Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nature ; 560(7719): 504-508, 2018 08.
Article in English | MEDLINE | ID: mdl-30051891

ABSTRACT

Histone H3 lysine 9 methylation (H3K9me) mediates heterochromatic gene silencing and is important for genome stability and the regulation of gene expression1-4. The establishment and epigenetic maintenance of heterochromatin involve the recruitment of H3K9 methyltransferases to specific sites on DNA, followed by the recognition of pre-existing H3K9me by the methyltransferase and methylation of proximal histone H35-11. This positive feedback loop must be tightly regulated to prevent deleterious epigenetic gene silencing. Extrinsic anti-silencing mechanisms involving histone demethylation or boundary elements help to limit the spread of inappropriate H3K9me12-15. However, how H3K9 methyltransferase activity is locally restricted or prevented from initiating random H3K9me-which would lead to aberrant gene silencing and epigenetic instability-is not fully understood. Here we reveal an autoinhibited conformation in the conserved H3K9 methyltransferase Clr4 (also known as Suv39h) of the fission yeast Schizosaccharomyces pombe that has a critical role in preventing aberrant heterochromatin formation. Biochemical and X-ray crystallographic data show that an internal loop in Clr4 inhibits the catalytic activity of this enzyme by blocking the histone H3K9 substrate-binding pocket, and that automethylation of specific lysines in this loop promotes a conformational switch that enhances the H3K9me activity of Clr4. Mutations that are predicted to disrupt this regulation lead to aberrant H3K9me, loss of heterochromatin domains and inhibition of growth, demonstrating the importance of the intrinsic inhibition and auto-activation of Clr4 in regulating the deposition of H3K9me and in preventing epigenetic instability. Conservation of the Clr4 autoregulatory loop in other H3K9 methyltransferases and the automethylation of a corresponding lysine in the human SUV39H2 homologue16 suggest that the mechanism described here is broadly conserved.


Subject(s)
Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Epigenesis, Genetic , Histone Methyltransferases/chemistry , Histone Methyltransferases/metabolism , Histones/metabolism , Methyltransferases/chemistry , Methyltransferases/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/enzymology , Schizosaccharomyces/genetics , Evolution, Molecular , Gene Silencing , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Humans , Methylation , Protein Conformation
2.
Nature ; 547(7664): 463-467, 2017 07 27.
Article in English | MEDLINE | ID: mdl-28682306

ABSTRACT

Heterochromatic DNA domains have important roles in the regulation of gene expression and maintenance of genome stability by silencing repetitive DNA elements and transposons. From fission yeast to mammals, heterochromatin assembly at DNA repeats involves the activity of small noncoding RNAs (sRNAs) associated with the RNA interference (RNAi) pathway. Typically, sRNAs, originating from long noncoding RNAs, guide Argonaute-containing effector complexes to complementary nascent RNAs to initiate histone H3 lysine 9 di- and trimethylation (H3K9me2 and H3K9me3, respectively) and the formation of heterochromatin. H3K9me is in turn required for the recruitment of RNAi to chromatin to promote the amplification of sRNA. Yet, how heterochromatin formation, which silences transcription, can proceed by a co-transcriptional mechanism that also promotes sRNA generation remains paradoxical. Here, using Clr4, the fission yeast Schizosaccharomyces pombe homologue of mammalian SUV39H H3K9 methyltransferases, we design active-site mutations that block H3K9me3, but allow H3K9me2 catalysis. We show that H3K9me2 defines a functionally distinct heterochromatin state that is sufficient for RNAi-dependent co-transcriptional gene silencing at pericentromeric DNA repeats. Unlike H3K9me3 domains, which are transcriptionally silent, H3K9me2 domains are transcriptionally active, contain modifications associated with euchromatic transcription, and couple RNAi-mediated transcript degradation to the establishment of H3K9me domains. The two H3K9me states recruit reader proteins with different efficiencies, explaining their different downstream silencing functions. Furthermore, the transition from H3K9me2 to H3K9me3 is required for RNAi-independent epigenetic inheritance of H3K9me domains. Our findings demonstrate that H3K9me2 and H3K9me3 define functionally distinct chromatin states and uncover a mechanism for the formation of transcriptionally permissive heterochromatin that is compatible with its broadly conserved role in sRNA-mediated genome defence.


Subject(s)
Gene Silencing , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/chemistry , Histones/metabolism , RNA Interference , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Transcription, Genetic , Amino Acid Sequence , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Gene Silencing/drug effects , Heterochromatin/chemistry , Histone-Lysine N-Methyltransferase , Hydroxamic Acids/pharmacology , Methylation/drug effects , Methyltransferases/metabolism , Mutation , Repressor Proteins/metabolism , Schizosaccharomyces/drug effects , Schizosaccharomyces pombe Proteins/metabolism , Transcription, Genetic/drug effects
3.
J Biol Chem ; 296: 100397, 2021.
Article in English | MEDLINE | ID: mdl-33571527

ABSTRACT

Since the discovery of the prolyl hydroxylases domain (PHD) proteins and their canonical hypoxia-inducible factor (HIF) substrate two decades ago, a number of in vitro hydroxylation (IVH) assays for PHD activity have been developed to measure the PHD-HIF interaction. However, most of these assays either require complex proteomics mass spectrometry methods that rely on the specific PHD-HIF interaction or require the handling of radioactive material, as seen in the most commonly used assay measuring [14C]O2 release from labeled [14C]α-ketoglutarate. Here, we report an alternative rapid, cost-effective assay in which the consumption of α-ketoglutarate is monitored by its derivatization with 2,4-dinitrophenylhydrazine (2,4-DNPH) followed by treatment with concentrated base. We extensively optimized this 2,4-DNPH α-ketoglutarate assay to maximize the signal-to-noise ratio and demonstrated that it is robust enough to obtain kinetic parameters of the well-characterized PHD2 isoform comparable with those in published literature. We further showed that it is also sensitive enough to detect and measure the IC50 values of pan-PHD inhibitors and several PHD2 inhibitors in clinical trials for chronic kidney disease (CKD)-induced anemia. Given the efficiency of this assay coupled with its multiwell format, the 2,4-DNPH α-KG assay may be adaptable to explore non-HIF substrates of PHDs and potentially to high-throughput assays.


Subject(s)
Colorimetry/methods , Hypoxia-Inducible Factor-Proline Dioxygenases/analysis , Ketoglutaric Acids/analysis , Phenylhydrazines/chemistry , Enzyme Assays/methods , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Ketoglutaric Acids/chemistry , Kinetics , Substrate Specificity
4.
J Clin Gastroenterol ; 56(3): 218-223, 2022 03 01.
Article in English | MEDLINE | ID: mdl-33731598

ABSTRACT

GOALS: The aim was to measure bile acids in human saliva using a sensitive ultraperformance liquid chromatography tandem mass spectrometry analysis method to distinguish quantitative differences in refractory gastroesophageal reflux disease (GERD) patients as compared with proton pump inhibitor (PPI) controlled GERD patients and healthy volunteers. STUDY: Human saliva samples were analyzed from 2 separate studies. The first a meal-controlled pilot, in which premeal and postmeal saliva samples were analyzed from 20 healthy subjects and 20 patients with GERD symptoms controlled by PPIs. In a subsequent exploratory study, saliva was collected from 34 patients with continuing GERD symptoms despite PPI treatment (refractory GERD), 30 healthy subjects, and 30 PPI-controlled GERD patients at ≥4 hours postmeal. RESULTS: In the meal-controlled pilot study, both healthy subjects and patients with PPI-controlled GERD, had total saliva bile acid increase for the first hour after consumption of a meal and returned to baseline levels 4 hours later. There was no difference in bile acid levels between the 2 groups. In the exploratory study, the saliva from patients with refractory GERD had statistically significant higher levels of total bile acid concentration compared with those of healthy volunteers and patients with PPI-controlled GERD (P=0.0181). CONCLUSIONS: Bile acids can be detected and accurately quantitated in human saliva using a sensitive ultraperformance liquid chromatography tandem mass spectrometry assay. Increases above threshold could indicate an underlying disease.This method could potentially be used to evaluate biliary reflux as an underlying pathophysiology of refractory GERD.


Subject(s)
Gastroesophageal Reflux , Saliva , Bile Acids and Salts , Chromatography, Liquid , Gastroesophageal Reflux/diagnosis , Humans , Pilot Projects , Proton Pump Inhibitors , Tandem Mass Spectrometry , Treatment Outcome
5.
Proc Natl Acad Sci U S A ; 116(22): 11057-11062, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31085647

ABSTRACT

Endothelial dysfunction and reduced nitric oxide (NO) signaling are a key element of the pathophysiology of nonalcoholic steatohepatitis (NASH). Stimulators of soluble guanylate cyclase (sGC) enhance NO signaling; have been shown preclinically to reduce inflammation, fibrosis, and steatosis; and thus have been proposed as potential therapies for NASH and fibrotic liver diseases. Praliciguat, an oral sGC stimulator with extensive distribution to the liver, was used to explore the role of this signaling pathway in NASH. We found that sGC is expressed in hepatic stellate cells and stellate-derived myofibroblasts, but not in hepatocytes. Praliciguat acted directly on isolated hepatic stellate cells to inhibit fibrotic and inflammatory signaling potentially through regulation of AMPK and SMAD7. Using in vivo microdialysis, we demonstrated stimulation of the NO-sGC pathway by praliciguat in both healthy and fibrotic livers. In preclinical models of NASH, praliciguat treatment was associated with lower levels of liver fibrosis and lower expression of fibrotic and inflammatory biomarkers. Praliciguat treatment lowered hepatic steatosis and plasma cholesterol levels. The antiinflammatory and antifibrotic effects of praliciguat were recapitulated in human microtissues in vitro. These data provide a plausible cellular basis for the mechanism of action of sGC stimulators and suggest the potential therapeutic utility of praliciguat in the treatment of NASH.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Enzyme Activators/pharmacology , Hepatic Stellate Cells/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Soluble Guanylyl Cyclase , Animals , Anti-Inflammatory Agents/therapeutic use , Cells, Cultured , Coculture Techniques , Humans , Mice , Nitric Oxide/metabolism , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Signal Transduction/drug effects , Soluble Guanylyl Cyclase/drug effects , Soluble Guanylyl Cyclase/metabolism
6.
Gastroenterology ; 158(8): 2093-2103, 2020 06.
Article in English | MEDLINE | ID: mdl-32092310

ABSTRACT

BACKGROUND & AIMS: Refractory gastroesophageal reflux disease (GERD) reduces quality of life and creates significant financial burden on the health care system. Approximately 30% of patients with GERD who receive label-dose proton pump inhibitors (PPIs) still have symptoms. We performed a trial to evaluate the efficacy and safety of IW-3718, a bile acid sequestrant, as an adjunct to PPI therapy. METHODS: We performed a multicenter, double-blind, placebo-controlled trial, from March 2016 through April 2017, of 280 patients with confirmed GERD. The patients, stratified by esophagitis status, were randomly assigned (1:1:1:1) to groups given placebo or IW-3718 (500, 1000, or 1500 mg) twice daily, with ongoing label-dose PPI. The primary endpoint was percent change from baseline to week 8 in weekly heartburn severity score. We also analyzed percent change from baseline to week 8 in weekly regurgitation frequency score. RESULTS: Mean changes from baseline to week 8 in weekly heartburn severity scores were reductions of 46.0% in the placebo group, 49.0% in the 500 mg group, 55.1% in the 1000 mg group, and 58.0% in the 1500 mg IW-3718 group (dose-response P = .02). The treatment difference was 11.9% between the 1500 mg IW-3718 and placebo groups (P = .04, analysis of covariance). The mean change in weekly regurgitation frequency score from baseline to week 8 in the 1500 mg IW-3718 vs placebo groups was a reduction of 17.5% (95% confidence interval, reductions of 31.4% to 3.6%). The most common adverse event was constipation (in 8.1% of patients receiving IW-3718 and 7.1% of patients receiving placebo). There were no drug-related serious adverse events. CONCLUSIONS: In a randomized trial of patients with refractory GERD, adding 1500 mg IW-3718 to label-dose PPIs significantly reduced heartburn symptoms compared with adding placebo. Regurgitation symptoms also decreased. IW-3718 was well tolerated. (ClinicalTrials.gov, Number: NCT02637557).


Subject(s)
Bile Acids and Salts/metabolism , Colesevelam Hydrochloride/administration & dosage , Esophagitis/drug therapy , Gastroesophageal Reflux/drug therapy , Heartburn/drug therapy , Proton Pump Inhibitors/therapeutic use , Adult , Aged , Aged, 80 and over , Colesevelam Hydrochloride/adverse effects , Colesevelam Hydrochloride/metabolism , Delayed-Action Preparations , Double-Blind Method , Drug Therapy, Combination , Esophagitis/diagnosis , Esophagitis/metabolism , Female , Gastroesophageal Reflux/diagnosis , Gastroesophageal Reflux/metabolism , Heartburn/diagnosis , Heartburn/metabolism , Humans , Male , Middle Aged , Proton Pump Inhibitors/adverse effects , Remission Induction , Severity of Illness Index , Time Factors , Treatment Outcome , United States , Young Adult
7.
J Neuroinflammation ; 18(1): 213, 2021 Sep 18.
Article in English | MEDLINE | ID: mdl-34537066

ABSTRACT

BACKGROUND: Inflammation in the central nervous system (CNS) is observed in many neurological disorders. Nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) signaling plays an essential role in modulating neuroinflammation. CYR119 is a CNS-penetrant sGC stimulator that amplifies endogenous NO-sGC-cGMP signaling. We evaluated target engagement and the effects of CYR119 on markers of neuroinflammation in vitro in mouse microglial cells and in vivo in quinolinic acid (QA)-induced and high-fat diet-induced rodent neuroinflammation models. METHODS: Target engagement was verified in human embryonic kidney (HEK) cells, rat primary neurons, mouse SIM-A9 cells, and in rats by measuring changes in cGMP and downstream targets of sGC signaling [phosphorylated vasodilator-stimulated phosphoprotein (pVASP), phosphorylated cAMP-response element binding (pCREB)]. In SIM-A9 cells stimulated with lipopolysaccharides (LPS), markers of inflammation were measured when cells were treated with or without CYR119. In rats, microinjections of QA and vehicle were administered into the right and left hemispheres of striatum, respectively, and then rats were dosed daily with either CYR119 (10 mg/kg) or vehicle for 7 days. The activation of microglia [ionized calcium binding adaptor molecule 1 (Iba1)] and astrocytes [glial fibrillary acidic protein (GFAP)] was measured by immunohistochemistry. Diet-induced obese (DIO) mice were treated daily with CYR119 (10 mg/kg) for 6 weeks, after which inflammatory genetic markers were analyzed in the prefrontal cortex. RESULTS: In vitro, CYR119 synergized with exogenous NO to increase the production of cGMP in HEK cells and in primary rat neuronal cell cultures. In primary neurons, CYR119 stimulated sGC, resulting in accumulation of cGMP and phosphorylation of CREB, likely through the activation of protein kinase G (PKG). CYR119 attenuated LPS-induced elevation of interleukin 6 (IL-6) and tumor necrosis factor (TNF) in mouse microglial cells. Following oral dosing in rats, CYR119 crossed the blood-brain barrier (BBB) and stimulated an increase in cGMP levels in the cerebral spinal fluid (CSF). In addition, levels of proinflammatory markers associated with QA administration or high-fat diet feeding were lower in rodents treated with CYR119 than in those treated with vehicle. CONCLUSIONS: These data suggest that sGC stimulation could provide neuroprotective effects by attenuating inflammatory responses in nonclinical models of neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/metabolism , Central Nervous System/metabolism , Cyclic GMP/metabolism , Inflammation Mediators/metabolism , Neurons/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers/metabolism , Cells, Cultured , Central Nervous System/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Rats , Rats, Sprague-Dawley
8.
Diabetologia ; 63(4): 733-743, 2020 04.
Article in English | MEDLINE | ID: mdl-31858186

ABSTRACT

AIMS/HYPOTHESIS: Praliciguat (IW-1973), a soluble guanylate cyclase stimulator, amplifies nitric oxide signalling. This exploratory trial investigated the safety, tolerability, pharmacokinetic profile and pharmacodynamic effects of praliciguat in individuals with type 2 diabetes and hypertension. METHODS: This Phase IIA, double-blind, placebo-controlled trial investigated praliciguat in 26 participants with type 2 diabetes and hypertension on stable glucose- and BP-lowering therapies. Participants were randomly allocated in a 3:5:5 ratio to three groups: placebo (n = 6), praliciguat 40 mg once daily for days 1-14 (n = 10), or praliciguat 20 mg twice daily for days 1-7 then 40 mg once daily for days 8-14 (n = 10). Assessments were made in clinic and included treatment-emergent adverse events, pharmacokinetics, metabolic variables, 24 h BP and heart rate, platelet function, reactive hyperaemia index (RHI) and plasma biomarkers. Participants, the sponsor, the investigator and clinic study staff (except designated pharmacy personnel) were blinded to group assignment. RESULTS: Participants treated for 14 days with praliciguat had least-square mean change-from-baseline differences vs placebo (95% CI) of -0.7 (-1.8, 0.4) mmol/l for fasting plasma glucose, -0.7 (-1.1, -0.2) mmol/l for total cholesterol, -0.5 (-1.0, -0.1) mmol/l for LDL-cholesterol, -23 (-56, 9) for HOMA-IR in those not being treated with insulin, and -5 (-10, 1) mmHg and 3 (-1, 6) beats/min for average 24 h mean arterial pressure and heart rate, respectively. Apart from one serious adverse event (SAE; upper gastrointestinal haemorrhage), praliciguat was well tolerated. Praliciguat did not affect platelet function or RHI. Among exploratory biomarkers, plasma levels of asymmetric dimethylarginine decreased in praliciguat vs placebo recipients. CONCLUSIONS/INTERPRETATION: In participants with type 2 diabetes and hypertension on standard therapies, over 14 days praliciguat was well tolerated, except for a single SAE, and showed positive trends in metabolic and BP variables. These results support further clinical investigation of praliciguat. TRIAL REGISTRATION: ClinicalTrials.gov NCT03091920. FUNDING: This trial was funded by Cyclerion Therapeutics.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Hypertension/drug therapy , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Adult , Aged , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Diabetic Nephropathies/prevention & control , Double-Blind Method , Drug Therapy, Combination , Female , Guanylyl Cyclase C Agonists/pharmacokinetics , Guanylyl Cyclase C Agonists/therapeutic use , Humans , Hypertension/complications , Hypertension/metabolism , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Insulin/administration & dosage , Insulin/adverse effects , Male , Middle Aged , Treatment Outcome
9.
Am J Physiol Renal Physiol ; 318(1): F148-F159, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31608671

ABSTRACT

Reduced nitric oxide (NO) and a decrease in cGMP signaling mediated by soluble guanylate cyclase (sGC) has been linked to the development of several cardiorenal diseases. Stimulation of sGC is a potential means for enhancing cGMP production in conditions of reduced NO bioavailability. The purpose of our studies was to determine the effects of praliciguat, a clinical-stage sGC stimulator, in a model of cardiorenal failure. Dahl salt-sensitive rats fed a high-salt diet to induce hypertension and organ damage were treated with the sGC stimulator praliciguat to determine its effects on hemodynamics, biomarkers of inflammation, fibrosis, tissue function, and organ damage. Praliciguat treatment reduced blood pressure, improved cardiorenal damage, and attenuated the increase in circulating markers of inflammation and fibrosis. Notably, praliciguat affected markers of renal damage at a dose that had minimal effect on blood pressure. In addition, liver fibrosis and circulating markers of tissue damage were attenuated in praliciguat-treated rats. Stimulation of the NO-sGC-cGMP pathway by praliciguat attenuated or normalized indicators of chronic inflammation, fibrosis, and tissue dysfunction in the Dahl salt-sensitive rat model. Stimulation of sGC by praliciguat may present an effective mechanism for treating diseases linked to NO deficiency, particularly those associated with cardiac and renal failure. Praliciguat is currently being evaluated in patients with diabetic nephropathy and heart failure with preserved ejection fraction.


Subject(s)
Fibrosis/drug therapy , Guanylyl Cyclase C Agonists/pharmacology , Inflammation/drug therapy , Kidney/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Renal Insufficiency/drug therapy , Animals , Biomarkers/blood , Blood Pressure/drug effects , Chemokine CCL2/blood , Cyclic GMP/metabolism , Fibrosis/pathology , Guanylyl Cyclase C Agonists/therapeutic use , Inflammation/pathology , Kidney/pathology , Male , Natriuretic Peptide, Brain/blood , Nitric Oxide/metabolism , Osteopontin/blood , Peptide Fragments/blood , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Rats , Rats, Inbred Dahl , Renal Insufficiency/pathology , Signal Transduction/drug effects , Soluble Guanylyl Cyclase/metabolism , Tissue Inhibitor of Metalloproteinase-1/blood
10.
Am J Physiol Renal Physiol ; 319(4): F697-F711, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32865013

ABSTRACT

Praliciguat, a clinical-stage soluble guanylate cyclase (sGC) stimulator, increases cGMP via the nitric oxide-sGC pathway. Praliciguat has been shown to be renoprotective in rodent models of hypertensive nephropathy and renal fibrosis. In the present study, praliciguat alone and in combination with enalapril attenuated proteinuria in the obese ZSF1 rat model of diabetic nephropathy. Praliciguat monotherapy did not affect hemodynamics. In contrast, enalapril monotherapy lowered blood pressure but did not attenuate proteinuria. Renal expression of genes in pathways involved in inflammation, fibrosis, oxidative stress, and kidney injury was lower in praliciguat-treated obese ZSF1 rats than in obese control rats; fasting glucose and cholesterol were also lower with praliciguat treatment. To gain insight into how tubular mechanisms might contribute to its pharmacological effects on the kidneys, we studied the effects of praliciguat on pathological processes and signaling pathways in cultured human primary renal proximal tubular epithelial cells (RPTCs). Praliciguat inhibited the expression of proinflammatory cytokines and secretion of monocyte chemoattractant protein-1 in tumor necrosis factor-α-challenged RPTCs. Praliciguat treatment also attenuated transforming growth factor-ß-mediated apoptosis, changes to a mesenchyme-like cellular phenotype, and phosphorylation of SMAD3 in RPTCs. In conclusion, praliciguat improved proteinuria in the ZSF1 rat model of diabetic nephropathy, and its actions in human RPTCs suggest that tubular effects may contribute to its renal benefits, building upon strong evidence for the role of cGMP signaling in renal health.


Subject(s)
Apoptosis/drug effects , Diabetic Nephropathies/drug therapy , Guanylyl Cyclase C Agonists/pharmacology , Kidney Tubules, Proximal/drug effects , Nephritis/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Cell Line , Cytokines/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Models, Animal , Disease Progression , Enalapril/pharmacology , Humans , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Nephritis/metabolism , Nephritis/pathology , Phosphorylation , Rats, Zucker , Signal Transduction , Smad3 Protein/metabolism
11.
RNA ; 23(3): 346-354, 2017 03.
Article in English | MEDLINE | ID: mdl-27932585

ABSTRACT

S-adenosylmethionine (SAM)-dependent methyltransferases regulate a wide range of biological processes through the modification of proteins, nucleic acids, polysaccharides, as well as various metabolites. TYW3/Taw3 is a SAM-dependent methyltransferase responsible for the formation of a tRNA modification known as wybutosine and its derivatives that are required for accurate decoding in protein synthesis. Here, we report the crystal structure of Taw3, a homolog of TYW3 from Sulfolobus solfataricus, which revealed a novel α/ß fold. The sequence motif (S/T)xSSCxGR and invariant aspartate and histidine, conserved in TYW3/Taw3, cluster to form the catalytic center. These structural and sequence features indicate that TYW3/Taw3 proteins constitute a distinct class of SAM-dependent methyltransferases. Using site-directed mutagenesis along with in vivo complementation assays combined with mass spectrometry as well as ligand docking and cofactor binding assays, we have identified the active site of TYW3 and residues essential for cofactor binding and methyltransferase activity.


Subject(s)
Archaeal Proteins/chemistry , Methyltransferases/chemistry , Nucleosides/chemistry , S-Adenosylmethionine/chemistry , Sulfolobus solfataricus/chemistry , Amino Acid Motifs , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Catalytic Domain , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Kinetics , Methyltransferases/genetics , Methyltransferases/metabolism , Molecular Docking Simulation , Mutagenesis, Site-Directed , Nucleosides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , S-Adenosylmethionine/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Sulfolobus solfataricus/enzymology
12.
Am J Gastroenterol ; 113(1): 105-114, 2018 01.
Article in English | MEDLINE | ID: mdl-29091082

ABSTRACT

OBJECTIVES: Linaclotide is a guanylate cyclase-C agonist approved in the United States, Canada, and Mexico at a once-daily 145-µg dose for the treatment of chronic idiopathic constipation (CIC); a once-daily 72-µg dose for CIC recently received FDA approval. The trial objective was to evaluate the efficacy and safety of a 72-µg linaclotide dose in CIC patients. METHODS: This double-blind, placebo-controlled trial randomized patients with CIC (Rome III criteria) to once-daily linaclotide 72 µg or 145 µg, or placebo for 12 weeks. The primary endpoint, 12-week complete spontaneous bowel movement (CSBM) overall responder, required patients to have ≥3 CSBMs and an increase of ≥1 CSBM per week from baseline in the same week for ≥9 of 12 weeks of the treatment period. Secondary endpoints included 12-week change from baseline in bowel (SBM and CSBM frequency, stool consistency, straining) and abdominal (bloating, discomfort) symptoms, monthly CSBM responders, and 12-week CSBM responders among patients who averaged >1 SBM/week at baseline. Sustained response (12-week CSBM overall responders who met weekly criteria for 3 of the 4 final weeks (weeks 9-12) of treatment) was evaluated as an additional endpoint. Adverse events (AEs) were monitored. RESULTS: The intent-to-treat population included 1,223 patients (mean age=46 years, female=77%, white=71%). The primary endpoint was met by 13.4% of linaclotide 72-µg patients vs. 4.7% of placebo patients (P<0.0001, odds ratio=3.0; statistically significant controlling for multiplicity). Sustained response was achieved by 12.4% of linaclotide 72-µg patients vs. 4.2% of placebo patients (nominal P<0.0001). Linaclotide 72-µg patients met 9-of-10 secondary endpoints vs. placebo (P<0.05; abdominal discomfort, P=0.1028). Patients treated with linaclotide 145 µg also improved CIC symptoms for the primary (12.4%) and sustained responder endpoint parameters (11.4%) and for all 10 of the secondary endpoint parameters including abdominal discomfort (P<0.05). Diarrhea, the most common AE, was mild in most instances and resulted in discontinuation of 0, 2.4%, and 3.2% of patients in the placebo, linaclotide 72-µg, and linaclotide 145-µg groups, respectively. CONCLUSIONS: Once-daily linaclotide 72 µg significantly improved CIC symptoms in both men and women with a low rate of discontinuation due to diarrhea over 12 weeks of treatment.


Subject(s)
Constipation/drug therapy , Guanylyl Cyclase C Agonists/administration & dosage , Peptides/administration & dosage , Adult , Aged , Chronic Disease , Defecation , Diarrhea/chemically induced , Double-Blind Method , Female , Guanylyl Cyclase C Agonists/therapeutic use , Humans , Male , Medication Adherence , Middle Aged , Peptides/therapeutic use , Treatment Outcome
13.
J Pharmacol Exp Ther ; 365(3): 664-675, 2018 06.
Article in English | MEDLINE | ID: mdl-29643251

ABSTRACT

Soluble guanylate cyclase (sGC), a key signal-transduction enzyme, increases the conversion of guanosine-5'-triphosphate to cGMP upon binding of nitric oxide (NO). Endothelial dysfunction and/or reduced NO signaling have been implicated in cardiovascular disease pathogenesis and complications of diabetes and have been associated with other disease states and aging. Soluble guanylate cyclase (sGC) stimulators are small-molecule drugs that bind sGC and enhance NO-mediated cGMP signaling. The pharmacological characterization of IW-1973 [1,1,1,3,3,3-hexafluoro-2-(((5-fluoro-2-(1-(2-fluorobenzyl)-5-(isoxazol-3-yl)-1H-pyrazol-3-yl) pyrimidin-4-yl)amino)methyl)propan-2-ol], a novel clinical-stage sGC stimulator under clinical investigation for treatment of heart failure with preserved ejection fraction and diabetic nephropathy, is described. In the presence of NO, IW-1973 stimulated sGC in a human purified enzyme assay and a HEK-293 whole cell assay. sGC stimulation by IW-1973 in cells was associated with increased phosphorylation of vasodilator-stimulated phosphoprotein. IW-1973, at doses of 1-10 mg/kg, significantly lowered blood pressure in normotensive and spontaneously hypertensive rats. In a Dahl salt-sensitive hypertension model, IW-1973 significantly reduced blood pressure, inflammatory cytokine levels, and renal disease markers, including proteinuria and renal fibrotic gene expression. The results were affirmed in mouse lipopolysaccharide-induced inflammation and rat unilateral ureteral obstruction renal fibrosis models. A quantitative whole-body autoradiography study of IW-1973 revealed extensive tissue distribution and pharmacokinetic studies showed a large volume of distribution and a profile consistent with predicted once-a-day dosing in humans. In summary, IW-1973 is a potent, orally available sGC stimulator that exhibits renoprotective, anti-inflammatory, and antifibrotic effects in nonclinical models.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/pharmacokinetics , Antihypertensive Agents/pharmacology , Antihypertensive Agents/pharmacokinetics , Pyrazoles/pharmacology , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacology , Pyrimidines/pharmacokinetics , Soluble Guanylyl Cyclase/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Antihypertensive Agents/therapeutic use , Arteries/drug effects , Arteries/physiology , Blood Pressure/drug effects , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Fibrosis , HEK293 Cells , Humans , Kidney/drug effects , Kidney/pathology , Male , Mice , Nitric Oxide/metabolism , Proteinuria/drug therapy , Pyrazoles/therapeutic use , Pyrimidines/therapeutic use , Rats , Signal Transduction/drug effects , Tissue Distribution , Vasodilation/drug effects
14.
J Gastroenterol Hepatol ; 33(5): 980-989, 2018 May.
Article in English | MEDLINE | ID: mdl-29319191

ABSTRACT

BACKGROUND AND AIM: Linaclotide is a guanylate cyclase-C agonist approved in multiple countries to treat irritable bowel syndrome with constipation (IBS-C). China has unmet need for well-tolerated therapy that is effective in treating both bowel and abdominal symptoms of IBS-C. This trial evaluated linaclotide's efficacy and safety in IBS-C patients in China and other regions. METHODS: This Phase 3, double-blind trial randomized IBS-C patients to once-daily oral 290-µg linaclotide or placebo at centers in China, North America, and Oceania. Patients reported bowel and abdominal symptoms daily; adverse events were monitored. Co-primary and secondary endpoints were tested using a predefined three-step serial gatekeeping multiple comparisons procedure. RESULTS: The intent-to-treat population included 839 patients (mean age = 41 years; 82% female; 81% Asian). The trial met all co-primary and secondary endpoints. Co-primary responder criteria were met by 60.0% of linaclotide patients versus 48.8% of placebo patients for abdominal pain/discomfort (≥ 30% decrease for ≥ 6/12 weeks; P < 0.05), and 31.7% of linaclotide versus 15.4% of placebo patients for IBS degree of relief (score ≤ 2 for ≥ 6/12 weeks; P < 0.0001). Secondary 12-week change-from-baseline endpoints (spontaneous bowel movement/complete spontaneous bowel movement frequency, stool consistency, straining, abdominal pain, abdominal discomfort, and abdominal bloating) were significantly improved with linaclotide versus placebo (all P < 0.0001). Diarrhea was the most common adverse event (9.4% linaclotide, 1.2% placebo). Discontinuation rates due to diarrhea were low (0.7% linaclotide, 0.2% placebo). CONCLUSIONS: Once-daily 290-µg linaclotide improved bowel habits, abdominal symptoms, and global measures in a predominantly Chinese IBS-C population.


Subject(s)
Constipation/drug therapy , Constipation/etiology , Guanylyl Cyclase C Agonists/administration & dosage , Irritable Bowel Syndrome/complications , Irritable Bowel Syndrome/drug therapy , Peptides/administration & dosage , Administration, Oral , Adolescent , Adult , Aged , Aged, 80 and over , Diarrhea/chemically induced , Double-Blind Method , Female , Guanylyl Cyclase C Agonists/adverse effects , Humans , Male , Middle Aged , Peptides/adverse effects , Treatment Outcome , Young Adult
15.
Proc Natl Acad Sci U S A ; 110(21): 8495-500, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23650358

ABSTRACT

The regulated binding of effector proteins to the nucleosome plays a central role in the activation and silencing of eukaryotic genes. How this binding changes the properties of chromatin to mediate gene activation or silencing is not fully understood. Here we provide evidence that association of the budding yeast silent information regulator 3 (Sir3) silencing protein with the nucleosome induces a conformational change in the amino terminus of histone H4 that promotes interactions between the conserved H4 arginines 17 and 19 (R17 and R19) and nucleosomal DNA. Substitutions of H4R17 and R19 with alanine abolish silencing in vivo, but have little or no effect on binding of Sir3 to nucleosomes or histone H4 peptides in vitro. Furthermore, in both the previously reported crystal structure of the Sir3-bromo adjacent homology (BAH) domain bound to the Xenopus laevis nucleosome core particle and the crystal structure of the Sir3-BAH domain bound to the yeast nucleosome core particle described here, H4R17 and R19 make contacts with nucleosomal DNA rather than with Sir3. These results suggest that Sir3 binding generates a more stable nucleosome by clamping H4R17 and R19 to nucleosomal DNA, and raise the possibility that such induced changes in histone-DNA contacts play major roles in the regulation of chromatin structure.


Subject(s)
Chromatin Assembly and Disassembly/physiology , DNA, Fungal/metabolism , Histones/metabolism , Nucleosomes/metabolism , Saccharomyces cerevisiae/metabolism , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Amino Acid Substitution , Animals , DNA, Fungal/chemistry , DNA, Fungal/genetics , Histones/chemistry , Histones/genetics , Mutation, Missense , Nucleosomes/chemistry , Nucleosomes/genetics , Protein Binding , Protein Structure, Tertiary , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Silent Information Regulator Proteins, Saccharomyces cerevisiae/chemistry , Silent Information Regulator Proteins, Saccharomyces cerevisiae/genetics , Xenopus Proteins/chemistry , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis
16.
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
17.
J Biol Chem ; 288(11): 7978-7985, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23341454

ABSTRACT

Clostridium thermocellum produces the prototypical cellulosome, a large multienzyme complex that efficiently hydrolyzes plant cell wall polysaccharides into fermentable sugars. This ability has garnered great interest in its potential application in biofuel production. The core non-catalytic scaffoldin subunit, CipA, bears nine type I cohesin modules that interact with the type I dockerin modules of secreted hydrolytic enzymes and promotes catalytic synergy. Because the large size and flexibility of the cellulosome preclude structural determination by traditional means, the structural basis of this synergy remains unclear. Small angle x-ray scattering has been successfully applied to the study of flexible proteins. Here, we used small angle x-ray scattering to determine the solution structure and to analyze the conformational flexibility of two overlapping N-terminal cellulosomal scaffoldin fragments comprising two type I cohesin modules and the cellulose-specific carbohydrate-binding module from CipA in complex with Cel8A cellulases. The pair distribution functions, ab initio envelopes, and rigid body models generated for these two complexes reveal extended structures. These two N-terminal cellulosomal fragments are highly dynamic and display no preference for extended or compact conformations. Overall, our work reveals structural and dynamic features of the N terminus of the CipA scaffoldin that may aid in cellulosome substrate recognition and binding.


Subject(s)
Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Cellulase/chemistry , Clostridium thermocellum/metabolism , Multienzyme Complexes/chemistry , Cellulase/metabolism , Crystallography, X-Ray/methods , Models, Molecular , Molecular Conformation , Multienzyme Complexes/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Scattering, Radiation , Scattering, Small Angle , Substrate Specificity , X-Rays
18.
N Engl J Med ; 365(6): 527-36, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21830967

ABSTRACT

BACKGROUND: Linaclotide is a minimally absorbed peptide agonist of the guanylate cyclase C receptor. In two trials, we aimed to determine the efficacy and safety of linaclotide in patients with chronic constipation. METHODS: We conducted two randomized, 12-week, multicenter, double-blind, parallel-group, placebo-controlled, dual-dose trials (Trials 303 and 01) involving 1276 patients with chronic constipation. Patients received either placebo or linaclotide, 145 µg or 290 µg, once daily for 12 weeks. The primary efficacy end point was three or more complete spontaneous bowel movements (CSBMs) per week and an increase of one or more CSBMs from baseline during at least 9 of the 12 weeks. Adverse events were also monitored. RESULTS: For Trials 303 and 01, respectively, the primary end point was reached by 21.2% and 16.0% of the patients who received 145 µg of linaclotide and by 19.4% and 21.3% of the patients who received 290 µg of linaclotide, as compared with 3.3% and 6.0% of those who received placebo (P<0.01 for all comparisons of linaclotide with placebo). Improvements in all secondary end points were significantly greater in both linaclotide groups than in the placebo groups. The incidence of adverse events was similar among all study groups, with the exception of diarrhea, which led to discontinuation of treatment in 4.2% of patients in both linaclotide groups. CONCLUSIONS: In these two 12-week trials, linaclotide significantly reduced bowel and abdominal symptoms in patients with chronic constipation. Additional studies are needed to evaluate the potential long-term risks and benefits of linaclotide in chronic constipation. (Funded by Ironwood Pharmaceuticals and Forest Research Institute; ClinicalTrials.gov numbers, NCT00765882 and NCT00730015.).


Subject(s)
Constipation/drug therapy , Laxatives/therapeutic use , Peptides/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Chronic Disease , Defecation/drug effects , Diarrhea/chemically induced , Double-Blind Method , Female , Guanylate Cyclase , Humans , Laxatives/adverse effects , Male , Middle Aged , Peptides/adverse effects , Quality of Life , Receptors, Guanylate Cyclase-Coupled/agonists , Withholding Treatment , Young Adult
19.
Clin Gastroenterol Hepatol ; 12(4): 616-23, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24075889

ABSTRACT

BACKGROUND & AIMS: Patients with irritable bowel syndrome with constipation (IBS-C) have abdominal symptoms that vary in severity. Linaclotide, a guanylate cyclase-C agonist, improves abdominal and bowel symptoms in these patients. We examined the prevalence of severe abdominal symptoms in patients with IBS-C and assessed the effects of linaclotide on abdominal symptoms, global measures, and quality of life (QOL). METHODS: In two phase 3 trials, patients who met modified Rome II criteria for IBS-C were randomly assigned to groups given oral, once-daily linaclotide (290 µg) or placebo for 12 weeks. During the baseline (2 weeks prior to treatment) and treatment periods, patients rated abdominal pain, discomfort, bloating, fullness, and cramping daily (from 0 = none to 10 = very severe). Linaclotide's effects on abdominal symptoms, global measures, and IBS-related QOL were assessed in subpopulations of patients who rated specific individual abdominal symptoms as severe (≥ 7.0) at baseline. RESULTS: In the intent-to-treat population (1602 patients; 797 receiving placebo and 805 receiving linaclotide), baseline prevalence values for severe abdominal symptoms were 44% for bloating, 44% for fullness, 32% for discomfort, 23% for pain, and 22% for cramping, with considerable overlap among symptoms. In patients with severe symptoms, linaclotide reduced all abdominal symptoms; mean changes from baseline severity scores ranged from -2.7 to -3.4 for linaclotide vs -1.4 to -1.9 for placebo (P < .0001). Linaclotide improved global measures (P < .0001) and IBS-QOL scores (P < .01) compared with placebo. Diarrhea was the most common adverse event of linaclotide in patients with severe abdominal symptoms (18.8%-21.0%). CONCLUSIONS: Of 5 severe abdominal symptoms assessed, bloating and fullness were most prevalent in patients with IBS-C. Linaclotide significantly improved all abdominal symptoms, global measures, and IBS-QOL in subpopulations of IBS-C patients with severe abdominal symptoms. Clinicaltrials.gov NUMBERS: NCT00938717, NCT00948818.


Subject(s)
Constipation/drug therapy , Gastrointestinal Agents/administration & dosage , Irritable Bowel Syndrome/complications , Irritable Bowel Syndrome/drug therapy , Peptides/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Placebos/administration & dosage , Treatment Outcome , Young Adult
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL