Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Trends Pharmacol Sci ; 43(2): 110-122, 2022 02.
Article in English | MEDLINE | ID: mdl-34865885

ABSTRACT

Irritable bowel syndrome (IBS) is a chronic gastrointestinal disorder characterized by abdominal pain and altered bowel habit that affects ~11% of the global population. Over the past decade, preclinical and clinical studies have revealed a variety of novel mechanisms relating to the visceral analgesic effects of guanylate cyclase-C (GC-C) agonists. Here we discuss the mechanisms by which GC-C agonists target the GC-C/cyclic guanosine-3',5'-monophosphate (cGMP) pathway, resulting in visceral analgesia as well as clinically relevant relief of abdominal pain and other sensations in IBS patients. Due to the preponderance of evidence we focus on linaclotide, a 14-amino acid GC-C agonist with very low oral bioavailability that acts within the gut. Collectively, the weight of experimental and clinical evidence supports the concept that GC-C agonists act as peripherally acting visceral analgesics.


Subject(s)
Chronic Pain , Guanylyl Cyclase C Agonists , Irritable Bowel Syndrome , Visceral Pain , Abdominal Pain/drug therapy , Chronic Pain/drug therapy , Guanylate Cyclase/metabolism , Guanylate Cyclase/therapeutic use , Guanylyl Cyclase C Agonists/pharmacology , Guanylyl Cyclase C Agonists/therapeutic use , Humans , Irritable Bowel Syndrome/drug therapy , Visceral Pain/drug therapy
2.
Neurogastroenterol Motil ; 33(3): e14076, 2021 03.
Article in English | MEDLINE | ID: mdl-33373484

ABSTRACT

BACKGROUND: Psychological stress is a risk factor for irritable bowel syndrome, a functional gastrointestinal pain disorder featuring abnormal brain-gut connectivity. The guanylate cyclase-C (GC-C) agonist linaclotide has been shown to relieve abdominal pain in IBS-C and exhibits antinociceptive effects in rodent models of post-inflammatory visceral hypersensitivity. However, the role GC-C signaling plays in psychological stress-induced visceral hypersensitivity is unknown. Here, we test the hypothesis that GC-C agonism reverses stress-induced colonic hypersensitivity via inhibition of nociceptive afferent signaling resulting in normalization of stress-altered corticotropin-releasing factor (CRF) expression in brain regions involved in pain perception and modulation. METHODS: Adult female rats were exposed to water avoidance stress or sham stress for 10 days, and the effects of linaclotide on stress-induced changes in colonic sensitivity, corticolimbic phospho-extracellular signal-regulated kinase (pERK), and CRF expression were measured using a combination of behavioral assessments, immunohistochemistry, and qRT-PCR. KEY RESULTS: Stressed rats exhibited colonic hypersensitivity and elevated corticolimbic pERK on day 11, which was inhibited by linaclotide. qRT-PCR analysis revealed dysregulated CRF expression in the medial prefrontal cortex, paraventricular nucleus of the hypothalamus, and central nucleus of the amygdala on day 28. Dysregulated CRF expression was not affected by linaclotide treatment. CONCLUSIONS AND INFERENCES: Our results demonstrate that exposure to repeated stress induces chronic colonic hypersensitivity in conjunction with altered corticolimbic activation and CRF expression. GC-C agonism attenuated stress-induced colonic hypersensitivity and ERK phosphorylation, but had no effect on CRF expression, suggesting the analgesic effects of linaclotide occur independent of stress-driven CRF gene expression in corticolimbic circuitry.


Subject(s)
Brain/drug effects , Colon/drug effects , Corticotropin-Releasing Hormone/genetics , Guanylyl Cyclase C Agonists/pharmacology , Nociception/drug effects , Peptides/pharmacology , Receptors, Enterotoxin/metabolism , Stress, Psychological/metabolism , Animals , Brain/metabolism , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Colon/metabolism , Corticotropin-Releasing Hormone/drug effects , Corticotropin-Releasing Hormone/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Limbic System/drug effects , Limbic System/metabolism , Pain Threshold , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Stress, Psychological/physiopathology
3.
Front Neurosci ; 14: 590871, 2020.
Article in English | MEDLINE | ID: mdl-33192275

ABSTRACT

Interstitial cystitis/bladder pain syndrome (IC/BPS) is a chronic urological condition characterised by urinary urgency, frequency and pelvic pain, that significantly impacts the quality of life for ∼5% of women. Bladder sensation is coordinated by primary afferent sensory neurons that innervate the bladder wall, translating bladder stretch into signals that travel to the brain via the spinal cord. Whilst the pathophysiology of IC/BPS remains unknown, an increase in the permeability of the bladder urothelium has been proposed as an initiating cause. Here we experimentally increased bladder permeability and tracked bladder afferent sensitivity for up to 28 days. We found that one day after increasing bladder epithelial permeability with in vivo bladder infusion of protamine sulfate, mechanosensitive bladder afferents exhibited significant hypersensitivity to bladder filling. This mechanical hypersensitivity was characterised by significantly increased peak afferent firing rates and a decrease in the activation threshold of individual afferents. Bladder afferent hypersensitivity occurred in the absence of inflammation and changes in bladder muscle compliance, indicating a direct sensitisation of peripheral afferent endings. Bladder afferent mechanosensitive responses to distension returned to control levels by day 7 post-protamine sulfate treatment and remained at control levels at 28-days post-treatment. Here we demonstrate, contrary to the prevailing hypothesis, that increased bladder permeability alone does not induce chronic bladder afferent sensitisation. Whilst experimentally induced changes in bladder permeability are able to induce transient bladder afferent hypersensitivity in the absence of inflammation, highly regulated homeostatic mechanisms exist to rapidly repair the urothelial barrier and normalise bladder afferent mechanosensitivity. Together, these data suggest that additional pathophysiology is required to induce chronic bladder dysfunction.

