Network


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

Hotspot


Dive into the research topics where Arthur Beyder is active.

Publication


Featured researches published by Arthur Beyder.


The Journal of Physiology | 2010

Mechanosensitivity of Nav1.5, a voltage‐sensitive sodium channel

Arthur Beyder; James L. Rae; Cheryl E. Bernard; Peter R. Strege; Frederick Sachs; Gianrico Farrugia

The voltage‐sensitive sodium channel Nav1.5 (encoded by SCN5A) is expressed in electromechanical organs and is mechanosensitive. This study aimed to determine the mechanosensitive transitions of Nav1.5 at the molecular level. Nav1.5 was expressed in HEK 293 cells and mechanosensitivity was studied in cell‐attached patches. Patch pressure up to −50 mmHg produced increases in current and large hyperpolarizing shifts of voltage dependence with graded shifts of half‐activation and half‐inactivation voltages (ΔV1/2) by ∼0.7 mV mmHg−1. Voltage dependence shifts affected channel kinetics by a single constant. This suggested that stretch accelerated only one of the activation transitions. Stretch accelerated voltage sensor movement, but not rate constants for gate opening and fast inactivation. Stretch also appeared to stabilize the inactivated states, since recovery from inactivation was slowed with stretch. Unitary conductance and maximum open probability were unaffected by stretch, but peak current was increased due to an increased number of active channels. Stretch effects were partially reversible, but recovery following a single stretch cycle required minutes. These data suggest that mechanical activation of Nav1.5 results in dose‐dependent voltage dependence shifts of activation and inactivation due to mechanical modulation of the voltage sensors.


Journal of Biological Chemistry | 2011

Altered expression of ANO1 variants in human diabetic gastroparesis

Amelia Mazzone; Cheryl E. Bernard; Peter R. Strege; Arthur Beyder; Luis J. V. Galietta; Pankaj J. Pasricha; James L. Rae; Henry P. Parkman; David R. Linden; Joseph H. Szurszewski; Tamas Ordog; Simon J. Gibbons; Gianrico Farrugia

Diabetes affects many organs including the stomach. Altered number and function of interstitial cells of Cajal (ICC), the gastrointestinal pacemaker cells, underlie a number of gastrointestinal motility disorders, including diabetic gastroparesis. In the muscle layers, ICC selectively express Ano1, thought to underlie classical Ca2+-activated Cl− currents. Mice homozygous for Ano1 knock-out exhibit abnormal ICC function and motility. Several transcripts for Ano1 are generated by alternative splicing of four exons. Here, we report expression levels of transcripts encoded by alternative splicing of Ano1 gene in gastric muscles of patients with diabetic gastroparesis and nondiabetic control tissues. Expression of mRNA from two alternatively transcribed exons are significantly different between patients and controls. Furthermore, patients with diabetic gastroparesis express mRNA for a previously unknown variant of Ano1. The 5′ end of this novel variant lacks exons 1 and 2 and part of exon 3. Expression of this variant in HEK cells produces a decreased density of Ca2+-activated Cl− currents that exhibit slower kinetics compared with the full-length Ano1. These results identify important changes in expression and splicing of Ano1 in patients with diabetic gastroparesis that alter the electrophysiological properties of the channel. Changes in Ano1 expression in ICC may directly contribute to diabetic gastroparesis.


Hepatology | 2015

Chronic Passive Venous Congestion drives Hepatic Fibrogenesis via Sinusoidal Thrombosis and Mechanical Forces

Douglas A. Simonetto; Hui yin Yang; Meng Yin; Thiago de Assuncao; Jung Hee Kwon; Moira Hilscher; Shuchong Pan; Liu Yang; Yan Bi; Arthur Beyder; Sheng Cao; Robert D. Simari; Richard L. Ehman; Patrick S. Kamath; Vijay H. Shah

Chronic passive hepatic congestion (congestive hepatopathy) leads to hepatic fibrosis; however, the mechanisms involved in this process are not well understood. We developed a murine experimental model of congestive hepatopathy through partial ligation of the inferior vena cava (pIVCL). C57BL/6 and transgenic mice overexpressing tissue factor pathway inhibitor (SM22α‐TFPI) were subjected to pIVCL or sham. Liver and blood samples were collected and analyzed in immunohistochemical, morphometric, real‐time polymerase chain reaction, and western blot assays. Hepatic fibrosis and portal pressure were significantly increased after pIVCL concurrent with hepatic stellate cell (HSC) activation. Liver stiffness, as assessed by magnetic resonance elastography, correlated with portal pressure and preceded fibrosis in our model. Hepatic sinusoidal thrombosis as evidenced by fibrin deposition was demonstrated both in mice after pIVCL as well as in humans with congestive hepatopathy. Warfarin treatment and TFPI overexpression both had a protective effect on fibrosis development and HSC activation after pIVCL. In vitro studies show that congestion stimulates HSC fibronectin (FN) fibril assembly through direct effects of thrombi as well as by virtue of mechanical strain. Pretreatment with either Mab13 or Cytochalasin‐D, to inhibit β‐integrin or actin polymerization, respectively, significantly reduced fibrin and stretch‐induced FN fibril assembly. Conclusion: Chronic hepatic congestion leads to sinusoidal thrombosis and strain, which in turn promote hepatic fibrosis. These studies mechanistically link congestive hepatopathy to hepatic fibrosis. (Hepatology 2015;61:648‐659)


Circulation | 2012

Ranolazine Decreases Mechanosensitivity of the Voltage-Gated Sodium Ion Channel NaV1.5 A Novel Mechanism of Drug Action

Arthur Beyder; Peter R. Strege; Santiago Reyes; Cheryl E. Bernard; Andre Terzic; Jonathan C. Makielski; Michael J. Ackerman; Gianrico Farrugia

Background— NaV1.5 is a mechanosensitive voltage-gated sodium-selective ion channel responsible for the depolarizing current and maintenance of the action potential plateau in the heart. Ranolazine is a NaV1.5 antagonist with antianginal and antiarrhythmic properties. Methods and Results— Mechanosensitivity of NaV1.5 was tested in voltage-clamped whole cells and cell-attached patches by bath flow and patch pressure, respectively. In whole cells, bath flow increased peak inward current in both murine ventricular cardiac myocytes (24±8%) and human embryonic kidney 293 cells heterologously expressing NaV1.5 (18±3%). The flow-induced increases in peak current were blocked by ranolazine. In cell-attached patches from cardiac myocytes and NaV1.5-expressing human embryonic kidney 293 cells, negative pressure increased NaV peak currents by 27±18% and 18±4% and hyperpolarized voltage dependence of activation by −11 mV and −10 mV, respectively. In human embryonic kidney 293 cells, negative pressure also increased the window current (250%) and increased late open channel events (250%). Ranolazine decreased pressure-induced shift in the voltage dependence (IC50 54 &mgr;mol/L) and eliminated the pressure-induced increases in window current and late current event numbers. Block of NaV1.5 mechanosensitivity by ranolazine was not due to the known binding site on DIVS6 (F1760). The effect of ranolazine on mechanosensitivity of NaV1.5 was approximated by lidocaine. However, ionized ranolazine and charged lidocaine analog (QX-314) failed to block mechanosensitivity. Conclusions— Ranolazine effectively inhibits mechanosensitivity of NaV1.5. The block of NaV1.5 mechanosensitivity by ranolazine does not utilize the established binding site and may require bilayer partitioning. Ranolazine block of NaV1.5 mechanosensitivity may be relevant in disorders of mechanoelectric dysfunction.


Frontiers in Physiology | 2011

Biophysically Based Modeling of the Interstitial Cells of Cajal: Current Status and Future Perspectives

Rachel Lees-Green; Peng Du; Gregory O'Grady; Arthur Beyder; Gianrico Farrugia; Andrew J. Pullan

Gastrointestinal motility research is progressing rapidly, leading to significant advances in the last 15 years in understanding the cellular mechanisms underlying motility, following the discovery of the central role played by the interstitial cells of Cajal (ICC). As experimental knowledge of ICC physiology has expanded, biophysically based modeling has become a valuable tool for integrating experimental data, for testing hypotheses on ICC pacemaker mechanisms, and for applications in in silico studies including in multiscale models. This review is focused on the cellular electrophysiology of ICC. Recent evidence from both experimental and modeling domains have called aspects of the existing pacemaker theories into question. Therefore, current experimental knowledge of ICC pacemaker mechanisms is examined in depth, and current theories of ICC pacemaking are evaluated and further developed. Existing biophysically based ICC models and their physiological foundations are then critiqued in light of the recent advances in experimental knowledge, and opportunities to improve these models are identified. The review concludes by examining several potential clinical applications of biophysically based ICC modeling from the subcellular through to the organ level, including ion channelopathies and ICC network degradation.


Neurogastroenterology and Motility | 2010

Lack of Serotonin 5-HT2B Receptor Alters Proliferation and Network Volume of Interstitial Cells of Cajal in Vivo

Vivek S. Tharayil; Mira M. Wouters; Jennifer E. Stanich; Jaime L. Roeder; Sha Lei; Arthur Beyder; Pedro J. Gomez-Pinilla; Michael D. Gershon; Luc Maroteaux; Simon J. Gibbons; Gianrico Farrugia

Background  Normal gastrointestinal motility requires intact networks of interstitial cells of Cajal (ICC). Interstitial cells of Cajal numbers are maintained by a balance between cell loss factors and survival/trophic/growth factors. Activation of 5‐HT2B receptors expressed on ICC increases ICC proliferation in vitro. It is not known whether 5‐HT2B receptors on ICC are activated in vivo. The aims of this study were to investigate if adult ICC proliferate, whether the proliferation of ICC in vivo is affected by knocking out the 5‐HT2B receptor, and if alterations in proliferation affect ICC networks.


The Journal of Physiology | 2013

The bioelectrical basis and validity of gastrointestinal extracellular slow wave recordings

Timothy R. Angeli; Peng Du; Niranchan Paskaranandavadivel; Patrick W. M. Janssen; Arthur Beyder; Roger G. Lentle; Ian P. Bissett; Leo K. Cheng; Gregory O’Grady

•  Extracellular recording techniques are commonly used to measure bioelectrical activity. However, the validity of gastrointestinal extracellular recordings has recently been challenged. •  In this joint experimental and modelling study, slow waves were recorded during contractile inhibition, biphasic and monophasic slow wave potentials were recorded simultaneously, and the biophysical basis of extracellular potentials was modelled with comparison to experimental data. •  The results showed that in vivo extracellular techniques reliably recorded slow waves in the absence of contractions, and potentials recorded using conventional serosal electrodes (biphasic) were concordant in phase and morphology with those recorded using suction electrodes (monophasic). •  Modelling further demonstrated that the morphology of experimental recordings is consistent with the biophysics underlying slow wave depolarisation. •  In total, these results demonstrate that gastrointestinal extracellular recordings are valid when performed and analysed correctly, reliably representing bioelectrical slow wave events. Motion suppression is not routinely required for in vivo extracellular studies.


Therapeutic Advances in Gastroenterology | 2012

Targeting ion channels for the treatment of gastrointestinal motility disorders

Arthur Beyder; Gianrico Farrugia

Gastrointestinal (GI) functional and motility disorders are highly prevalent and responsible for long-term morbidity and sometimes mortality in the affected patients. It is estimated that one in three persons has a GI functional or motility disorder. However, diagnosis and treatment of these widespread conditions remains challenging. This partly stems from the multisystem pathophysiology, including processing abnormalities in the central and peripheral (enteric) nervous systems and motor dysfunction in the GI wall. Interstitial cells of Cajal (ICCs) are central to the generation and propagation of the cyclical electrical activity and smooth muscle cells (SMCs) are responsible for electromechanical coupling. In these and other excitable cells voltage-sensitive ion channels (VSICs) are the main molecular units that generate and regulate electrical activity. Thus, VSICs are potential targets for intervention in GI motility disorders. Research in this area has flourished with advances in the experimental methods in molecular and structural biology and electrophysiology. However, our understanding of the molecular mechanisms responsible for the complex and variable electrical behavior of ICCs and SMCs remains incomplete. In this review, we focus on the slow waves and action potentials in ICCs and SMCs. We describe the constituent VSICs, which include voltage-gated sodium (NaV), calcium (CaV), potassium (KV, KCa), chloride (Cl–) and nonselective ion channels (transient receptor potentials [TRPs]). VSICs have significant structural homology and common functional mechanisms. We outline the approaches and limitations and provide examples of targeting VSICs at the pores, voltage sensors and alternatively spliced sites. Rational drug design can come from an integrated view of the structure and mechanisms of gating and activation by voltage or mechanical stress.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Hydrogen sulfide is a partially redox-independent activator of the human jejunum Na+ channel, Nav1.5

Peter R. Strege; Cheryl E. Bernard; Robert E. Kraichely; Amelia Mazzone; Lei Sha; Arthur Beyder; Simon J. Gibbons; David R. Linden; Michael L. Kendrick; Michael G. Sarr; Joseph H. Szurszewski; Gianrico Farrugia

Hydrogen sulfide (H(2)S) is produced endogenously by L-cysteine metabolism. H(2)S modulates several ion channels with an unclear mechanism of action. A possible mechanism is through reduction-oxidation reactions attributable to the redox potential of the sulfur moiety. The aims of this study were to determine the effects of the H(2)S donor NaHS on Na(V)1.5, a voltage-dependent sodium channel expressed in the gastrointestinal tract in human jejunum smooth muscle cells and interstitial cells of Cajal, and to elucidate whether H(2)S acts on Na(V)1.5 by redox reactions. Whole cell Na(+) currents were recorded in freshly dissociated human jejunum circular myocytes and Na(V)1.5-transfected human embryonic kidney-293 cells. RT-PCR amplified mRNA for H(2)S enzymes cystathionine β-synthase and cystathionine γ-lyase from the human jejunum. NaHS increased native Na(+) peak currents and shifted the half-point (V(1/2)) of steady-state activation and inactivation by +21 ± 2 mV and +15 ± 3 mV, respectively. Similar effects were seen on the heterologously expressed Na(V)1.5 α subunit with EC(50)s in the 10(-4) to 10(-3) M range. The reducing agent dithiothreitol (DTT) mimicked in part the effects of NaHS by increasing peak current and positively shifting steady-state activation. DTT together with NaHS had an additive effect on steady-state activation but not on peak current, suggesting that the latter may be altered via reduction. Pretreatment with the Hg(2+)-conjugated oxidizer thimerosal or the alkylating agent N-ethylmaleimide inhibited or decreased NaHS induction of Na(V)1.5 peak current. These studies show that H(2)S activates the gastrointestinal Na(+) channel, and the mechanism of action of H(2)S is partially redox independent.


The Journal of Physiology | 2017

Mechanosensitive ion channel Piezo2 is important for enterochromaffin cell response to mechanical forces

Fan Wang; Kaitlyn R. Knutson; Constanza Alcaino; David R. Linden; Simon J. Gibbons; Purna C. Kashyap; Madhusudan Grover; Richard Oeckler; Philip A. Gottlieb; Hui Joyce Li; Andrew B. Leiter; Gianrico Farrugia; Arthur Beyder

The gastrointestinal epithelial enterochromaffin (EC) cell synthesizes the vast majority of the bodys serotonin. As a specialized mechanosensor, the EC cell releases this serotonin in response to mechanical forces. However, the molecular mechanism of EC cell mechanotransduction is unknown. In the present study, we show, for the first time, that the mechanosensitive ion channel Piezo2 is specifically expressed by the human and mouse EC cells. Activation of Piezo2 by mechanical forces results in a characteristic ionic current, the release of serotonin and stimulation of gastrointestinal secretion. Piezo2 inhibition by drugs or molecular knockdown decreases mechanosensitive currents, serotonin release and downstream physiological effects. The results of the present study suggest that the mechanosensitive ion channel Piezo2 is specifically expressed by the EC cells of the human and mouse small bowel and that it is important for EC cell mechanotransduction.

Collaboration


Dive into the Arthur Beyder's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew B. Leiter

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge