TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis.

Standard

TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis. / Schattling, Benjamin; Steinbach, Karin; Thies, Edda; Kruse, Martin; Menigoz, Aurélie; Ufer, Friederike; Flockerzi, Veit; Brück, Wolfgang; Pongs, Olaf; Vennekens, Rudi; Kneussel, Matthias; Freichel, Marc; Merkler, Doron; Friese, Manuel A.

In: NAT MED, Vol. 18, No. 12, 12, 2012, p. 1805-1811.

Research output: SCORING: Contribution to journalSCORING: Journal articleResearchpeer-review

Harvard

Schattling, B, Steinbach, K, Thies, E, Kruse, M, Menigoz, A, Ufer, F, Flockerzi, V, Brück, W, Pongs, O, Vennekens, R, Kneussel, M, Freichel, M, Merkler, D & Friese, MA 2012, 'TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis.', NAT MED, vol. 18, no. 12, 12, pp. 1805-1811. <http://www.ncbi.nlm.nih.gov/pubmed/23160238?dopt=Citation>

APA

Schattling, B., Steinbach, K., Thies, E., Kruse, M., Menigoz, A., Ufer, F., Flockerzi, V., Brück, W., Pongs, O., Vennekens, R., Kneussel, M., Freichel, M., Merkler, D., & Friese, M. A. (2012). TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis. NAT MED, 18(12), 1805-1811. [12]. http://www.ncbi.nlm.nih.gov/pubmed/23160238?dopt=Citation

Vancouver

Bibtex

@article{dbdac53cf24746ad8abf39f380cf7f6f,
title = "TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis.",
abstract = "In multiple sclerosis, an inflammatory disease of the central nervous system (CNS), axonal and neuronal loss are major causes for irreversible neurological disability. However, which molecules contribute to axonal and neuronal injury under inflammatory conditions remains largely unknown. Here we show that the transient receptor potential melastatin 4 (TRPM4) cation channel is crucial in this process. TRPM4 is expressed in mouse and human neuronal somata, but it is also expressed in axons in inflammatory CNS lesions in experimental autoimmune encephalomyelitis (EAE) in mice and in human multiple sclerosis tissue. Deficiency or pharmacological inhibition of TRPM4 using the antidiabetic drug glibenclamide resulted in reduced axonal and neuronal degeneration and attenuated clinical disease scores in EAE, but this occurred without altering EAE-relevant immune function. Furthermore, Trpm4(-/-) mouse neurons were protected against inflammatory effector mechanisms such as excitotoxic stress and energy deficiency in vitro. Electrophysiological recordings revealed TRPM4-dependent neuronal ion influx and oncotic cell swelling upon excitotoxic stimulation. Therefore, interference with TRPM4 could translate into a new neuroprotective treatment strategy.",
author = "Benjamin Schattling and Karin Steinbach and Edda Thies and Martin Kruse and Aur{\'e}lie Menigoz and Friederike Ufer and Veit Flockerzi and Wolfgang Br{\"u}ck and Olaf Pongs and Rudi Vennekens and Matthias Kneussel and Marc Freichel and Doron Merkler and Friese, {Manuel A.}",
year = "2012",
language = "English",
volume = "18",
pages = "1805--1811",
journal = "NAT MED",
issn = "1078-8956",
publisher = "NATURE PUBLISHING GROUP",
number = "12",

}

RIS

TY - JOUR

T1 - TRPM4 cation channel mediates axonal and neuronal degeneration in experimental autoimmune encephalomyelitis and multiple sclerosis.

AU - Schattling, Benjamin

AU - Steinbach, Karin

AU - Thies, Edda

AU - Kruse, Martin

AU - Menigoz, Aurélie

AU - Ufer, Friederike

AU - Flockerzi, Veit

AU - Brück, Wolfgang

AU - Pongs, Olaf

AU - Vennekens, Rudi

AU - Kneussel, Matthias

AU - Freichel, Marc

AU - Merkler, Doron

AU - Friese, Manuel A.

PY - 2012

Y1 - 2012

N2 - In multiple sclerosis, an inflammatory disease of the central nervous system (CNS), axonal and neuronal loss are major causes for irreversible neurological disability. However, which molecules contribute to axonal and neuronal injury under inflammatory conditions remains largely unknown. Here we show that the transient receptor potential melastatin 4 (TRPM4) cation channel is crucial in this process. TRPM4 is expressed in mouse and human neuronal somata, but it is also expressed in axons in inflammatory CNS lesions in experimental autoimmune encephalomyelitis (EAE) in mice and in human multiple sclerosis tissue. Deficiency or pharmacological inhibition of TRPM4 using the antidiabetic drug glibenclamide resulted in reduced axonal and neuronal degeneration and attenuated clinical disease scores in EAE, but this occurred without altering EAE-relevant immune function. Furthermore, Trpm4(-/-) mouse neurons were protected against inflammatory effector mechanisms such as excitotoxic stress and energy deficiency in vitro. Electrophysiological recordings revealed TRPM4-dependent neuronal ion influx and oncotic cell swelling upon excitotoxic stimulation. Therefore, interference with TRPM4 could translate into a new neuroprotective treatment strategy.

AB - In multiple sclerosis, an inflammatory disease of the central nervous system (CNS), axonal and neuronal loss are major causes for irreversible neurological disability. However, which molecules contribute to axonal and neuronal injury under inflammatory conditions remains largely unknown. Here we show that the transient receptor potential melastatin 4 (TRPM4) cation channel is crucial in this process. TRPM4 is expressed in mouse and human neuronal somata, but it is also expressed in axons in inflammatory CNS lesions in experimental autoimmune encephalomyelitis (EAE) in mice and in human multiple sclerosis tissue. Deficiency or pharmacological inhibition of TRPM4 using the antidiabetic drug glibenclamide resulted in reduced axonal and neuronal degeneration and attenuated clinical disease scores in EAE, but this occurred without altering EAE-relevant immune function. Furthermore, Trpm4(-/-) mouse neurons were protected against inflammatory effector mechanisms such as excitotoxic stress and energy deficiency in vitro. Electrophysiological recordings revealed TRPM4-dependent neuronal ion influx and oncotic cell swelling upon excitotoxic stimulation. Therefore, interference with TRPM4 could translate into a new neuroprotective treatment strategy.

M3 - SCORING: Journal article

VL - 18

SP - 1805

EP - 1811

JO - NAT MED

JF - NAT MED

SN - 1078-8956

IS - 12

M1 - 12

ER -