EXCEED THE SPACE PROVIDED. Mechanosensitivity is required for the efficient and effective processing of intestinal content. The overall objective of this proposal is to identify in human intestinal smooth muscle cells and in interstitial cells of Cajal (ICC) the mechanisms and physiological and patho_physiological relevance of mechanosensitivity. Our preliminary data2+,suggest that mechanosensltive' (MS) Ca 2 channels in human intestinal smooth muscle regulate Ca influx in response to mechanical stimuli and that their expression is altered in disease states. Our preliminary data also suggest that human intestinal smooth muscle and ICC express a novel Na + channel that is MS and that motility may be altered in patients with mutations in the Na + channel or in patients with altered expression of the channel. Based on this work we have generated the novel hypothesis that intestinal contractility is regulated by MS L-type Ca 2+ channels expressed in intestinal smooth muscle and MS Na channels expressed in intestinal smooth muscle and ICC and that disturbed meehanosensitivity may lead to motility disorders. The proposal has two specific aims.
Our first aim will focus on the physiological and pathophysiological role of MS L-type Ca 2+ channels in intestinal smooth muscle by d!rectly testing the hypotheses that: a) activation9f_ MS L-type. Cat channels by stretch increases' Ca entry and . contraction; b) the expressed L-type Ca channel IS MS; and c) upregulation of MS L-type Ca channels in obstruction results in increased contractile force.
Our second aim will focus on the physiological and pathophysiologieal role of MS Na + channels in human intestinal smooth muscle cells and ICC by directly testing the hypotheses that: a) M S Na + channels are present in smooth muscle cells and ICC ayjd that mechanoactivation increases Na _ entrv and regulates the membrane potential; b) the mammalian gene -I-' _-' + encoding the alpha subunit of the Na channel is SCN5A; and c) altered function of MS Na channels may contribute to the development of intestinal pseudo-obstruction. The studies will employ patch clamp techniques on freshly dissociated human smooth muscle cells and ICC, muscle strip techniques, optical laser tweezers, laser capture micro-dissection, immunohistochemistry, single cell PCR and standard molecular biology techniques to answer the questions raised. We are now uniquely poised to pursue our long term goal of providing new information of the role of MS ion channels in the regulation of intestinal contractility in both health and disease states. This work has the potential of identifying the pathophysiology of a subset of motility disorders such as intestinal pseudo-obstruction as well as provide novel therapeutic strategies. PERFORMANCE SITE ========================================Section End===========================================

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK052766-08
Application #
6822006
Study Section
General Medicine A Subcommittee 2 (GMA)
Program Officer
Hamilton, Frank A
Project Start
1997-09-01
Project End
2007-12-31
Budget Start
2005-01-01
Budget End
2005-12-31
Support Year
8
Fiscal Year
2005
Total Cost
$289,080
Indirect Cost
Name
Mayo Clinic, Rochester
Department
Type
DUNS #
006471700
City
Rochester
State
MN
Country
United States
Zip Code
55905
Treichel, Anthony J; Farrugia, Gianrico; Beyder, Arthur (2018) The touchy business of gastrointestinal (GI) mechanosensitivity. Brain Res 1693:197-200
Knutson, Katilyn; Strege, Peter R; Li, Joyce et al. (2018) Whole Cell Electrophysiology of Primary Cultured Murine Enterochromaffin Cells. J Vis Exp :
Strege, Peter R; Mazzone, Amelia; Bernard, Cheryl E et al. (2018) Irritable bowel syndrome patients have SCN5A channelopathies that lead to decreased NaV1.5 current and mechanosensitivity. Am J Physiol Gastrointest Liver Physiol 314:G494-G503
Alcaino, Constanza; Knutson, Kaitlyn R; Treichel, Anthony J et al. (2018) A population of gut epithelial enterochromaffin cells is mechanosensitive and requires Piezo2 to convert force into serotonin release. Proc Natl Acad Sci U S A 115:E7632-E7641
Strege, Peter R; Knutson, Kaitlyn; Eggers, Samuel J et al. (2017) Sodium channel NaV1.3 is important for enterochromaffin cell excitability and serotonin release. Sci Rep 7:15650
Alcaino, C; Farrugia, G; Beyder, A (2017) Mechanosensitive Piezo Channels in the Gastrointestinal Tract. Curr Top Membr 79:219-244
Wang, Fan; Knutson, Kaitlyn; Alcaino, Constanza et al. (2017) Mechanosensitive ion channel Piezo2 is important for enterochromaffin cell response to mechanical forces. J Physiol 595:79-91
Beyder, A; Gibbons, S J; Mazzone, A et al. (2016) Expression and function of the Scn5a-encoded voltage-gated sodium channel NaV 1.5 in the rat jejunum. Neurogastroenterol Motil 28:64-73
Beyder, Arthur; Farrugia, Gianrico (2016) Ion channelopathies in functional GI disorders. Am J Physiol Gastrointest Liver Physiol 311:G581-G586
Neshatian, Leila; Strege, Peter R; Rhee, Poong-Lyul et al. (2015) Ranolazine inhibits voltage-gated mechanosensitive sodium channels in human colon circular smooth muscle cells. Am J Physiol Gastrointest Liver Physiol 309:G506-12

Showing the most recent 10 out of 36 publications