4.
Pain ; 160(11): 2566-2579, 2019 11.
Article in English | MEDLINE | ID: mdl-31335750

ABSTRACT

Endometriosis, an estrogen-dependent chronic inflammatory disease, is the most common cause of chronic pelvic pain. Here, we investigated the effects of linaclotide, a Food and Drug Administration-approved treatment for IBS-C, in a rat model of endometriosis. Eight weeks after endometrium transplantation into the intestinal mesentery, rats developed endometrial lesions as well as vaginal hyperalgesia to distension and decreased mechanical hind paw withdrawal thresholds. Daily oral administration of linaclotide, a peripherally restricted guanylate cyclase-C (GC-C) agonist peptide acting locally within the gastrointestinal tract, increased pain thresholds to vaginal distension and mechanical hind paw withdrawal thresholds relative to vehicle treatment. Furthermore, using a cross-over design, administering linaclotide to rats previously administered vehicle resulted in increased hind paw withdrawal thresholds, whereas replacing linaclotide with vehicle treatment decreased hind paw withdrawal thresholds. Retrograde tracing of sensory afferent nerves from the ileum, colon, and vagina revealed that central terminals of these afferents lie in close apposition to one another within the dorsal horn of the spinal cord. We also identified dichotomizing dual-labelled ileal/colon innervating afferents as well as colon/vaginal dual-labelled neurons and a rare population of triple traced ileal/colon/vaginal neurons within thoracolumbar DRG. These observations provide potential sources of cross-organ interaction at the level of the DRG and spinal cord. GC-C expression is absent in the vagina and endometrial cysts suggesting that the actions of linaclotide are shared through nerve pathways between these organs. In summary, linaclotide may offer a novel therapeutic option not only for treatment of chronic endometriosis-associated pain, but also for concurrent treatment of comorbid chronic pelvic pain syndromes.


Subject(s)
Endometriosis/chemically induced , Hyperalgesia/etiology , Pain/drug therapy , Peptides/pharmacology , Afferent Pathways/drug effects , Afferent Pathways/metabolism , Animals , Endometriosis/complications , Female , Hyperalgesia/drug therapy , Peptides/adverse effects , Peptides/therapeutic use , Rats, Sprague-Dawley , Spinal Cord/drug effects , Spinal Cord/metabolism
5.
JCI Insight ; 3(19)2018 10 04.
Article in English | MEDLINE | ID: mdl-30282832

ABSTRACT

Irritable bowel syndrome (IBS) patients suffer from chronic abdominal pain and extraintestinal comorbidities, including overactive bladder (OAB) and interstitial cystitis/painful bladder syndrome (IC-PBS). Mechanistic understanding of the cause and time course of these comorbid symptoms is lacking, as are clinical treatments. Here, we report that colitis triggers hypersensitivity of colonic afferents, neuroplasticity of spinal cord circuits, and chronic abdominal pain, which persists after inflammation. Subsequently, and in the absence of bladder pathology, colonic hypersensitivity induces persistent hypersensitivity of bladder afferent pathways, resulting in bladder-voiding dysfunction, indicative of OAB/IC-PBS. Daily administration of linaclotide, a guanylate cyclase-C (GC-C) agonist that is restricted to and acts within the gastrointestinal tract, reverses colonic afferent hypersensitivity, reverses neuroplasticity-induced alterations in spinal circuitry, and alleviates chronic abdominal pain in mice. Intriguingly, daily linaclotide administration also reverses persistent bladder afferent hypersensitivity to mechanical and chemical stimuli and restores normal bladder voiding. Linaclotide itself does not inhibit bladder afferents, rather normalization of bladder function by daily linaclotide treatment occurs via indirect inhibition of bladder afferents via reduced nociceptive signaling from the colon. These data support the concepts that cross-organ sensitization underlies the development and maintenance of visceral comorbidities, while pharmaceutical treatments that inhibit colonic afferents may also improve urological symptoms through common sensory pathways.


