Network


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

Hotspot


Dive into the research topics where Marco Kessler is active.

Publication


Featured researches published by Marco Kessler.


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.


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.


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.


Investigative Ophthalmology & Visual Science | 2016

The Soluble Guanylate Cyclase Stimulator IWP-953 Increases Conventional Outflow Facility in Mouse Eyes.

Pei Ge; Iris Navarro; Marco Kessler; Sylvie G. Bernier; Nicholas Robert Perl; Renee Sarno; Jaime Masferrer; Gerhard Hannig; W. Daniel Stamer

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.


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.


JCI insight | 2017

Guanylate cyclase 2C agonism corrects CFTR mutants

Kavisha Arora; Yunjie Huang; Kyushik Mun; Sunitha Yarlagadda; Nambirajan Sundaram; Marco Kessler; Gerhard Hannig; Caroline B. Kurtz; Inmaculada Silos-Santiago; Michael A. Helmrath; Joseph J. Palermo; John P. Clancy; Kris A. Steinbrecher; Anjaparavanda P. Naren

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.


BMC Clinical Pharmacology | 2013

Metabolic fate of orally administered cGMP in rats

Elaine Liong; Christopher Graul; Samuel Rivers; Marco Kessler; Gerhard Hannig; Inmaculada Silos-Santiago

Background Activation of membrane guanylate cyclase type C (GC-C) on the luminal surface of the intestinal epithelium by GCC agonists results in an increase in both intracellular and extracellular levels of cyclic guanosine monophosphate (cGMP). Elevation in intracellular cGMP results in the secretion of chloride and bicarbonate anions into the intestinal lumen through the activation of the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel. Physiologically, this results in increased intestinal fluid and accelerated transit. Furthermore, an accompanying increase in the luminal and submucosal concentration of cGMP is observed as intracellular cGMP is transported out of epithelial cells. In animal models, decreased activity of pain-sensing afferent fibers is likely mediated by increased extracellular cGMP in the submucosa [1]. Compared to the pharmacological effects elicited by the increase in cGMP, less is known about the distribution and metabolic fate of this second messenger that is transported out into the luminal space.


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 Marco Kessler's collaboration.

Top Co-Authors

Avatar

Mark G. Currie

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerhard Hannig

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Robert Solinga

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pei Ge

Ironwood Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge