Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Boris Tchernychev is active.

Publication


Featured researches published by Boris Tchernychev.


Pain | 2013

Gastrointestinal pain: unraveling a novel endogenous pathway through uroguanylin/guanylate cyclase-C/cGMP activation.

Inmaculada Silos-Santiago; Gerhard Hannig; Helene Eutamene; Elena E. Ustinova; Sylvie G. Bernier; Pei Ge; Christopher Graul; Sarah Jacobson; Hong Jin; Elaine Liong; Marco Kessler; Tammi Reza; Samuel Rivers; Courtney Shea; Boris Tchernychev; Alexander P. Bryant; Caroline B. Kurtz; Lionel Bueno; Michael A. Pezzone; Mark G. Currie

Summary Uroguanylin activation of the guanylate cyclase‐C/cyclic guanosine monophosphate pathway elicits analgesic effects in animal models of colonic hypersensitivity, unraveling a novel pathway to treat abdominal pain. 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.


Thrombosis and Haemostasis | 2007

The von Willebrand factor antagonist (GPG-290) prevents coronary thrombosis without prolongation of bleeding time

Michael Wadanoli; Dianne Sako; Gray Shaw; Robert G. Schaub; Qin Wang; Boris Tchernychev; Jin Xu; Thomas J. Porter; Qinheng Huang

The interaction between von Willebrand factor (VWF) and platelet glycoprotein Ibalpha (GPIbalpha) is a critical step that allows platelet adhesion, activation and subsequent thrombus formation to the injured vessel wall under high-shear conditions. In this study, we sought to investigate 1) whether GPG-290, a recombinant human GPIbalpha chimeric protein, would prevent thrombosis in a canine model of coronary thrombosis by blocking VWF-GPIbalpha interaction; and 2) whether desmopressin (DDAVP), a VWF release stimulant, could reduce the prolonged bleeding time caused by a 10x efficacious dose of GPG-290. The antithrombotic efficacy of GPG-290 was evaluated by the in-vivo ability to prevent cyclic flow reductions (CFRs) and ex-vivo inhibition of platelet adhesion/aggregation reflected by prolongation of Platelet Function Analyzer (PFA-100) collagen/ADP closure time. The anti-hemostatic effect was assessed by template bleeding time. GPG-290 at doses of 25, 50 and 100 microg/kg abolished CFRs in 67%, 100% and 100% of the treated dogs without bleeding time prolongation, respectively; GPG-290 dose-dependently prolonged the ex-vivo collagen/ADP-closure time, while it had no effects on plasma VWF antigen level (VWF:Ag) and VWF-collagen binding activity (VWF:CB); the prolonged template bleeding time caused by 500 microg/kg of GPG-290 was prevented by intravenous infusion of DDAVP (0.3 microg/kg). In conclusion, GPG-290 appears to be an effective agent for treating arterial thrombosis without bleeding time prolongation.


Journal of Pharmacology and Experimental Therapeutics | 2007

Characterization of the Novel P-Selectin Inhibitor PSI-697 [2-(4-Chlorobenzyl)-3-hydroxy-7,8,9,10-tetrahydrobenzo[h] Quinoline-4-carboxylic acid] in Vitro and in Rodent Models of Vascular Inflammation and Thrombosis

Patricia W. Bedard; Valerie Clerin; Natalia Sushkova; Boris Tchernychev; Thomas M. Antrilli; Christine Resmini; James C. Keith; James K. Hennan; Neelu Kaila; Silvano DeBernardo; Kristin Janz; Qin Wang; David L. Crandall; Robert G. Schaub; Gray Shaw; Laura L. Carter

P-selectin plays a significant and well documented role in vascular disease by mediating leukocyte and platelet rolling and adhesion. This study characterizes the in vitro activity, pharmacokinetic properties, and the anti-inflammatory and antithrombotic efficacy of the orally active P-selectin small-molecule antagonist PSI-697 [2-(4-chlorobenzyl)-3-hydroxy-7,8,9,10-tetrahydrobenzo[h] quinoline-4-carboxylic acid; molecular mass, 367.83]. Biacore and cell-based assays were used to demonstrate the ability of PSI-697 to dose dependently inhibit the binding of human P-selectin to human P-selectin glycoprotein ligand-1, inhibiting 50% of binding at 50 to 125 μM. The pharmacokinetics of PSI-697 in rats were characterized by low clearance, short half-life, low volume of distribution, and moderate apparent oral bioavailability. A surgical inflammation model, using exteriorized rat cremaster venules, demonstrated that PSI-697 (50 mg/kg p.o.) significantly reduced the number of rolling leukocytes by 39% (P < 0.05) versus vehicle control. In a rat venous thrombosis model, PSI-697 (100 mg/kg p.o.) reduced thrombus weight by 18% (P < 0.05) relative to vehicle, without prolonging bleeding time. Finally, in a rat carotid injury model, PSI-697 (30 or 15 mg/kg p.o.) administered 1 h before arterial injury and once daily thereafter for 13 days resulted in dose-dependent decreases in intima/media ratios of 40.2% (P = 0.025) and 25.7% (P = 0.002) compared with vehicle controls. These data demonstrate the activity of PSI-697 in vitro and after oral administration in animal models of both arterial and venous injury and support the clinical evaluation of this novel antagonist of P-selectin in atherothrombotic and venous thrombotic indications.


Journal of Medicinal Chemistry | 2010

Discovery of 2-[1-(4-Chlorophenyl)cyclopropyl]-3-hydroxy-8-(trifluoromethyl)quinoline-4-carboxylic Acid (PSI-421), a P-Selectin Inhibitor with Improved Pharmacokinetic Properties and Oral Efficacy in Models of Vascular Injury

Adrian Huang; Alessandro Moretto; Kristin Janz; Michael Dennis Lowe; Patricia W. Bedard; Steve Tam; Li Di; Valerie Clerin; Natalia Sushkova; Boris Tchernychev; Desiree H.H. Tsao; James C. Keith; Gray Shaw; Robert G. Schaub; Qin Wang; Neelu Kaila

Previously, we reported the discovery of PSI-697 (1a), a C-2 benzyl substituted quinoline salicylic acid-based P-selectin inhibitor. It is active in a variety of animal models of cardiovascular disease. Compound 1a has also been shown to be well tolerated and safe in healthy volunteers at doses of up to 1200 mg in a phase 1 single ascending dose study. However, its oral bioavailability was low. Our goal was to identify a back up compound with equal potency, increased solubility, and increased exposure. We expanded our structure-activity studies in this series by branching at the alpha position of the C-2 benzyl side chain and through modification of substituents on the carboxylic A-ring of the quinoline. This resulted in discovery of PSI-421 with marked improvement in aqueous solubility and pharmacokinetic properties. This compound has shown oral efficacy in animal models of arterial and venous injury and was selected as a preclinical development compound for potential treatment of such diseases as atherosclerosis and deep vein thrombosis.


Frontiers in Molecular Neuroscience | 2014

Guanylate cyclase-C/cGMP: an emerging pathway in the regulation of visceral pain

Gerhard Hannig; Boris Tchernychev; Caroline B. Kurtz; Alexander P. Bryant; Mark G. Currie; Inmaculada Silos-Santiago

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.


Journal of Pharmacology and Experimental Therapeutics | 2015

MRP4 Modulation of the Guanylate Cyclase-C/cGMP Pathway: Effects on Linaclotide-Induced Electrolyte Secretion and cGMP Efflux

Boris Tchernychev; Pei Ge; Marco Kessler; Robert Solinga; Derek Wachtel; Jenny Tobin; Sara Thomas; Craig E. Lunte; Angelika Fretzen; Gerhard Hannig; Alexander P. Bryant; Caroline B. Kurtz; Mark G. Currie; Inmaculada Silos-Santiago

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.