Subject(s)
Guanylyl Cyclase C Agonists/administration & dosage , Hyperalgesia/drug therapy , Irritable Bowel Syndrome/drug therapy , Neuronal Plasticity/drug effects , Peptides/administration & dosage , Urinary Bladder, Overactive/drug therapy , Afferent Pathways/drug effects , Animals , Colitis/chemically induced , Colon/drug effects , Colon/innervation , Disease Models, Animal , Drug Administration Schedule , Humans , Hyperalgesia/chemically induced , Hyperalgesia/complications , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/complications , Male , Mice , Nociception/drug effects , Treatment Outcome , Trinitrobenzenesulfonic Acid/toxicity , Urinary Bladder/innervation , Urinary Bladder, Overactive/etiology
6.
J Pharmacol Exp Ther ; 366(2): 274-281, 2018 08.
Article in English | MEDLINE | ID: mdl-29784661

ABSTRACT

Bladder pain syndrome (BPS) is poorly understood; however, there is a female predominance and comorbidity with irritable bowel syndrome (IBS). Here we test the hypothesis that linaclotide, a guanylate cyclase-C (GC-C) agonist approved for the treatment of IBS with constipation (IBS-C), may represent a novel therapeutic for BPS acting through a mechanism involving an inhibition of visceral organ cross-sensitization. We showed previously that infusion of dilute protamine sulfate (PS) into the bladder increased sensitivity and permeability in the bladder and colon. PS was infused into the bladder of female rats; sensitivity was assessed via application of von Frey filaments applied to the suprapubic area and the frequency of withdrawal responses was recorded. Colonic sensitivity was measured via visceromotor behavioral response to graded pressures of colorectal distension (CRD). Permeability was measured in vitro via transepithelial electrical resistance (TEER) and conductance (G). Linaclotide (3 µg/kg, p.o.) or vehicle was administered daily for 7 days prior to experiments. Rats treated with PS bladder infusion exhibited visceral hyperalgesia, as shown by a significantly higher response frequency to individual von Frey filaments and increased behavioral responses to CRD. Linaclotide attenuated bladder and colonic hyperalgesia to control levels. PS infusion into the bladder increased bladder and colon permeability measured as a decrease in TEER and increased G. Linaclotide significantly inhibited PS-induced colonic hyperpermeability while having no effect on bladder hyperpermeability. Our findings suggest a novel treatment paradigm for GC-C agonism in IBS-C and BPS mediated through a mechanism involving visceral organ crosstalk.


Subject(s)
Colon/drug effects , Colon/metabolism , Guanylate Cyclase/metabolism , Peptides/pharmacology , Urinary Bladder/drug effects , Urinary Bladder/metabolism , Animals , Enzyme Activation/drug effects , Female , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord/cytology , Spinal Cord/drug effects
7.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978796

ABSTRACT

Cystic fibrosis (CF) is a genetic disorder in which epithelium-generated fluid flow from the lung, intestine, and pancreas is impaired due to mutations disrupting CF transmembrane conductance regulator (CFTR) channel function. CF manifestations of the pancreas and lung are present in the vast majority of CF patients, and 15% of CF infants are born with obstructed gut or meconium ileus. However, constipation is a significantly underreported outcome of CF disease, affecting 47% of the CF patients, and management becomes critical in the wake of increasing life span of CF patients. In this study, we unraveled a potentially novel molecular role of a membrane-bound cyclic guanosine monophosphate-synthesizing (cGMP-synthesizing) intestinal enzyme, guanylate cyclase 2C (GCC) that could be targeted to ameliorate CF-associated intestinal fluid deficit. We demonstrated that GCC agonism results in functional rescue of murine F508del/F508del and R117H/R117H Cftr and CFTR mutants in CF patient-derived intestinal spheres. GCC coexpression and activation facilitated processing and ER exit of F508del CFTR and presented a potentially novel rescue modality in the intestine, similar to the CF corrector VX-809. Our findings identify GCC as a biological CFTR corrector and potentiator in the intestine.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Receptors, Enterotoxin/physiology , Animals , Cystic Fibrosis/enzymology , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Humans , Intestinal Mucosa/metabolism , Mice, Mutant Strains , Mutation , Organoids/metabolism , Organoids/pathology , Stem Cells/metabolism
8.
Physiol Rep ; 5(11)2017 Jun.
Article in English | MEDLINE | ID: mdl-28592587

ABSTRACT

The transmembrane receptor guanylyl cyclase-C (GC-C), expressed on enterocytes along the intestine, is the molecular target of the GC-C agonist peptide linaclotide, an FDA-approved drug for treatment of adult patients with Irritable Bowel Syndrome with Constipation and Chronic Idiopathic Constipation. Polarized human colonic intestinal cells (T84, CaCo-2BBe) rat and human intestinal tissues were employed to examine cellular signaling and cystic fibrosis transmembrane conductance regulator (CFTR)-trafficking pathways activated by linaclotide using confocal microscopy, in vivo surface biotinylation, and protein kinase-II (PKG-II) activity assays. Expression and activity of GC-C/cGMP pathway components were determined by PCR, western blot, and cGMP assays. Fluid secretion as a marker of CFTR cell surface translocation was determined using in vivo rat intestinal loops. Linaclotide treatment (30 min) induced robust fluid secretion and translocation of CFTR from subapical compartments to the cell surface in rat intestinal loops. Similarly, linaclotide treatment (30 min) of T84 and CaCo-2BBe cells increased cell surface CFTR levels. Linaclotide-induced activation of the GC-C/cGMP/PKGII signaling pathway resulted in elevated intracellular cGMP and pVASPser239 phosphorylation. Inhibition or silencing of PKGII significantly attenuated linaclotide-induced CFTR trafficking to the apical membrane. Inhibition of protein kinase-A (PKA) also attenuated linaclotide-induced CFTR cell surface trafficking, implying cGMP-dependent cross-activation of PKA pathway. Together, these findings support linaclotide-induced activation of the GC-C/cGMP/PKG-II/CFTR pathway as the major pathway of linaclotide-mediated intestinal fluid secretion, and that linaclotide-dependent CFTR activation and recruitment/trafficking of CFTR from subapical vesicles to the cell surface is an important step in this process.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Guanylyl Cyclase C Agonists/pharmacology , Intestinal Mucosa/metabolism , Peptides/pharmacology , Signal Transduction , Animals , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinase Type II/metabolism , Humans , Intestinal Mucosa/drug effects , Male , Protein Transport , Rats , Rats, Sprague-Dawley , Receptors, Guanylate Cyclase-Coupled/metabolism
9.
Invest Ophthalmol Vis Sci ; 57(3): 1317-26, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26998718

ABSTRACT

PURPOSE: The nitric oxide (NO)-cyclic guanosine-3',5'-monophosphate (cGMP) pathway regulates aqueous humor outflow and therefore, intraocular pressure. We investigated the pharmacologic effects of the soluble guanylate cyclase (sGC) stimulator IWP-953 on primary human trabecular meshwork (HTM) cells and conventional outflow facility in mouse eyes. METHODS: Cyclic GMP levels were determined in vitro in HEK-293 cells and four HTM cell strains (HTM120/HTM123: predominantly myofibroblast-like phenotype, HTM130/HTM141: predominantly endothelial-like phenotype), and in HTM cell culture supernatants. Conventional outflow facility was measured following intracameral injection of IWP-953 or DETA-NO using a computerized pressure-controlled perfusion system in enucleated mouse eyes ex vivo. RESULTS: IWP-953 markedly stimulated cGMP production in HEK-293 cells in the presence and absence of DETA-NO (half maximal effective concentrations: 17 nM, 9.5 µM). Similarly, IWP-953 stimulated cGMP production in myofibroblast-like HTM120 and HTM123 cells, an effect that was greatly amplified by the presence of DETA-NO. In contrast, IWP-953 stimulation of cGMP production in endothelial-like HTM130 and HTM141 cells was observed, but was markedly less prominent than in HTM120 and HTM123 cells. Notably, cGMP was found in all HTM culture supernatants, following IWP-953/DETA-NO stimulation. In paired enucleated mouse eyes, IWP-953 at 10, 30, 60, and 100 µM concentration-dependently increased outflow facility. This effect (89.5%) was maximal at 100 µM (P = 0.002) and in magnitude comparable to DETA-NO at 100 µM (97.5% increase, P = 0.030). CONCLUSIONS: These data indicate that IWP-953, via modulation of the sGC-cGMP pathway, increases aqueous outflow facility in mouse eyes, suggesting therapeutic potential for sGC stimulators as novel ocular hypotensive drugs.


Subject(s)
Aqueous Humor/chemistry , Cyclic GMP/metabolism , Enzyme Inhibitors/therapeutic use , Glaucoma, Open-Angle/drug therapy , Guanylate Cyclase/drug effects , Intraocular Pressure/drug effects , Trabecular Meshwork/metabolism , Adult , Animals , Cells, Cultured , Child, Preschool , Disease Models, Animal , Glaucoma, Open-Angle/pathology , Glaucoma, Open-Angle/physiopathology , Guanylate Cyclase/metabolism , Humans , Infant , Mice , Mice, Inbred C57BL , Trabecular Meshwork/pathology
10.
J Pharmacol Exp Ther ; 355(1): 48-56, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26216942

ABSTRACT

MRP4 mediates the efflux of cGMP and cAMP and acts as an important regulator of these secondary messengers, thereby affecting signaling events mediated by cGMP and cAMP. Immunofluorescence staining showed high MRP4 expression localized predominantly in the apical membrane of rat colonic epithelium. In vitro studies were performed using a rat colonic mucosal layer mounted in an Ussing chamber. Linaclotide activation of the guanylate cyclase-C (GC-C)/cGMP pathway induced a concentration-dependent increase in transepithelial ion current [short-circuit current (Isc)] across rat colonic mucosa (EC50: 9.2 nM). Pretreatment of colonic mucosa with the specific MRP4 inhibitor MK571 potentiated linaclotide-induced electrolyte secretion and augmented linaclotide-stimulated intracellular cGMP accumulation. Notably, pretreatment with the phosphodiesterase 5 inhibitor sildenafil increased basal Isc, but had no amplifying effect on linaclotide-induced Isc. MRP4 inhibition selectively affected the activation phase, but not the deactivation phase, of linaclotide. In contrast, incubation with a GC-C/Fc chimera binding to linaclotide abrogated linaclotide-induced Isc, returning to baseline. Furthermore, linaclotide activation of GC-C induced cGMP secretion from the apical and basolateral membranes of colonic epithelium. MRP4 inhibition blocked cGMP efflux from the apical membrane, but not the basolateral membrane. These data reveal a novel, previously unrecognized mechanism that functionally couples GC-C-induced luminal electrolyte transport and cGMP secretion to spatially restricted, compartmentalized regulation by MRP4 at the apical membrane of intestinal epithelium. These findings have important implications for gastrointestinal disorders with symptoms associated with dysregulated fluid homeostasis, such as irritable bowel syndrome with constipation, chronic idiopathic constipation, and secretory diarrhea.


Subject(s)
Cyclic GMP/metabolism , Electrolytes/metabolism , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Peptides/pharmacology , Propionates/pharmacology , Quinolines/pharmacology , Receptors, Guanylate Cyclase-Coupled/metabolism , Receptors, Peptide/metabolism , Signal Transduction/drug effects , Animals , Biological Transport/drug effects , Colon/cytology , Colon/drug effects , Colon/metabolism , Colon/physiology , Electrophysiological Phenomena/drug effects , Female , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Kinetics , Rats , Rats, Sprague-Dawley , Receptors, Enterotoxin
11.
Front Mol Neurosci ; 7: 31, 2014.
Article in English | MEDLINE | ID: mdl-24795564

ABSTRACT

Activation of guanylate cyclase-C (GC-C) expressed predominantly on intestinal epithelial cells by guanylin, uroguanylin or the closely related GC-C agonist peptide, linaclotide, stimulates generation, and release of cyclic guanosine-3',5'-monophosphate (cGMP). Evidence that the visceral analgesic effects of linaclotide are mediated by a novel, GC-C-dependent peripheral sensory mechanism was first demonstrated in animal models of visceral pain. Subsequent studies with uroguanylin or linaclotide have confirmed the activation of a GC-C/cGMP pathway leading to increased submucosal cGMP mediated by cGMP efflux pumps, which modulates intestinal nociceptor function resulting in peripheral analgesia. These effects can be reproduced by the addition of exogenous cGMP and support a role for GC-C/cGMP signaling in the regulation of visceral sensation, a physiological function that has not previously been linked to the GC-C/cGMP pathway. Notably, targeting the GC-C/cGMP pathway for treatment of gastrointestinal pain and abdominal sensory symptoms has now been validated in the clinic. In 2012, linaclotide was approved in the United States and European Union for the treatment of adult patients with irritable bowel syndrome with constipation.

12.
Gastroenterology ; 145(6): 1334-46.e1-11, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23958540

ABSTRACT

BACKGROUND & AIMS: Linaclotide is a minimally absorbed agonist of guanylate cyclase-C (GUCY2C or GC-C) that reduces symptoms associated with irritable bowel syndrome with constipation (IBS-C). Little is known about the mechanism by which linaclotide reduces abdominal pain in patients with IBS-C. METHODS: We determined the effects of linaclotide on colonic sensory afferents in healthy mice and those with chronic visceral hypersensitivity. We assessed pain transmission by measuring activation of dorsal horn neurons in the spinal cord in response to noxious colorectal distention. Levels of Gucy2c messenger RNA were measured in tissues from mice using quantitative reverse transcription polymerase chain reaction and in situ hybridization. We used human intestinal cell lines to measure release of cyclic guanosine-3',5'-monophosphate (cGMP) by linaclotide. We performed a post-hoc analysis of data from a phase III, double-blind, parallel-group study in which 805 patients with IBS-C were randomly assigned to groups given an oral placebo or 290 µg linaclotide once daily for 26 weeks. We quantified changes in IBS-C symptoms, including abdominal pain. RESULTS: In mice, linaclotide inhibited colonic nociceptors with greater efficacy during chronic visceral hypersensitivity. Intra-colonic administration of linaclotide reduced signaling of noxious colorectal distention to the spinal cord. The colonic mucosa, but not neurons, was found to express linaclotide's target, GC-C. The downstream effector of GC-C, cGMP, was released after administration of linaclotide and also inhibited nociceptors. The effects of linaclotide were lost in Gucy2c(-/-) mice and prevented by inhibiting cGMP transporters or removing the mucosa. During 26 weeks of linaclotide administration, a significantly greater percentage of patients (70%) had at least a 30% reduction in abdominal pain compared with patients given placebo (50%). CONCLUSIONS: We have identified an analgesic mechanism of linaclotide: it activates GC-C expressed on mucosal epithelial cells, resulting in the production and release of cGMP. This extracellular cGMP acts on and inhibits nociceptors, thereby reducing nociception. We also found that linaclotide reduces chronic abdominal pain in patients with IBS-C.


Subject(s)
Abdominal Pain/prevention & control , Colon/innervation , Cyclic GMP/physiology , Guanylate Cyclase/physiology , Nociceptors/drug effects , Peptides/pharmacology , Peptides/therapeutic use , Abdominal Pain/chemically induced , Adult , Aged , Aged, 80 and over , Animals , Caco-2 Cells , Cell Line , Colon/drug effects , Colon/pathology , Disease Models, Animal , Double-Blind Method , Female , Humans , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/prevention & control , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Natriuretic Peptides/pharmacology , Nociceptors/physiology , Receptors, Atrial Natriuretic Factor/physiology , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled/physiology , Receptors, Peptide/physiology , Treatment Outcome , Trinitrobenzenesulfonic Acid/adverse effects
13.
Pain ; 154(9): 1820-1830, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23748116

ABSTRACT

The natural hormone uroguanylin regulates intestinal fluid homeostasis and bowel function through activation of guanylate cyclase-C (GC-C), resulting in increased intracellular cyclic guanosine-3',5'-monophosphate (cGMP). We report the effects of uroguanylin-mediated activation of the GC-C/cGMP pathway in vitro on extracellular cGMP transport and in vivo in rat models of inflammation- and stress-induced visceral hypersensitivity. In vitro exposure of intestinal Caco-2 cells to uroguanylin stimulated bidirectional, active extracellular transport of cGMP into luminal and basolateral spaces. cGMP transport was significantly and concentration dependently decreased by probenecid, an inhibitor of cGMP efflux pumps. In ex vivo Ussing chamber assays, uroguanylin stimulated cGMP secretion from the basolateral side of rat colonic epithelium into the submucosal space. In a rat model of trinitrobenzene sulfonic acid (TNBS)-induced visceral hypersensitivity, orally administered uroguanylin increased colonic thresholds required to elicit abdominal contractions in response to colorectal distension (CRD). Oral administration of cGMP mimicked the antihyperalgesic effects of uroguanylin, significantly decreasing TNBS- and restraint stress-induced visceromotor response to graded CRD in rats. The antihyperalgesic effects of cGMP were not associated with increased colonic spasmolytic activity, but were linked to significantly decreased firing rates of TNBS-sensitized colonic afferents in rats in response to mechanical stimuli. In conclusion, these data suggest that the continuous activation of the GC-C/cGMP pathway along the intestinal tract by the endogenous hormones guanylin and uroguanylin results in significant reduction of gastrointestinal pain. Extracellular cGMP produced on activation of GC-C is the primary mediator in this process via modulation of sensory afferent activity.


Subject(s)
Guanylate Cyclase/metabolism , Natriuretic Peptides/metabolism , Signal Transduction/physiology , Visceral Pain/metabolism , Acetylcholine/pharmacology , Acetylglucosamine/analogs & derivatives , Acetylglucosamine/pharmacology , Adenocarcinoma/pathology , Animals , Cell Differentiation/drug effects , Cell Line, Tumor , Colitis/chemically induced , Colitis/complications , Colon/drug effects , Colon/metabolism , Colorectal Neoplasms/pathology , Cyclic GMP/analogs & derivatives , Cyclic GMP/metabolism , Cyclic GMP/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Electric Stimulation , Female , Gastrointestinal Diseases/complications , Gastrointestinal Diseases/etiology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hyperalgesia/physiopathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/physiology , Male , Mast Cells/drug effects , Mast Cells/metabolism , Morphine/therapeutic use , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Natriuretic Peptides/therapeutic use , Organic Anion Transporters, Sodium-Independent/genetics , Organic Anion Transporters, Sodium-Independent/metabolism , Peroxidase/metabolism , RNA, Messenger , Rats , Rats, Sprague-Dawley , Rats, Wistar , Restraint, Physical , Trinitrobenzenesulfonic Acid/toxicity , Visceral Pain/drug therapy , Visceral Pain/etiology
14.
J Pharmacol Exp Ther ; 344(1): 196-206, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23090647

ABSTRACT

Linaclotide, a potent guanylate cyclase C agonist, is a therapeutic peptide approved in the United States for the treatment of irritable bowel syndrome with constipation and chronic idiopathic constipation. We present for the first time the metabolism, degradation, and disposition of linaclotide in animals and humans. We examined the metabolic stability of linaclotide in conditions that mimic the gastrointestinal tract and characterized the metabolite MM-419447 (CCEYCCNPACTGC), which contributes to the pharmacologic effects of linaclotide. Systemic exposure to these active peptides is low in rats and humans, and the low systemic and portal vein concentrations of linaclotide and MM-419447 observed in the rat confirmed both peptides are minimally absorbed after oral administration. Linaclotide is stable in the acidic environment of the stomach and is converted to MM-419447 in the small intestine. The disulfide bonds of both peptides are reduced in the small intestine, where they are subsequently proteolyzed and degraded. After oral administration of linaclotide, <1% of the dose was excreted as active peptide in rat feces and a mean of 3-5% in human feces; in both cases MM-419447 was the predominant peptide recovered. MM-419447 exhibits high-affinity binding in vitro to T84 cells, resulting in a significant, concentration-dependent accumulation of intracellular cyclic guanosine-3',5'-monophosphate (cGMP). In rat models of gastrointestinal function, orally dosed MM-419447 significantly increased fluid secretion into small intestinal loops, increased intraluminal cGMP, and caused a dose-dependent acceleration in gastrointestinal transit. These results demonstrate the importance of the active metabolite in contributing to linaclotide's pharmacology.


Subject(s)
Constipation/drug therapy , Irritable Bowel Syndrome/drug therapy , Peptides/pharmacology , Alkylation , Animals , Area Under Curve , Biological Availability , Biotransformation , Constipation/complications , Cyclic AMP/metabolism , Feces/chemistry , Female , Gastrointestinal Transit/drug effects , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Intestinal Mucosa/metabolism , Intestines/drug effects , Irritable Bowel Syndrome/complications , Male , Peptide Hydrolases/chemistry , Peptides/pharmacokinetics , Peptides/therapeutic use , Radioligand Assay , Rats , Rats, Sprague-Dawley
15.
Eur J Pharmacol ; 649(1-3): 328-35, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20863829

ABSTRACT

Linaclotide is a first-in-class, orally administered 14-amino acid peptide that is in development for the treatment of irritable bowel syndrome with constipation and chronic constipation. We have characterized the solution structure of linaclotide, the in vitro binding and agonist activity to guanylate cyclase C receptors, the stability of linaclotide under conditions mimicking the gastric environment, oral bioavailability, and the pharmacodynamic effects in rat models of gastrointestinal transit and intestinal secretion. Nuclear magnetic resonance spectroscopy analysis determined that the molecular structure of linaclotide is stabilized by three intramolecular disulfide bridges. Linaclotide exhibited high affinity and pH-independent binding (K(i): 1.23-1.64 nM) to guanylate cyclase C receptors on human colon carcinoma T84 cells and concomitantly, linaclotide binding resulted in a significant, concentration-dependent accumulation of intracellular cyclic guanosine-3', 5'-monophosphate (cGMP) (EC50:99 nM). Linaclotide was stable after 3 h incubation in simulated gastric fluid (pH 1) and similarly, was completely resistant to hydrolysis by pepsin. Pharmacokinetic analysis of linaclotide showed very low oral bioavailability (0.1%). Orally administered linaclotide elicited a significant, dose-dependent increase in gastrointestinal transit rates in rats at doses of ≥5 µg/kg. Exposure of surgically ligated small intestinal loops to linaclotide induced a significant increase in fluid secretion, accompanied by a significant increase in intraluminal cGMP levels. These results suggest that the guanylate cyclase C agonist linaclotide elicits potent pharmacological responses locally in the gastrointestinal tract, and that orally administered guanylate cyclase C agonists may be capable of improving bowel habits in patients suffering from irritable bowel syndrome with constipation and chronic constipation.


Subject(s)
Gastrointestinal Transit/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Laxatives/pharmacology , Peptides/pharmacology , Receptors, Guanylate Cyclase-Coupled/agonists , Receptors, Peptide/agonists , Animals , Binding, Competitive , Biological Availability , Cell Line , Cells, Cultured , Constipation/drug therapy , Cyclic GMP/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Female , Humans , Intestinal Mucosa/cytology , Intestinal Secretions/metabolism , Irritable Bowel Syndrome/drug therapy , Laxatives/chemistry , Laxatives/metabolism , Laxatives/pharmacokinetics , Male , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacokinetics , Protein Conformation , Protein Stability , Rats , Receptors, Enterotoxin
16.
Bioorg Med Chem Lett ; 20(8): 2520-4, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20304639

ABSTRACT

In pursuit of a potent and highly selective sphingosine-1-phosphate receptor agonists with an improved in vivo conversion of the precursor to the active phospho-drug, we have utilized previously reported phenylamide and phenylimidazole scaffolds to identify a selectivity enhancing moiety (SEM) and selectivity enhancing orientation (SEO) within both pharmacophores. SEM and SEO have allowed for over 100 to 500-fold improvement in selectivity for S1P receptor subtype 1 over subtype 3. Utility of SEM and SEO and further SAR study allowed for discovery of a potent and selective preclinical candidate PPI-4955 (21b) with an excellent in vivo potency and dose responsiveness and markedly improved overall in vivo pharmacodynamic properties upon oral administration.


Subject(s)
Amino Alcohols/pharmacology , Receptors, Lysosphingolipid/agonists , Administration, Oral , Amino Alcohols/administration & dosage , Animals , Mice , Structure-Activity Relationship
17.
Life Sci ; 86(19-20): 760-5, 2010 May 08.
Article in English | MEDLINE | ID: mdl-20307554

ABSTRACT

AIMS: Linaclotide is an orally administered 14-amino acid peptide being developed for the treatment of constipation-predominant irritable bowel syndrome (IBS-C) and chronic constipation. We determined the stability of linaclotide in the intestine, measured the oral bioavailability, and investigated whether the pharmacodynamic effects elicited in rodent models of gastrointestinal function are mechanistically linked to the activation of intestinal guanylate cyclase C (GC-C). MAIN METHODS: Linaclotide binding to intestinal mucosal membranes was assessed in competitive binding assays. Stability and oral bioavailability of linaclotide were measured in small intestinal fluid and serum, respectively, and models of gastrointestinal function were conducted using wild type (wt) and GC-C null mice. KEY FINDINGS: Linaclotide inhibited in vitro [(125)I]-STa binding to intestinal mucosal membranes from wt mice in a concentration-dependent manner. In contrast, [(125)I]-STa binding to these membranes from GC-C null mice was significantly decreased. After incubation in vitro in jejunal fluid for 30 min, linaclotide was completely degraded. Pharmacokinetic analysis showed very low oral bioavailability (0.10%). In intestinal secretion and transit models, linaclotide exhibited significant pharmacological effects in wt, but not in GC-C null mice: induction of increased fluid secretion into surgically ligated jejunal loops was accompanied by the secretion of elevated levels of cyclic guanosine-3',5'-monophosphate and accelerated gastrointestinal transit. SIGNIFICANCE: Linaclotide is a potent and selective GC-C agonist that elicits pharmacological effects locally in the gastrointestinal tract. This pharmacological profile suggests that orally administered linaclotide may be capable of improving the abdominal symptoms and bowel habits of patients suffering from IBS-C and chronic constipation.


Subject(s)
Gastrointestinal Agents/pharmacology , Peptides/pharmacology , Receptors, Peptide/agonists , Administration, Oral , Animals , Biological Availability , Dose-Response Relationship, Drug , Female , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/pharmacokinetics , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Gastrointestinal Transit/drug effects , Guanylate Cyclase/genetics , Intestinal Mucosa/metabolism , Intestinal Secretions/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides/administration & dosage , Peptides/pharmacokinetics , Receptors, Enterotoxin , Receptors, Guanylate Cyclase-Coupled , Receptors, Peptide/genetics
18.
J Med Chem ; 52(24): 8047-56, 2009 Dec 24.
Article in English | MEDLINE | ID: mdl-19929003

ABSTRACT

Inhibition of methionine aminopeptidase-2 (MetAP2) represents a novel approach to antiangiogenic therapy. We describe the synthesis and activity of fumagillin analogues that address the pharmacokinetic and safety liabilities of earlier candidates in this compound class. Two-step elaboration of fumagillol with amines yielded a diverse series of carbamates at C6 of the cyclohexane spiroepoxide. The most potent of these compounds exhibited subnanomolar inhibition of cell proliferation in HUVEC and BAEC assays. Although a range of functionalities were tolerated at this position, alpha-trisubstituted amines possessed markedly decreased inhibitory activity, and this could be rationalized by modeling based on the known fumagillin-MetAP2 crystal structure. The lead compound resulting from these studies, (3R,4S,5S,6R)-5-methoxy-4-((2R,3R)-2-methyl-3-(3-methylbut-2-enyl)oxiran-2-yl)-1-oxaspiro[2.5]octan-6-yl (R)-1-amino-3-methyl-1-oxobutan-2-ylcarbamate, (PPI-2458), demonstrated an improved pharmacokinetic profile relative to the earlier clinical candidate TNP-470, and has advanced into phase I clinical studies in non-Hodgkin's lymphoma and solid cancers.


Subject(s)
Aminopeptidases/antagonists & inhibitors , Carbamates/chemistry , Carbamates/pharmacology , Cyclohexanes/chemistry , Cyclohexanes/pharmacology , Fatty Acids, Unsaturated/chemistry , Fatty Acids, Unsaturated/pharmacology , Metalloendopeptidases/antagonists & inhibitors , Amino Acids/chemistry , Animals , Cattle , Cell Growth Processes/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Humans , Models, Molecular , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Structure-Activity Relationship
19.
Bioorg Med Chem Lett ; 19(8): 2315-9, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-19282175

ABSTRACT

In pursuit of potent and selective sphingosine-1-phosphate receptor agonists, we have utilized previously reported phenylamide and phenylimidazole scaffolds to explore extensive side-chain modifications to generate new molecular entities. A number of designed molecules demonstrate good selectivity and excellent in vitro and in vivo potency in both mouse and rat models. Oral administration of the lead molecule 11c (PPI-4667) demonstrated potent and dose-responsive lymphopenia.


Subject(s)
Amides/chemical synthesis , Imidazoles/chemical synthesis , Receptors, Lysosphingolipid/agonists , Amides/pharmacology , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Fingolimod Hydrochloride , Imidazoles/pharmacology , Mice , Propylene Glycols/chemistry , Propylene Glycols/pharmacology , Protein Subunits/agonists , Protein Subunits/physiology , Receptors, Lysosphingolipid/physiology , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Sphingosine/pharmacology
20.
Bioorg Med Chem Lett ; 19(2): 369-72, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19081720

ABSTRACT

In the design of potent and selective sphingosine-1-phosphate receptor agonists, we were able to identify two series of molecules based on phenylamide and phenylimidazole analogs of FTY-720. Several designed molecules in these scaffolds have demonstrated selectivity for S1P receptor subtype 1 versus 3 and excellent in vivo activity in mouse. Two molecules PPI-4621 (4b) and PPI-4691 (10a), demonstrated dose responsive lymphopenia, when administered orally.


Subject(s)
Amides/chemical synthesis , Amides/pharmacology , Imidazoles/chemical synthesis , Imidazoles/pharmacology , Receptors, Lysosphingolipid/agonists , Amides/chemistry , Animals , Dose-Response Relationship, Drug , Humans , Imidazoles/chemistry , Mice , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...