)
Colon/cytology , Enteroendocrine Cells/cytology , Hormones/metabolism , Intestine, Small/cytology , Animals , Colon/metabolism , Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Intestine, Small/metabolism , Mice, Inbred C57BL , Peptide YY/metabolism , Sus scrofa
17.
J Med Chem ; 57(23): 9933-44, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25393758

ABSTRACT

α-Conotoxin RgIA is both an antagonist of the α9α10 nicotinic acetylcholine receptor (nAChR) subtype and an inhibitor of high-voltage-activated N-type calcium channel currents. RgIA has therapeutic potential for the treatment of pain, but reduction of the disulfide bond framework under physiological conditions represents a potential liability for clinical applications. We synthesized four RgIA analogues that replaced native disulfide pairs with nonreducible dicarba bridges. Solution structures were determined by NMR, activity assessed against biological targets, and stability evaluated in human serum. [3,12]-Dicarba analogues retained inhibition of ACh-evoked currents at α9α10 nAChRs but not N-type calcium channel currents, whereas [2,8]-dicarba analogues displayed the opposite pattern of selectivity. The [2,8]-dicarba RgIA analogues were effective in HEK293 cells stably expressing human Cav2.2 channels and transfected with human GABAB receptors. The analogues also exhibited improved serum stability over the native peptide. These selectively acting dicarba analogues may represent mechanistic probes to explore analgesia-related biological receptors.


Subject(s)
Conotoxins/pharmacology , Receptors, Nicotinic/drug effects , Amino Acid Sequence , Analgesics , Animals , Calcium Channels, N-Type/drug effects , Conotoxins/chemistry , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Rats
18.
PLoS One ; 9(10): e111804, 2014.
Article in English | MEDLINE | ID: mdl-25350563

ABSTRACT

In rat tail artery (RTA), spinal cord injury (SCI) increases nerve-evoked contractions and the contribution of L-type Ca2+ channels to these responses. In RTAs from unoperated rats, these channels play a minor role in contractions and Bay K8644 (L-type channel agonist) mimics the effects of SCI. Here we investigated the mechanisms underlying the facilitatory actions of SCI and Bay K8644 on nerve-evoked contractions of RTAs and the hypothesis that Ca2+ entering via L-type Ca2+ channels is rapidly sequestered by the sarcoplasmic reticulum (SR) limiting its role in contraction. In situ electrochemical detection of noradrenaline was used to assess if Bay K8644 increased noradrenaline release. Perforated patch recordings were used to assess if SCI changed the Ca2+ current recorded in RTA myocytes. Wire myography was used to assess if SCI modified the effects of Bay K8644 and of interrupting SR Ca2+ uptake on nerve-evoked contractions. Bay K8644 did not change noradrenaline-induced oxidation currents. Neither the size nor gating of Ca2+ currents differed between myocytes from sham-operated (control) and SCI rats. Bay K8644 increased nerve-evoked contractions in RTAs from both control and SCI rats, but the magnitude of this effect was reduced by SCI. By contrast, depleting SR Ca2+ stores with ryanodine or cyclopiazonic acid selectively increased nerve-evoked contractions in control RTAs. Cyclopiazonic acid also selectively increased the blockade of these responses by nifedipine (L-type channel blocker) in control RTAs, whereas ryanodine increased the blockade produced by nifedipine in both groups of RTAs. These findings suggest that Ca2+ entering via L-type channels is normally rapidly sequestered limiting its access to the contractile mechanism. Furthermore, the findings suggest SCI reduces the role of this mechanism.


Subject(s)
Calcium/metabolism , Cytoplasm/metabolism , Spinal Cord Injuries/pathology , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/chemistry , Animals , Calcium Channels/drug effects , Calcium Channels, L-Type/metabolism , Electrochemistry , Female , Indoles/chemistry , Muscle Cells/drug effects , Muscle Contraction/drug effects , Nifedipine/chemistry , Norepinephrine/chemistry , Rats , Rats, Sprague-Dawley , Ryanodine/chemistry , Sarcoplasmic Reticulum/metabolism , Tail/blood supply
19.
Pharmacol Rev ; 66(4): 984-1001, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25107984

ABSTRACT

The only molecularly identified ghrelin receptor is the growth hormone secretagogue receptor GHSR1a. Its natural ligand, ghrelin, is an acylated peptide whose unacylated counterpart (UAG) is almost inactive at GHSR1a. A truncated, nonfunctional receptor, GHSR1b, derives from the same gene. We have critically evaluated evidence for effects of ghrelin receptor ligands that are not consistent with actions at GHSR1a. Effects of ghrelin are observed in cells or tissues where the expression of GHSR1a is not detectable or after the Ghsr gene has been inactivated. In several, effects of ghrelin are mimicked by UAG, and ghrelin binding is competitively reduced by UAG. Effects in the absence of GHSR1a and sites at which ghrelin and UAG have similar potency suggest the presence of novel nonspecific ghrelin receptors (ghrelin receptor-like receptors [GRLRs]). A third class of receptor, the UAG receptors, at which UAG, but not ghrelin, is an agonist has been proposed. None of the novel receptors, with the exception of the glycoprotein CD36, which accounts for ghrelin action at a limited number of sites, have been identified. GHSR1a and GHSR1b combine with other G protein-coupled receptors to form heterodimers, whose pharmacologies differ from their components. Thus, it is feasible some GRLRs and some UAG receptors are heterodimers. Effects mediated through GRLRs or UAG receptors include adipocyte lipid accumulation, myoblast differentiation, osteoblast proliferation, insulin release, cardioprotection, coronary artery constriction, vascular endothelial cell proliferation, and tumor cell proliferation. The molecular identification and pharmacologic characterization of novel ghrelin receptors are thus important objectives.


Subject(s)
Ghrelin/analogs & derivatives , Ghrelin/metabolism , Receptors, Ghrelin/metabolism , Animals , Humans , Receptors, Ghrelin/biosynthesis
20.
Breast Cancer Res Treat ; 147(1): 193-201, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25056185

ABSTRACT

Aromatase converts androgens into estrogens and its expression within adipose stromal cells (ASCs) is believed to be the major driver of estrogen-dependent cancers in older women. Ghrelin is a gut-hormone that is involved in the regulation of appetite and known to bind to and activate the cognate ghrelin receptor, GHSR1a. The unacylated form of ghrelin, des-acyl ghrelin, binds weakly to GHSR1a but has been shown to play an important role in regulating a number of physiological processes. The aim of this study was to determine the effect of ghrelin and des-acyl ghrelin on aromatase in primary human ASCs. Primary human ASCs were isolated from adipose tissue of women undergoing cosmetic surgery. Real-time PCR and tritiated water-release assays were performed to examine the effect of treatment on aromatase transcript expression and aromatase activity, respectively. Treatments included ghrelin, des-acyl ghrelin, obestatin, and capromorelin (GHSR1a agonist). GHSR1a protein expression was assessed by Western blot and effects of treatment on Ca(2+) and cAMP second messenger systems were examined using the Flexstation assay and the Lance Ultra cAMP kit, respectively. Results demonstrate that pM concentrations of ghrelin and des-acyl ghrelin inhibit aromatase transcript expression and activity in ASCs under basal conditions and in PGE2-stimulated cells. Moreover, the effects of ghrelin and des-acyl ghrelin are mediated via effects on aromatase promoter PII-specific transcripts. Neither the GHSR1a-specific agonist capromorelin nor obestatin had any effect on aromatase transcript expression or activity. Moreover, GHSR1a protein was undetectable by Western blot and neither ghrelin nor capromorelin elicited a calcium response in ASCs. Finally, ghrelin caused a significant decrease in basal and forskolin-stimulated cAMP in ASC. These findings suggest that ghrelin acts at alternate receptors in ASCs by decreasing intracellular cAMP levels. Ghrelin mimetics may be useful in the treatment of estrogen-dependent breast cancer.


Subject(s)
Adipose Tissue/enzymology , Aromatase/chemistry , Breast/enzymology , Cyclic AMP/metabolism , Ghrelin/pharmacology , Stromal Cells/enzymology , Adipose Tissue/cytology , Adipose Tissue/drug effects , Aromatase/genetics , Aromatase/metabolism , Blotting, Western , Breast/cytology , Breast/drug effects , Calcium/metabolism , Cells, Cultured , Female , Humans , Piperidines/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Ghrelin/agonists , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology , Stromal Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...