Physiological Reports | 2017

Linaclotide activates guanylate cyclase-C/cGMP/protein kinase-II-dependent trafficking of CFTR in the intestine

Md. Kaimul Ahsan; Boris Tchernychev; Marco Kessler; Robert Solinga; David Arthur; Cristina I. Linde; Inmaculada Silos-Santiago; Gerhard Hannig; Nadia A. Ameen

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.


BMC Clinical Pharmacology | 2013

Linaclotide induces secretion of cGMP from mouse colonic epithelium

Boris Tchernychev; Sarah Jacobson; Caroline B. Kurtz; Mark G. Currie; Inmaculada Silos-Santiago

Linaclotide is a novel receptor guanylyl cyclase C (GC-C) agonist approved for treatment of abdominal pain and constipation in patients with irritable bowel syndrome with constipation (IBS-C). Linaclotide effects on bowel movements are mediated by the intracellular cGMP that is produced upon activation of GC-C. It is hypothesized that the effects of linaclotide on abdominal pain are mediated by extracellular cGMP, which was shown to decrease the activity of pain-sensing nerves [1]. Here we used an ex vivo Ussing chamber assay to measure the secretion of cGMP from the mouse colonic mucosa in response to linaclotide treatment. Ion transport and epithelial barrier function were monitored by measuring short-circuit current (Isc) and trans-epithelial electrical resistance (TEER). Stimulation with linaclotide (1 µM) elicited a robust short-circuit current across mouse colonic epithelium. Isc reached a maximum within ten minutes following stimulation with linaclotide and remained steady during the duration of the study (60 min). Treatment of colonic mucosa with linaclotide induced release of cGMP from the apical, as well as, the basolateral side of the epithelium. The time course of cGMP accumulation in the basolateral bath of the Ussing chamber was linear with an estimated cGMP secretion rate equal to 23 fmol/min×cm2. The trans-epithelial electrical resistance of the colonic mucosa remained high over the course of the study indicating that the barrier to diffusion of cGMP between apical and basolateral sides remained intact throughout the study.. In summary, these data demonstrate that linaclotide-stimulated mouse colonic epithelium secretes cGMP from both the apical and basolateral sides and that cGMP is available in the submucosal interstitial space to inhibit colonic nociceptors.


BMC Clinical Pharmacology | 2015

Linaclotide-induced electrolyte secretion in human and rat colon: Ussing chamber studies

Boris Tchernychev; Gerhard Hannig; David Arthur; Pei Ge; Jenny Tobin; Inmaculada Silos-Santiago

Background Linaclotide is a guanylyl cyclase C (GC-C) agonist approved for treatment of adult patients with irritable bowel syndrome with constipation (IBS-C). Linaclotide binding to the extracellular receptor domain of GC-C activates the intracellular catalytic domain of GC-C, resulting in the generation of cyclic GMP (cGMP). cGMP, via a pathway leading to activation of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel, stimulates Cland HCO3-secretion and concomitant inhibition of Na+ absorption by sodium/hydrogen exchanger 3 (NHE3), driving the efflux of water into the lumen and accelerating bowel transit. However, the effect of linaclotide on electrolyte transport in the lower gastrointestinal tract has not yet been characterized. This study specifically examined the effect of linaclotide on electrolyte transport in the colon. We used an in vitro Ussing chamber assay to measure the secretion of electrolytes in human ascending colon segments and in rat proximal colon in response to linaclotide treatment.


Gastroenterology | 2014

Mo1752 Linaclotide Induces Endocytosis of the Sodium/Hydrogen Exchanger 3 (NHE3) and Inhibits Sodium Absorption

Nadia A. Ameen; Robert L. Jakab; Gerhard Hannig; Boris Tchernychev; David Arthur; Inmaculada Silos-Santiago

Collaboration


Dive into the Boris Tchernychev's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerhard Hannig

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Kessler

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Mark G. Currie

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Pei Ge

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qin Wang

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Solinga

Ironwood Pharmaceuticals

View shared research outputs
Researchain Logo
Decentralizing Knowledge