Network


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

Hotspot


Dive into the research topics where Robert Solinga is active.

Publication


Featured researches published by Robert Solinga.


Journal of Pharmacology and Experimental Therapeutics | 2013

Pharmacologic Properties, Metabolism, and Disposition of Linaclotide, a Novel Therapeutic Peptide Approved for the Treatment of Irritable Bowel Syndrome with Constipation and Chronic Idiopathic Constipation

Robert W. Busby; Marco Kessler; Wilmin Bartolini; Alexander P. Bryant; Gerhard Hannig; Carolyn S. Higgins; Robert Solinga; Jenny Tobin; Caroline B. Kurtz; Mark G. Currie

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.


The Journal of Neuroscience | 2013

Activation of Guanylate Cyclase-C Attenuates Stretch Responses and Sensitization of Mouse Colorectal Afferents

Bin Feng; Michael E. Kiyatkin; Jun Ho La; Pei Ge; Robert Solinga; Inmaculada Silos-Santiago; G.F. Gebhart

Irritable bowel syndrome (IBS) is characterized by altered bowel habits, persistent pain and discomfort, and typically colorectal hypersensitivity. Linaclotide, a peripherally restricted 14 aa peptide approved for the treatment of IBS with constipation, relieves constipation and reduces IBS-associated pain in these patients presumably by activation of guanylate cyclase-C (GC-C), which stimulates production and release of cyclic guanosine monophosphate (cGMP) from intestinal epithelial cells. We investigated whether activation of GC-C by the endogenous agonist uroguanylin or the primary downstream effector of that activation, cGMP, directly modulates responses and sensitization of mechanosensitive colorectal primary afferents. The distal 2 cm of mouse colorectum with attached pelvic nerve was harvested and pinned flat mucosal side up for in vitro single-fiber recordings, and the encoding properties of mechanosensitive afferents (serosal, mucosal, muscular, and muscular–mucosal; M/M) to probing and circumferential stretch studied. Both cGMP (10–300 μm) and uroguanylin (1–1000 nm) applied directly to colorectal receptive endings significantly reduced responses of muscular and M/M afferents to stretch; serosal and mucosal afferents were not affected. Sensitized responses (i.e., increased responses to stretch) of muscular and M/M afferents were reversed by cGMP, returning responses to stretch to control. Blocking the transport of cGMP from colorectal epithelia by probenecid, a mechanism validated by studies in cultured intestinal T84 cells, abolished the inhibitory effect of uroguanylin on M/M afferents. These results suggest that GC-C agonists like linaclotide alleviate colorectal pain and hypersensitivity by dampening stretch-sensitive afferent mechanosensitivity and normalizing afferent sensitization.


Hypertension | 2009

The Natriuretic Peptide Uroguanylin Elicits Physiologic Actions Through 2 Distinct Topoisomers

Nicholas G. Moss; Dorothy A. Riguera; Robert Solinga; Marco Kessler; Daniel P. Zimmer; William J. Arendshorst; Mark G. Currie; Michael F. Goy

The peptide uroguanylin regulates electrolyte transport in the intestine and kidney. Human uroguanylin has 2 conformations that can be stably isolated because of their slow interconversion rate. The A isomer potently activates the guanylate cyclase C receptor found primarily in the intestine. The B isomer, by contrast, is a very weak agonist of this receptor, leading to a widely held assumption that it is physiologically irrelevant. We show here, however, that human uroguanylin B has potent natriuretic activity in the kidney. Interestingly, uroguanylin A and B both induce saluretic responses, but the activity profiles for the 2 peptides differ markedly. The uroguanylin B dose-response curve is sigmoidal with a threshold dose of ≈10 nmol/kg of body weight, whereas uroguanylin A has a comparable threshold but a bell-shaped dose-response curve. In addition, our study indicates a unique interplay between the A and B isoforms, such that the A form at high concentrations antagonizes the natriuretic action of the B form. These data show that the kidney contains a uroguanylin receptor of which the pharmacological profile does not match that of the well-defined intestinal uroguanylin receptor (guanylate cyclase C), an observation consistent with previous studies showing that the kidney of the guanylate cyclase C knockout mouse remains responsive to uroguanylin. The results presented here also support the unconventional notion that distinct conformations of a single endocrine peptide can elicit different responses in different tissues.


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.


