In electro-mechanical organs, such as the gastrointestinal (GI) tract, ion channels are required to generate electrical activity that drives contractions. In turn, mechanical forces affect ion channel function and therefore electrical activity, which is termed mechano-electric feedback. Therefore, ion channel mechanosensitivity is important for normal function, and abnormalities can lead to disease. In the previous grant cycles we have shown that a mechano-sensitive voltage-gated sodium channel NaV1.5, encoded by SCN5A, is present in gastrointestinal smooth muscle cells of the human small bowel and colon. Further, SCN5A mutations are associated with irritable bowel syndrome (IBS). The contribution of NaV1.5 current density and mechanosensitivity to normal and abnormal mechano-electric feedback is not known. The central hypothesis of this proposal is that NaV1.5 mechanosensitivity and current density are critical for control of human GI smooth muscle excitability, and both are regulated and targetable. We will test the central hypothesis in 3 specific aims (SAs). We will determine in: SA1 how IBS NaV1.5 mutations affect mechanosensitivity and how changes in mechanosensitivity affect mechano-electric feedback; SA2 how NaV1.5 pore determines mechanosensitivity and mechanosensitivity block by certain drugs; SA3 how NaV1.5 is regulated by miRNAs in smooth muscle cells and the effects of NaV1.5 regulation by miRNA and drugs on GI smooth muscle cell function. The SAs are supported by extensive preliminary data. 1) 30% of IBS-associated SCN5A mutations result abnormal mechanosensitivity reducing mechano-electric feedback. 2) NaV1.5 mechanosensitivity depends on ion channel pore, and NaV1.5 mechanosensitivity blockade by drugs such as ranolazine is mechanistically separate from peak current block. 3) In GI smooth muscle from slow transit constipation patients NaV1.5 is down-regulated while a small set of miRNAs is upregulated and miRNA let-7f correlates with NaV1.5 expression, down-regulates NaV1.5 current and alters electrical slow wave activity. 4) Patients on ranolazine have delayed colon transit and rat GI transit is delayed by ranolazine. To investigate the central hypothesis we use a wide variety of cutting-edge techniques, including whole-cell and single-channel voltage- and current-clamp electrophysiology and optogenetics in combination with ultra-fast pressure delivery, CRISPR-Cas to introduce patient mutations into cells, bacterial NaV channels with designer functional domains, Western blots, IHC, delivery of miRNA mimics by lentivirus, rat organotypic cultures, and a prospective clinical trial. Successful completion of the proposed studies has both basic significance and clinical impact. As a result of the work done in the previous grant cycles and the preliminary data presented in this proposal, we will significantly advance our understanding of the molecular mechanisms of NaV1.5 channel mechanosensitivity, the contribution of NaV1.5 mechanosensitivity to mechano-electric feedback and regulation of NaV1.5 in GI smooth muscle in order to understand how to target NaV1.5 to modulate abnormal GI function.

Public Health Relevance

In the gastrointestinal tract ion channels are critical for normal electrical and mechanical function. Voltage- gated sodium channel NaV1.5, encoded by SCN5A, is important for excitability and responds to force by opening (mechanosensitivity). In some patients NaV1.5 mutations are associated with irritable bowel syndrome, which affects about 15% of US population. In this project we plan to study the basic mechanism of NaV1.5 mechanosensitivity, how certain drugs impact NaV1.5 mechanosensitivity, how IBS patient NaV1.5 mutations impact the channel's mechanosensitivity and how NaV1.5 density is regulated by microRNA in slow transit constipation patients. We will translate our findings by examining how manipulation of NaV1.5 and its mechanosensitivity can be used to impact gastrointestinal transit.

Agency
National Institute of Health (NIH)
Institute
National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK)
Type
Research Project (R01)
Project #
5R01DK052766-21
Application #
9690661
Study Section
Clinical, Integrative and Molecular Gastroenterology Study Section (CIMG)
Program Officer
Hamilton, Frank A
Project Start
1997-09-01
Project End
2022-04-30
Budget Start
2019-05-01
Budget End
2020-04-30
Support Year
21
Fiscal Year
2019
Total Cost
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, Arthur; Farrugia, Gianrico (2016) Ion channelopathies in functional GI disorders. Am J Physiol Gastrointest Liver Physiol 311:G581-G586
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
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