Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 28
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
EMBO J ; 43(11): 2264-2290, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38671253

RÉSUMÉ

Transient receptor potential (TRP) ion channels are involved in the surveillance or regulation of the acid-base balance. Here, we demonstrate that weak carbonic acids, including acetic acid, lactic acid, and CO2 activate and sensitize TRPV2 through a mechanism requiring permeation through the cell membrane. TRPV2 channels in cell-free inside-out patches maintain weak acid-sensitivity, but protons applied on either side of the membrane do not induce channel activation or sensitization. The involvement of proton modulation sites for weak acid-sensitivity was supported by the identification of titratable extracellular (Glu495, Glu561) and intracellular (His521) residues on a cryo-EM structure of rat TRPV2 (rTRPV2) treated with acetic acid. Molecular dynamics simulations as well as patch clamp experiments on mutant rTRPV2 constructs confirmed that these residues are critical for weak acid-sensitivity. We also demonstrate that the pore residue Glu609 dictates an inhibition of weak acid-induced currents by extracellular calcium. Finally, TRPV2-expression in HEK293 cells is associated with an increased weak acid-induced cytotoxicity. Together, our data provide new insights into weak acids as endogenous modulators of TRPV2.


Sujet(s)
Canaux cationiques TRPV , Canaux cationiques TRPV/métabolisme , Canaux cationiques TRPV/génétique , Canaux cationiques TRPV/composition chimique , Humains , Cellules HEK293 , Animaux , Rats , Simulation de dynamique moléculaire , Cryomicroscopie électronique , Calcium/métabolisme , Techniques de patch-clamp , Acides/métabolisme
3.
Nat Biotechnol ; 39(6): 737-746, 2021 06.
Article de Anglais | MEDLINE | ID: mdl-33558697

RÉSUMÉ

Organoid models of early tissue development have been produced for the intestine, brain, kidney and other organs, but similar approaches for the heart have been lacking. Here we generate complex, highly structured, three-dimensional heart-forming organoids (HFOs) by embedding human pluripotent stem cell aggregates in Matrigel followed by directed cardiac differentiation via biphasic WNT pathway modulation with small molecules. HFOs are composed of a myocardial layer lined by endocardial-like cells and surrounded by septum-transversum-like anlagen; they further contain spatially and molecularly distinct anterior versus posterior foregut endoderm tissues and a vascular network. The architecture of HFOs closely resembles aspects of early native heart anlagen before heart tube formation, which is known to require an interplay with foregut endoderm development. We apply HFOs to study genetic defects in vitro by demonstrating that NKX2.5-knockout HFOs show a phenotype reminiscent of cardiac malformations previously observed in transgenic mice.


Sujet(s)
Coeur/embryologie , Intestins/embryologie , Organoïdes/embryologie , Plan d'organisation du corps , Développement embryonnaire , Techniques de knock-down de gènes , Protéines à fluorescence verte/génétique , Facteur nucléaire hépatocytaire HNF-4/génétique , Protéine homéotique Nkx-2.5/génétique , Humains , Facteurs de transcription SOX-B1/génétique , Facteurs de transcription SOX-F/génétique , Analyse de séquence d'ARN
4.
Stem Cell Reports ; 14(5): 788-802, 2020 05 12.
Article de Anglais | MEDLINE | ID: mdl-32302556

RÉSUMÉ

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) represent an attractive model to investigate CM function and disease mechanisms. One characteristic marker of ventricular specificity of human CMs is expression of the ventricular, slow ß-myosin heavy chain (MyHC), as opposed to the atrial, fast α-MyHC. The main aim of this study was to investigate at the single-cell level whether contraction kinetics and electrical activity of hESC-CMs are influenced by the relative expression of α-MyHC versus ß-MyHC. For effective assignment of functional parameters to the expression of both MyHC isoforms at protein and mRNA levels in the very same hESC-CMs, we developed a single-cell mapping technique. Surprisingly, α- versus ß-MyHC was not related to specific contractile or electrophysiological properties of the same cells. The multiparametric cell-by-cell analysis suggests that in hESC-CMs the expression of genes associated with electrical activity, contraction, calcium handling, and MyHCs is independently regulated.


Sujet(s)
Potentiels d'action , Myosines cardiaques/métabolisme , Cellules souches embryonnaires humaines/cytologie , Contraction myocardique , Myocytes cardiaques/métabolisme , Chaînes lourdes de myosine/métabolisme , Myosines cardiaques/génétique , Différenciation cellulaire , Cellules cultivées , Cellules souches embryonnaires humaines/métabolisme , Humains , Myocytes cardiaques/cytologie , Myocytes cardiaques/physiologie , Chaînes lourdes de myosine/génétique , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Analyse sur cellule unique
5.
Nat Commun ; 11(1): 633, 2020 01 31.
Article de Anglais | MEDLINE | ID: mdl-32005803

RÉSUMÉ

Despite proven efficacy of pharmacotherapies targeting primarily global neurohormonal dysregulation, heart failure (HF) is a growing pandemic with increasing burden. Treatments mechanistically focusing at the cardiomyocyte level are lacking. MicroRNAs (miRNA) are transcriptional regulators and essential drivers of disease progression. We previously demonstrated that miR-132 is both necessary and sufficient to drive the pathological cardiomyocytes growth, a hallmark of adverse cardiac remodelling. Therefore, miR-132 may serve as a target for HF therapy. Here we report further mechanistic insight of the mode of action and translational evidence for an optimized, synthetic locked nucleic acid antisense oligonucleotide inhibitor (antimiR-132). We reveal the compound's therapeutic efficacy in various models, including a clinically highly relevant pig model of HF. We demonstrate favourable pharmacokinetics, safety, tolerability, dose-dependent PK/PD relationships and high clinical potential for the antimiR-132 treatment scheme.


Sujet(s)
Thérapie génétique/méthodes , Défaillance cardiaque/génétique , Défaillance cardiaque/thérapie , microARN/génétique , Oligonucléotides antisens/génétique , Animaux , Évaluation préclinique de médicament , Femelle , Régulation de l'expression des gènes , Défaillance cardiaque/métabolisme , Humains , microARN/métabolisme , Myocytes cardiaques/métabolisme , Oligonucléotides antisens/métabolisme , Oligonucléotides antisens/pharmacocinétique , Suidae
6.
Proc Natl Acad Sci U S A ; 116(48): 24359-24365, 2019 11 26.
Article de Anglais | MEDLINE | ID: mdl-31719194

RÉSUMÉ

Thermosensitive transient receptor potential (TRP) ion channels detect changes in ambient temperature to regulate body temperature and temperature-dependent cellular activity. Rodent orthologs of TRP vanilloid 2 (TRPV2) are activated by nonphysiological heat exceeding 50 °C, and human TRPV2 is heat-insensitive. TRPV2 is required for phagocytic activity of macrophages which are rarely exposed to excessive heat, but what activates TRPV2 in vivo remains elusive. Here we describe the molecular mechanism of an oxidation-induced temperature-dependent gating of TRPV2. While high concentrations of H2O2 induce a modest sensitization of heat-induced inward currents, the oxidant chloramine-T (ChT), ultraviolet A light, and photosensitizing agents producing reactive oxygen species (ROS) activate and sensitize TRPV2. This oxidation-induced activation also occurs in excised inside-out membrane patches, indicating a direct effect on TRPV2. The reducing agent dithiothreitol (DTT) in combination with methionine sulfoxide reductase partially reverses ChT-induced sensitization, and the substitution of the methionine (M) residues M528 and M607 to isoleucine almost abolishes oxidation-induced gating of rat TRPV2. Mass spectrometry on purified rat TRPV2 protein confirms oxidation of these residues. Finally, macrophages generate TRPV2-like heat-induced inward currents upon oxidation and exhibit reduced phagocytosis when exposed to the TRP channel inhibitor ruthenium red (RR) or to DTT. In summary, our data reveal a methionine-dependent redox sensitivity of TRPV2 which may be an important endogenous mechanism for regulation of TRPV2 activity and account for its pivotal role for phagocytosis in macrophages.


Sujet(s)
Méthionine/métabolisme , Canaux cationiques TRPV/composition chimique , Canaux cationiques TRPV/métabolisme , Canaux calciques/composition chimique , Canaux calciques/génétique , Canaux calciques/métabolisme , Chloramines/composition chimique , Escherichia coli/génétique , Température élevée , Humains , Peroxyde d'hydrogène/composition chimique , Macrophages , Méthionine/composition chimique , Mutation , Oxydants/composition chimique , Oxydoréduction , Techniques de patch-clamp , Phagocytose , Canaux cationiques TRPM/composition chimique , Canaux cationiques TRPM/métabolisme , Canaux cationiques TRPV/génétique , Composés tosyliques/composition chimique
8.
Stem Cell Res ; 40: 101542, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31473565

RÉSUMÉ

CFTR encodes for a chloride ion channel expressed primarily in secretory epithelia in the airways, intestine, liver and other tissues. Mutations in the CFTR gene have been identified in people suffering from Cystic Fibrosis. Here, we established a CFTR knock-in reporter cell line from a human iPSC line (MHHi006-A) using TALEN technology. The reporter enables the monitoring and optimization of the differentiation of pluripotent stem cells into CFTR expressing epithelia on a single cell level, as well as the enrichment of CFTR positive cells, which represent an excellent tool for Cystic Fibrosis disease modelling, drug screening and ultimately cellular therapies.


Sujet(s)
Protéine CFTR/génétique , Édition de gène , Cellules souches pluripotentes induites/cytologie , Potentiels d'action/effets des médicaments et des substances chimiques , Différenciation cellulaire , Lignée cellulaire , Reprogrammation cellulaire , Colforsine/pharmacologie , Humains , Cellules souches pluripotentes induites/métabolisme , Caryotype , Mâle , Nucléases effectrices de type activateur de transcription/génétique
9.
Sci Rep ; 9(1): 11173, 2019 08 01.
Article de Anglais | MEDLINE | ID: mdl-31371804

RÉSUMÉ

Loss-of-function mutations of the SCN5A gene encoding for the sodium channel α-subunit NaV1.5 result in the autosomal dominant hereditary disease Brugada Syndrome (BrS) with a high risk of sudden cardiac death in the adult. We here engineered human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) carrying the CRISPR/Cas9 introduced BrS-mutation p.A735V-NaV1.5 (g.2204C > T in exon 14 of SCN5A) as a novel model independent of patient´s genetic background. Recent studies raised concern regarding the use of hiPSC-CMs for studying adult-onset hereditary diseases due to cells' immature phenotype. To tackle this concern, long-term cultivation of hiPSC-CMs on a stiff matrix (27-42 days) was applied to promote maturation. Patch clamp recordings of A735V mutated hiPSC-CMs revealed a substantially reduced upstroke velocity and sodium current density, a prominent rightward shift of the steady state activation curve and decelerated recovery from inactivation as compared to isogenic hiPSC-CMs controls. These observations were substantiated by a comparative study on mutant A735V-NaV1.5 channels heterologously expressed in HEK293T cells. In contrast to mutated hiPSC-CMs, a leftward shift of sodium channel inactivation was not observed in HEK293T, emphasizing the importance of investigating mechanisms of BrS in independent systems. Overall, our approach supports hiPSC-CMs' relevance for investigating channelopathies in a dish.


Sujet(s)
Syndrome de Brugada/génétique , Cellules souches pluripotentes induites/cytologie , Mutation , Myocytes cardiaques/anatomopathologie , Canal sodique voltage-dépendant NAV1.5/génétique , Adulte , Syndrome de Brugada/anatomopathologie , Systèmes CRISPR-Cas , Cellules HEK293 , Humains , Techniques de patch-clamp
10.
Stem Cell Reports ; 13(2): 366-379, 2019 08 13.
Article de Anglais | MEDLINE | ID: mdl-31353227

RÉSUMÉ

Aiming at clinical translation, robust directed differentiation of human pluripotent stem cells (hPSCs), preferentially in chemically defined conditions, is a key requirement. Here, feasibility of suspension culture based hPSC-cardiomyocyte (hPSC-CM) production in low-cost, xeno-free media compatible with good manufacturing practice standards is shown. Applying stirred tank bioreactor systems at increasing dimensions, our advanced protocol enables routine production of about 1 million hPSC-CMs/mL, yielding ∼1.3 × 108 CM in 150 mL and ∼4.0 × 108 CMs in 350-500 mL process scale at >90% lineage purity. Process robustness and efficiency is ensured by uninterrupted chemical WNT pathway control at early stages of differentiation and results in the formation of almost exclusively ventricular-like CMs. Modulated WNT pathway regulation also revealed the previously unappreciated role of ROR1/CD13 as superior surrogate markers for predicting cardiac differentiation efficiency as soon as 72 h of differentiation. This monitoring strategy facilitates process upscaling and controlled mass production of hPSC derivatives.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Milieux de culture/pharmacologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Bioréacteurs , Antigènes CD13/génétique , Antigènes CD13/métabolisme , Techniques de culture cellulaire/méthodes , Milieux de culture/composition chimique , Humains , Mésoderme/métabolisme , Myocytes cardiaques/cytologie , Myocytes cardiaques/métabolisme , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/métabolisme , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme
11.
J Biol Chem ; 293(22): 8626-8637, 2018 06 01.
Article de Anglais | MEDLINE | ID: mdl-29674316

RÉSUMÉ

ClC-K channels belong to the CLC family of chloride channels and chloride/proton antiporters. They contribute to sodium chloride reabsorption in Henle's loop of the kidney and to potassium secretion into the endolymph by the stria vascularis of the inner ear. Their accessory subunit barttin stabilizes the ClC-K/barttin complex, promotes its insertion into the surface membrane, and turns the pore-forming subunits into a conductive state. Barttin mutations cause Bartter syndrome type IV, a salt-wasting nephropathy with sensorineural deafness. Here, studying ClC-K/barttin channels heterologously expressed in MDCK-II and HEK293T cells with confocal imaging and patch-clamp recordings, we demonstrate that the eight-amino-acids-long barttin N terminus is required for channel trafficking and activation. Deletion of the complete N terminus (Δ2-8 barttin) retained barttin and human hClC-Ka channels in intracellular compartments. Partial N-terminal deletions did not compromise subcellular hClC-Ka trafficking but drastically reduced current amplitudes. Sequence deletions encompassing Thr-6, Phe-7, or Arg-8 in barttin completely failed to activate hClC-Ka. Analyses of protein expression and whole-cell current noise revealed that inactive channels reside in the plasma membrane. Substituting the deleted N terminus with a polyalanine sequence was insufficient for recovering chloride currents, and single amino acid substitutions highlighted that the correct sequence is required for proper function. Fast and slow gate activation curves obtained from rat V166E rClC-K1/barttin channels indicated that mutant barttin fails to constitutively open the slow gate. Increasing expression of barttin over that of ClC-K partially recovered this insufficiency, indicating that N-terminal modifications of barttin alter both binding affinities and gating properties.


Sujet(s)
Membrane cellulaire/métabolisme , Canaux chlorure/métabolisme , Ouverture et fermeture des portes des canaux ioniques/physiologie , Rein/métabolisme , Mutation , Transport biologique , Cellules cultivées , Canaux chlorure/génétique , Cellules HEK293 , Humains , Domaines protéiques
12.
PLoS One ; 12(11): e0188008, 2017.
Article de Anglais | MEDLINE | ID: mdl-29141003

RÉSUMÉ

BACKGROUND: Local anaesthetics (LA) reduce neuronal excitability by inhibiting voltage-gated Na+ channels. When applied at high concentrations in the direct vicinity of nerves, LAs can also induce relevant irritation and neurotoxicity via mechanisms involving an increase of intracellular Ca2+. In the present study we explored the role of the Ca2+-permeable ion channels TRPA1 and TRPV1 for lidocaine-induced Ca2+-influx, neuropeptide release and neurotoxicity in mouse sensory neurons. METHODS: Cultured dorsal root ganglion (DRG) neurons from wildtype and mutant mice lacking TRPV1, TRPA1 or both channels were explored by means of calcium imaging, whole-cell patch clamp recordings and trypan blue staining for cell death. Release of calcitonin gene-related peptide (CGRP) from isolated mouse peripheral nerves was determined with ELISA. RESULTS: Lidocaine up to 10 mM induced a concentration-dependent reversible increase in intracellular Ca2+ in DRG neurons from wildtype and mutant mice lacking one of the two receptors, but not in neurons lacking both TRPA1 and TRPV1. 30 mM lidocaine also released Ca2+ from intracellular stores, presumably from the endoplasmic reticulum. While 10 mM lidocaine evoked an axonal CGRP release requiring expression of either TRPA1 or TRPV1, CGRP release induced by 30 mM lidocaine again mobilized internal Ca2+ stores. Lidocaine-evoked cell death required neither TRPV1 nor TRPA1. SUMMARY: Depending on the concentration, lidocaine employs TRPV1, TRPA1 and intracellular Ca2+ stores to induce a Ca2+-dependent release of the neuropeptide CGRP. Lidocaine-evoked cell death does not seem to require Ca2+ influx through TRPV1 or TRPV1.


Sujet(s)
Calcium/métabolisme , Lidocaïne/pharmacologie , Cellules réceptrices sensorielles/effets des médicaments et des substances chimiques , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire/physiologie , Canaux cationiques TRPV/physiologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Peptide relié au gène de la calcitonine/métabolisme , Cellules cultivées , Femelle , Ganglions sensitifs des nerfs spinaux/cytologie , Ganglions sensitifs des nerfs spinaux/effets des médicaments et des substances chimiques , Ganglions sensitifs des nerfs spinaux/métabolisme , Transport des ions , Mâle , Souris , Souris knockout , Techniques de patch-clamp , Cellules réceptrices sensorielles/métabolisme
13.
Sci Rep ; 7(1): 12775, 2017 10 06.
Article de Anglais | MEDLINE | ID: mdl-28986540

RÉSUMÉ

The irritant receptor TRPA1 was suggested to mediate analgesic, antipyretic but also pro-inflammatory effects of the non-opioid analgesic acetaminophen, presumably due to channel activation by the reactive metabolites parabenzoquinone (pBQ) and N-acetyl-parabenzoquinonimine (NAPQI). Here we explored the effects of these metabolites on the capsaicin receptor TRPV1, another redox-sensitive ion channel expressed in sensory neurons. Both pBQ and NAPQI, but not acetaminophen irreversibly activated and sensitized recombinant human and rodent TRPV1 channels expressed in HEK 293 cells. The reducing agents dithiothreitol and N-acetylcysteine abolished these effects when co-applied with the metabolites, and both pBQ and NAPQI failed to gate TRPV1 following substitution of the intracellular cysteines 158, 391 and 767. NAPQI evoked a TRPV1-dependent increase in intracellular calcium and a potentiation of heat-evoked currents in mouse spinal sensory neurons. Although TRPV1 is expressed in mouse hepatocytes, inhibition of TRPV1 did not alleviate acetaminophen-induced hepatotoxicity. Finally, intracutaneously applied NAPQI evoked burning pain and neurogenic inflammation in human volunteers. Our data demonstrate that pBQ and NAQPI activate and sensitize TRPV1 by interacting with intracellular cysteines. While TRPV1 does not seem to mediate acetaminophen-induced hepatotoxicity, our data identify TRPV1 as a target of acetaminophen with a potential relevance for acetaminophen-induced analgesia, antipyresia and inflammation.


Sujet(s)
Acétaminophène/métabolisme , Capsaïcine/pharmacologie , Métabolome , Canaux cationiques TRPV/métabolisme , Animaux , Benzoquinones/pharmacologie , Calcium/métabolisme , Mort cellulaire/effets des médicaments et des substances chimiques , Cystéine/métabolisme , Ganglions sensitifs des nerfs spinaux/effets des médicaments et des substances chimiques , Ganglions sensitifs des nerfs spinaux/métabolisme , Cellules HEK293 , Hépatocytes/cytologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Humains , Imines/pharmacologie , Espace intracellulaire/métabolisme , Ouverture et fermeture des portes des canaux ioniques/effets des médicaments et des substances chimiques , Mâle , Souris de lignée C57BL , Douleur/physiopathologie , Phosphorylation/effets des médicaments et des substances chimiques , Réducteurs/pharmacologie , Réflexe/effets des médicaments et des substances chimiques , Débit sanguin régional/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Canaux cationiques TRPV/agonistes
14.
Reg Anesth Pain Med ; 42(4): 462-468, 2017.
Article de Anglais | MEDLINE | ID: mdl-28394849

RÉSUMÉ

BACKGROUND AND OBJECTIVES: Regional anesthesia includes application of local anesthetics (LAs) into the vicinity of peripheral nerves. Prolongation or improvement of nerve blocks with LAs can be accomplished by coapplication with adjuvants, including buprenorphine, ketamine, and clonidine. While the mechanisms mediating prolonged or improved LA-induced effects by adjuvants are poorly understood, we hypothesized that they are likely to increase LA-induced block of voltage-gated Na channels. In this study, we investigated the inhibitory effects of the LA bupivacaine alone and in combination with the adjuvants on neuronal Na channels. METHODS: Effects of bupivacaine, buprenorphine, ketamine, and clonidine on endogenous Na channels in ND7/23 neuroblastoma cells were investigated with whole-cell patch clamp. RESULTS: Bupivacaine, buprenorphine, ketamine, and clonidine are concentration- and state-dependent inhibitors of Na currents in ND7/23 cells. Tonic block of resting channels revealed an order of potency of bupivacaine (half-maximal inhibitory concentration [IC50] 178 ± 8 µM) > buprenorphine (IC50 172 ± 25) > clonidine (IC50 824 ± 55 µM) > ketamine (IC50 1377 ± 92 µM). Bupivacaine and buprenorphine, but not clonidine and ketamine, induced a strong use-dependent block at 10 Hz. Except for clonidine, all substances enhanced fast and slow inactivation. The combination of bupivacaine with one of the adjuvants resulted in a concentration-dependent potentiation bupivacaine-induced block. CONCLUSIONS: We demonstrate that buprenorphine, ketamine, and clonidine directly inhibit Na channels and that they potentiate the blocking efficacy of bupivacaine on Na channels. These data indicate that block of Na channels may account for the additive effects of adjuvants used for regional anesthesia.


Sujet(s)
Potentiels d'action/effets des médicaments et des substances chimiques , Bupivacaïne/administration et posologie , Buprénorphine/administration et posologie , Clonidine/administration et posologie , Kétamine/administration et posologie , Bloqueurs de canaux sodiques voltage-dépendants/administration et posologie , Potentiels d'action/physiologie , Anesthésiques locaux/administration et posologie , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Synergie des médicaments , Humains
15.
Stem Cell Reports ; 8(2): 305-317, 2017 02 14.
Article de Anglais | MEDLINE | ID: mdl-28089668

RÉSUMÉ

Subtype-specific human cardiomyocytes (CMs) are valuable for basic and applied research. Induction of cardiomyogenesis and enrichment of nodal-like CMs was described for mouse pluripotent stem cells (mPSCs) in response to 1-ethyl-2-benzimidazolinone (EBIO), a chemical modulator of small-/intermediate-conductance Ca2+-activated potassium channels (SKs 1-4). Investigating EBIO in human pluripotent stem cells (PSCs), we have applied three independent differentiation protocols of low to high cardiomyogenic efficiency. Equivalent to mPSCs, timed EBIO supplementation during hPSC differentiation resulted in dose-dependent enrichment of up to 80% CMs, including an increase in nodal- and atrial-like phenotypes. However, our study revealed extensive EBIO-triggered cell loss favoring cardiac progenitor preservation and, subsequently, CMs with shortened action potentials. Proliferative cells were generally more sensitive to EBIO, presumably via an SK-independent mechanism. Together, EBIO did not promote cardiogenic differentiation of PSCs, opposing previous findings, but triggered lineage-selective survival at a cardiac progenitor stage, which we propose as a pharmacological strategy to modulate CM subtype composition.


Sujet(s)
Benzimidazoles/pharmacologie , Agonistes des canaux calciques/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Myocytes cardiaques/cytologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Cellules souches pluripotentes/cytologie , Cellules souches pluripotentes/effets des médicaments et des substances chimiques , Marqueurs biologiques , Différenciation cellulaire/génétique , Lignée cellulaire , Lignage cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Régulation de l'expression des gènes au cours du développement , Humains , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Canaux potassiques calcium-dépendants de conductance intermédiaire/génétique , Canaux potassiques calcium-dépendants de conductance intermédiaire/métabolisme , Mésoderme/cytologie , Mésoderme/effets des médicaments et des substances chimiques , Mésoderme/embryologie , Myocytes cardiaques/métabolisme , Cellules souches pluripotentes/métabolisme
16.
Sci Rep ; 6: 36740, 2016 11 09.
Article de Anglais | MEDLINE | ID: mdl-27827430

RÉSUMÉ

Tissue ischemia results in an accumulation of lactate and local or systemic lactic acidosis. In nociceptive sensory neurons, lactate was reported to sensitize or activate the transient receptor potential ion channel TRPA1 and acid-sensing ion channels (ASICs). However, it is unclear how lactate modulates the TRPV1 regarded as the main sensor for acidosis in sensory neurons. In this study we investigated the effects of lactate (LA) on recombinant and native TRPV1 channels and on TRPV1-mediated release of neuropeptides from mouse nerves. TRPV1-mediated membrane currents evoked by protons, capsaicin or heat are inhibited by LA at concentrations ranging from 3 µM to 100 mM. LA inhibits TRPV1-mediated proton-induced Ca2+-influx in dorsal root ganglion neurons as well as proton-evoked neuropeptide release from mouse nerves. Inhibition of TRPV1 by LA is significantly stronger on inward currents as compared to outward currents since LA affects channel gating, shifting the activation curve towards more positive potentials. The mutation I680A in the pore lower gate displays no LA inhibition. Cell-attached as well as excised inside- and outside-out patches suggest an interaction through an extracellular binding site. In conclusion, our data demonstrate that lactate at physiologically relevant concentrations is a potent endogenous inhibitor of TRPV1.


Sujet(s)
Signalisation calcique/effets des médicaments et des substances chimiques , Acide lactique/métabolisme , Acide lactique/pharmacologie , Canaux cationiques TRPV/antagonistes et inhibiteurs , Animaux , Calcium/métabolisme , Cellules HEK293 , Humains , Souris , Neuropeptides/métabolisme , Rats , Canaux cationiques TRPV/génétique , Canaux cationiques TRPV/métabolisme
17.
Nat Commun ; 5: 4381, 2014 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-25023795

RÉSUMÉ

Nitroxyl (HNO) is a redox sibling of nitric oxide (NO) that targets distinct signalling pathways with pharmacological endpoints of high significance in the treatment of heart failure. Beneficial HNO effects depend, in part, on its ability to release calcitonin gene-related peptide (CGRP) through an unidentified mechanism. Here we propose that HNO is generated as a result of the reaction of the two gasotransmitters NO and H2S. We show that H2S and NO production colocalizes with transient receptor potential channel A1 (TRPA1), and that HNO activates the sensory chemoreceptor channel TRPA1 via formation of amino-terminal disulphide bonds, which results in sustained calcium influx. As a consequence, CGRP is released, which induces local and systemic vasodilation. H2S-evoked vasodilatatory effects largely depend on NO production and activation of HNO-TRPA1-CGRP pathway. We propose that this neuroendocrine HNO-TRPA1-CGRP signalling pathway constitutes an essential element for the control of vascular tone throughout the cardiovascular system.


Sujet(s)
Peptide relié au gène de la calcitonine/métabolisme , Sulfure d'hydrogène/métabolisme , Monoxyde d'azote/pharmacologie , Oxydes d'azote/métabolisme , Canaux cationiques TRP/métabolisme , Animaux , Aorte/effets des médicaments et des substances chimiques , Aorte/métabolisme , Tronc cérébral/effets des médicaments et des substances chimiques , Tronc cérébral/métabolisme , Peptide relié au gène de la calcitonine/génétique , Humains , Immunohistochimie , Techniques in vitro , Souris , Souris knockout , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/génétique , Spectrométrie de masse MALDI , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire , Canaux cationiques TRP/génétique , Ganglion trigéminal/effets des médicaments et des substances chimiques , Ganglion trigéminal/métabolisme
18.
Anesth Analg ; 117(5): 1101-8, 2013 Nov.
Article de Anglais | MEDLINE | ID: mdl-24029851

RÉSUMÉ

BACKGROUND: Systemic administration of lipid emulsions is an established treatment for local anesthetic intoxication. However, it is unclear by which mechanisms lipids achieve this function. The high cardiac toxicity of the lipophilic local anesthetic bupivacaine probably results from a long-lasting inhibition of the cardiac Na channel Nav1.5. In this study, we sought to determine whether lipid emulsions functionally interact with Nav1.5 or counteract inhibition by bupivacaine. METHODS: Human embryonic kidney cells expressing human Nav1.5 were investigated by whole-cell patch clamp. The effects of Intralipid® and Lipofundin® were explored on functional properties and on bupivacaine-induced inhibition. RESULTS: Intralipid and Lipofundin did not affect the voltage dependency of activation, but induced a small hyperpolarizing shift of the steady-state fast inactivation and impaired the recovery from fast inactivation. Lipofundin, but not Intralipid, induced a concentration-dependent but voltage-independent tonic block (42% ± 4% by 3% Lipofundin). The half-maximal inhibitory concentration (IC50) values for tonic block by bupivacaine (50 ± 4 µM) were significantly increased when lipids were coapplied (5% Intralipid: 196 ± 22 µM and 5% Lipofundin: 103 ± 8 µM). Use-dependent block by bupivacaine at 10 Hz was also reduced by both lipid emulsions. Moreover, the recovery of inactivated channels from bupivacaine-induced block was faster in the presence of lipids. CONCLUSIONS: Our data indicate that lipid emulsions reduce rather than increase availability of Nav1.5. However, both Intralipid and Lipofundin partly relieve Nav1.5 from block by bupivacaine. These effects are likely to involve not only a direct interaction of lipids with Nav1.5 but also the ability of lipid emulsions to absorb bupivacaine and thus reduce its effective concentration.


Sujet(s)
Anesthésiques locaux/effets indésirables , Anesthésiques locaux/pharmacologie , Bupivacaïne/effets indésirables , Émulsion lipidique intraveineuse/pharmacologie , Canal sodique voltage-dépendant NAV1.5/effets des médicaments et des substances chimiques , Phospholipides/pharmacologie , Sorbitol/pharmacologie , Huile de soja/pharmacologie , Absorption , Relation dose-effet des médicaments , Association médicamenteuse , Électrophysiologie , Émulsions/pharmacologie , Cellules HEK293 , Humains , Concentration inhibitrice 50 , Rein/effets des médicaments et des substances chimiques , Lipides/composition chimique , Techniques de patch-clamp , Bloqueurs de canaux sodiques/pharmacologie
19.
J Biol Chem ; 288(28): 20280-92, 2013 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-23709225

RÉSUMÉ

The surveillance of acid-base homeostasis is concerted by diverse mechanisms, including an activation of sensory afferents. Proton-evoked activation of rodent sensory neurons is mainly mediated by the capsaicin receptor TRPV1 and acid-sensing ion channels. In this study, we demonstrate that extracellular acidosis activates and sensitizes the human irritant receptor TRPA1 (hTRPA1). Proton-evoked membrane currents and calcium influx through hTRPA1 occurred at physiological acidic pH values, were concentration-dependent, and were blocked by the selective TRPA1 antagonist HC030031. Both rodent and rhesus monkey TRPA1 failed to respond to extracellular acidosis, and protons even inhibited rodent TRPA1. Accordingly, mouse dorsal root ganglion neurons lacking TRPV1 only responded to protons when hTRPA1 was expressed heterologously. This species-specific activation of hTRPA1 by protons was reversed in both mouse and rhesus monkey TRPA1 by exchange of distinct residues within transmembrane domains 5 and 6. Furthermore, protons seem to interact with an extracellular interaction site to gate TRPA1 and not via a modification of intracellular N-terminal cysteines known as important interaction sites for electrophilic TRPA1 agonists. Our data suggest that hTRPA1 acts as a sensor for extracellular acidosis in human sensory neurons and should thus be taken into account as a yet unrecognized transduction molecule for proton-evoked pain and inflammation. The species specificity of this property is unique among known endogenous TRPA1 agonists, possibly indicating that evolutionary pressure enforced TRPA1 to inherit the role as an acid sensor in human sensory neurons.


Sujet(s)
Canaux calciques/métabolisme , Protéines de tissu nerveux/métabolisme , Protons , Canaux cationiques TRP/métabolisme , Animaux , Sites de fixation/génétique , Calcium/métabolisme , Canaux calciques/génétique , Capsaïcine/pharmacologie , Cellules cultivées , Cymènes , Ganglions sensitifs des nerfs spinaux/cytologie , Ganglions sensitifs des nerfs spinaux/métabolisme , Cellules HEK293 , Humains , Concentration en ions d'hydrogène , Ionomycine/pharmacologie , Macaca mulatta , Potentiels de membrane/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Souris knockout , Monoterpènes/pharmacologie , Mutation , Protéines de tissu nerveux/génétique , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/physiologie , Potassium/pharmacologie , Rats , Spécificité d'espèce , Membre-1 de la sous-famille A de canaux cationiques à potentiel de récepteur transitoire , Canaux cationiques TRP/génétique
20.
BMC Neurol ; 12: 104, 2012 Sep 24.
Article de Anglais | MEDLINE | ID: mdl-23006332

RÉSUMÉ

BACKGROUND: The mammalian neurological disorder hereditary hyperekplexia can be attributed to various mutations of strychnine sensitive glycine receptors. The clinical symptoms of "startle disease" predominantly occur in the newborn leading to convulsive hypertonia and an exaggerated startle response to unexpected mild stimuli. Amongst others, point mutations R271Q and R271L in the α1-subunit of strychnine sensitive glycine receptors show reduced glycine sensitivity and cause the clinical symptoms of hyperekplexia.Halogenation has been shown to be a crucial structural determinant for the potency of a phenolic compound to positively modulate glycine receptor function.The aim of this in vitro study was to characterize the effects of 4-chloropropofol (4-chloro-2,6-dimethylphenol) at four glycine receptor mutations. METHODS: Glycine receptor subunits were expressed in HEK 293 cells and experiments were performed using the whole-cell patch-clamp technique. RESULTS: 4-chloropropofol exerted a positive allosteric modulatory effect in a low sub-nanomolar concentration range at the wild type receptor (EC50 value of 0.08 ± 0.02 nM) and in a micromolar concentration range at the mutations (1.3 ± 0.6 µM, 0.1 ± 0.2 µM, 6.0 ± 2.3 µM and 55 ± 28 µM for R271Q, L, K and S267I, respectively). CONCLUSIONS: 4-chloropropofol might be an effective compound for the activation of mutated glycine receptors in experimental models of startle disease.


Sujet(s)
Chlore/métabolisme , Chlorophénols/administration et posologie , Épilepsie/métabolisme , Maladies génétiques liées au chromosome X/métabolisme , Ouverture et fermeture des portes des canaux ioniques/effets des médicaments et des substances chimiques , Potentiels de membrane/effets des médicaments et des substances chimiques , Récepteur de la glycine/agonistes , Récepteur de la glycine/métabolisme , Relation dose-effet des médicaments , Cellules HEK293 , Humains , Mutagenèse dirigée , Récepteur de la glycine/génétique , Réflexes anormaux , Xylènes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...