Journal of Biological Chemistry | 2017

Discovery of stimulator binding to a conserved pocket in the heme domain of soluble guanylyl cyclase

Jessica A. Wales; Cheng Yu Chen; Linda Breci; Andrzej Weichsel; Sylvie G. Bernier; James E. Sheppeck; Robert Solinga; Takashi Nakai; Paul Allan Renhowe; Joon Jung; William R. Montfort

Soluble guanylyl cyclase (sGC) is the receptor for nitric oxide and a highly sought-after therapeutic target for the management of cardiovascular diseases. New compounds that stimulate sGC show clinical promise, but where these stimulator compounds bind and how they function remains unknown. Here, using a photolyzable diazirine derivative of a novel stimulator compound, IWP-051, and MS analysis, we localized drug binding to the β1 heme domain of sGC proteins from the hawkmoth Manduca sexta and from human. Covalent attachments to the stimulator were also identified in bacterial homologs of the sGC heme domain, referred to as H-NOX domains, including those from Nostoc sp. PCC 7120, Shewanella oneidensis, Shewanella woodyi, and Clostridium botulinum, indicating that the binding site is highly conserved. The identification of photoaffinity-labeled peptides was aided by a signature MS fragmentation pattern of general applicability for unequivocal identification of covalently attached compounds. Using NMR, we also examined stimulator binding to sGC from M. sexta and bacterial H-NOX homologs. These data indicated that stimulators bind to a conserved cleft between two subdomains in the sGC heme domain. L12W/T48W substitutions within the binding pocket resulted in a 9-fold decrease in drug response, suggesting that the bulkier tryptophan residues directly block stimulator binding. The localization of stimulator binding to the sGC heme domain reported here resolves the longstanding question of where stimulators bind and provides a path forward for drug discovery.


Gastroenterology | 2015

Tu1863 Linaclotide Induces Secretion of cGMP Into the Colonic Submucosal Layer: An In Vivo Microdialysis Study in Rats

Jenny Tobin; Sara Thomas; Robert Solinga; Jaime L. Masferrer; Craig E. Lunte; Inmaculada Silos-Santiago

Figure 1: Physiologic response to intraluminal BAC{BR}A) Fecal output decreased significantly in the days after BAC exposure but then recovered toward baseline output. Output was significantly more depressed in mice treated with 10% BAC compared to 5% BAC on postexposure days 2 and 3. B) Body weight also decreased in a similar fashion but then trended towards recovery. (Note: the N of mice treated with 10% BAC changed due to illness or excessive weight loss). *: p<0.05 when compared to PBS group. ‡: p<0.05 when compared to BAC 5% group. Error bars are standard error.


BMC Clinical Pharmacology | 2013

Determination of cGMP levels in rodent tissues following oral dosing of a soluble guanylate cyclase stimulator

Nisha Perez; Christopher Graul; Peter Germano; Erik Solberg; Samuel Rivers; Robert Solinga; Joel Moore; Gerhard Hannig; Ada Silos-Santiago; Robert W. Busby; Daniel P. Zimmer

Background In the vasculature, nitric oxide (NO) binds and activates smooth muscle soluble guanylate cyclase (sGC), leading to increased intracellular cGMP, which triggers smooth muscle relaxation and vasodilation. sGC stimulators are a class of small molecule allosteric modulators, which stimulate cGMP production independently of NO but also act in synergy with NO. Evidence to date suggests that sGC stimulators may be balanced vasodilators, meaning that they elicit vasorelaxation in both the arterial and venous vasculature; however, there have been conflicting reports [1,2]. Our approach to developing a better understanding of the


European Journal of Pharmacology | 2010

Linaclotide, through activation of guanylate cyclase C, acts locally in the gastrointestinal tract to elicit enhanced intestinal secretion and transit.

Robert W. Busby; Alexander P. Bryant; Wilmin Bartolini; Etchell A. Cordero; Gerhard Hannig; Marco Kessler; Shalina Mahajan-Miklos; Christine M. Pierce; Robert Solinga; Li Jing Sun; Jenny Tobin; Caroline B. Kurtz; Mark G. Currie


Life Sciences | 2010

Linaclotide is a potent and selective guanylate cyclase C agonist that elicits pharmacological effects locally in the gastrointestinal tract

Alexander P. Bryant; Robert W. Busby; Wilmin Bartolini; Etchell A. Cordero; Gerhard Hannig; Marco Kessler; Christine M. Pierce; Robert Solinga; Jenny Tobin; Shalina Mahajan-Miklos; Mitchell B. Cohen; Caroline B. Kurtz; Mark G. Currie

Collaboration


Dive into the Robert Solinga's collaboration.

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

Gerhard Hannig

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jenny Tobin

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge