334 results on '"Trek-1"'
Search Results
102. Protective effects of TREK-1 against oxidative injury induced by SNP and H2O2.
- Author
-
Li-na SUN, Liao-liao LI, Zheng-bin LI, Ling WANG, and Xiao-liang WANG
- Subjects
CELL death ,OXIDATIVE stress ,HYDROGEN peroxide ,APOPTOSIS ,SODIUM nitroferricyanide - Abstract
Aim: TREK-1 (TWIK-related K
+ channel-1) is a 2-pore-domain K+ channel subtype. The present study investigated the role of TREK-1 in cell death induced by oxidative stress. Methods: The cell viability of wild-type Chinese hamster ovary (CHO) and TREK-1-transfected CHO cells (TREK-1/CHO cells) was measured using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay in the presence of sodium nitroprusside (SNP) or hydrogen peroxide (H2 O2 ). Apoptosis of wild-type CHO and TREK-1/CHO cells was detected using Hoechst33342 staining. Results: Both SNP and H2 O2 caused dose- and time-dependent growth inhibition of wild-type CHO and TREK-1/CHO cells. Following a 12 h exposure to SNP, the 50% inhibition (IC50 ) values for wild-type CHO and TREK-1/CHO cells were calculated as 0.69 mmol/L and 1.14 mmol/L, respectively. The IC50 values were 0.07 mmol/L and 0.09 mmol/L in H2 O2 -treated wild-type CHO and TREK-1/CHO cells, respectively, following 12 h exposure to H2 O2 . Moreover, SNP/H2 O2 induced less apoptosis in TREK-1/CHO cells than that in wild-type CHO cells ( P<0.05). Conclusion: The results demonstrated that TREK-1 played a protective role against oxidative injury. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
103. TWIK-related two-pore domain potassium channel TREK-1 in carotid endothelium of normotensive and hypertensive mice.
- Author
-
Pokojski, Sebastian, Busch, Christoph, Grgic, Ivica, Kacik, Michael, Salman, Waleed, Preisig-Müller, Regina, Heyken, Willm-Thomas, Daut, Jürgen, Hoyer, Joachim, and Köhler, Ralf
- Subjects
- *
POTASSIUM channels , *ENDOTHELIUM , *HYPERTENSION , *ION channels , *VASODILATION - Abstract
Aims: Potassium channels are essential elements of endothelial function. Recently, evidence emerged that the TWIK (tandem of P domains in a weak inwardly rectifying K+ channel)-related K+ channel (TREK-1) of the two-pore domain potassium channel gene family (K2P) may be involved in the regulation of vascular tone. However, the functional and molecular characterization of vascular TREK-1 is incomplete. In this study, we therefore analysed the functional expression of TREK-1 in the endothelium. Moreover, we hypothesized that changes in channel expression may contribute to altered endothelial vasodilator response under conditions of elevated blood pressure. [ABSTRACT FROM PUBLISHER]
- Published
- 2008
- Full Text
- View/download PDF
104. Curcumin inhibits bTREK-1 K+ channels and stimulates cortisol secretion from adrenocortical cells
- Author
-
Enyeart, Judith A., Liu, Haiyan, and Enyeart, John J.
- Subjects
- *
ADRENOCORTICOTROPIC hormone , *PHYTOCHEMICALS , *HYDROCORTISONE , *ELECTRIC potential - Abstract
Abstract: Bovine adrenal zona fasciculata (AZF) cells express bTREK-1 K+ channels that set the resting membrane potential. Inhibition of these channels by adrenocorticotropic hormone (ACTH) is coupled to membrane depolarization and cortisol secretion. Curcumin, a phytochemical with medicinal properties extracted from the spice turmeric, was found to modulate both bTREK-1 K+ currents and cortisol secretion from AZF cells. In whole-cell patch clamp experiments, curcumin inhibited bTREK-1 with an IC50 of 0.93μM by a mechanism that was voltage-independent. bTREK-1 inhibition by curcumin occurred through interaction with an external binding site and was independent of ATP hydrolysis. Curcumin produced a concentration-dependent increase in cortisol secretion that persisted for up to 24h. At a maximally effective concentration of 50μM, curcumin increased secretion as much as 10-fold. These results demonstrate that curcumin potently inhibits bTREK-1 K+ channels and stimulates cortisol secretion from bovine AZF cells. The inhibition of bTREK-1 by curcumin may be linked to cortisol secretion through membrane depolarization. Since TREK-1 is widely expressed in a variety of cells, it is likely that some of the biological actions of curcumin, including its therapeutic effects, may be mediated through inhibition of these K+ channels. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
105. Molecular basis of the mammalian pressure-sensitive ion channels: Focus on vascular mechanotransduction
- Author
-
Folgering, Joost H.A., Sharif-Naeini, Reza, Dedman, Alexandra, Patel, Amanda, Delmas, Patrick, and Honoré, Eric
- Subjects
- *
ION channels , *ACTIVE biological transport , *MEMBRANE proteins , *BLOOD circulation disorders - Abstract
Abstract: Mechano-gated ion channels are implicated in a variety of neurosensory functions ranging from touch sensitivity to hearing. In the heart, rhythm disturbance subsequent to mechanical effects is also associated with the activation of stretch-sensitive ion channels. Arterial autoregulation in response to hemodynamic stimuli, a vital process required for protection against hypertension-induced injury, is similarly dependent on the activity of force-sensitive ion channels. Seminal work in prokaryotes and invertebrates, including the nematode Caenorhabditis elegans and the fruit fly drosophila, greatly helped to identify the molecular basis of volume regulation, hearing and touch sensitivity. In mammals, more recent findings have indicated that members of several structural family of ion channels, namely the transient receptor potential (TRP) channels, the amiloride-sensitive ENaC/ASIC channels and the potassium channels K2P and Kir are involved in cellular mechanotransduction. In the present review, we will focus on the molecular and functional properties of these channel subunits and will emphasize on their role in the pressure-dependent arterial myogenic constriction and the flow-mediated vasodilation. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
106. Methionine and its derivatives increase bladder excitability by inhibiting stretch-dependent K(+) channels.
- Author
-
Baker, S. A., Hennig, G. W., Han, J., Britton, F. C., Smith, T. K., and Koh, S. D.
- Subjects
- *
BLADDER , *METHIONINE , *SULFUR amino acids , *SMOOTH muscle , *PHARMACOLOGY - Abstract
Background and purpose:During the bladder filling phase, the volume of the urinary bladder increases dramatically, with only minimal increases in intravesical pressure. To accomplish this, the smooth muscle of the bladder wall must remain relaxed during bladder filling. However, the mechanisms responsible for the stabilization of bladder excitability during stretch are unclear. We hypothesized that stretch-dependent K+ (TREK) channels in bladder smooth muscle cells may inhibit contraction in response to stretch.Experimental approaches:Bladder tissues from mouse, guinea pig and monkey were used for molecular, patch clamp, mechanical, electrical, Ca2+ imaging and cystometric responses to methionine and its derivatives, which are putative blockers of stretch-dependent K+ (SDK) channels.Key results:SDK channels are functionally expressed in bladder myocytes. The single channel conductance of SDK channels is 89pS in symmetrical K+ conditions and is blocked by L-methionine. Expressed TREK-1 currents are also inhibited by L-methioninol. All three types of bladder smooth muscle cells from mouse, guinea pig and monkey expressed TREK-1 genes. L-methionine, methioninol and methionine methyl ester but not D-methionine increased contractility in concentration-dependent manner. Methioninol further increased contractility and depolarized the membrane in the presence of blockers of Ca2+-activated K+ conductance. L-methionine induced Ca2+ waves that spread long distances through the tissue under stretched conditions and were associated with strong contractions. In cystometric assays, methioninol injection increased bladder excitability mimicking overactive bladder activity.Conclusions and implications:Methioninol-sensitive K+ (SDK, TREK-1) channels appear to be important to prevent spread of excitation through the syncitium during bladder filling.British Journal of Pharmacology (2008) 153, 1259–1271; doi:10.1038/sj.bjp.0707690; published online 21 January 2008 [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
107. Reduced inhibition of cortical glutamate and GABA release by halothane in mice lacking the K+ channel, TREK-1.
- Author
-
Westphalen, R. I., Krivitski, M., Amarosa, A., Guy, N., Hemmings Jr., H. C., and Hemmings, H C Jr
- Subjects
- *
GLUTAMIC acid , *GABA , *ANESTHETICS , *HALOTHANE , *IMMUNOBLOTTING , *BRAIN function localization , *CELL metabolism , *DRUG metabolism , *GLUTAMIC acid metabolism , *ANIMAL experimentation , *BIOCHEMISTRY , *CELL receptors , *CELLS , *CEREBRAL cortex , *IMMUNOHISTOCHEMISTRY , *LOCAL anesthetics , *PHENOMENOLOGY , *MICE , *NERVE endings , *NEUROTRANSMITTERS , *PULMONARY alveoli , *RESEARCH funding , *WESTERN immunoblotting , *INHALATION anesthetics , *PHARMACODYNAMICS - Abstract
Background and Purpose: Deletion of TREK-1, a two-pore domain K(+) channel (K(2P)) activated by volatile anaesthetics, reduces volatile anaesthetic potency in mice, consistent with a role for TREK-1 as an anaesthetic target. We used TREK-1 knockout mice to examine the presynaptic function of TREK-1 in transmitter release and its role in the selective inhibition of glutamate vs GABA release by volatile anaesthetics.Experimental Approach: The effects of halothane on 4-aminopyridine-evoked and basal [(3)H]glutamate and [(14)C]GABA release from cerebrocortical nerve terminals isolated from TREK-1 knockout (KO) and littermate wild-type (WT) mice were compared. TREK-1 was quantified by immunoblotting of nerve terminal preparations.Key Results: Deletion of TREK-1 significantly reduced the potency of halothane inhibition of 4-aminopyridine-evoked release of both glutamate and GABA without affecting control evoked release or the selective inhibition of glutamate vs GABA release. TREK-1 deletion also reduced halothane inhibition of basal glutamate release, but did not affect basal GABA release.Conclusions and Implications: The reduced sensitivity of glutamate and GABA release to inhibition by halothane in TREK-1 KO nerve terminals correlates with the reduced anaesthetic potency of halothane in TREK-1 KO mice observed in vivo. A presynaptic role for TREK-1 was supported by the enrichment of TREK-1 in isolated nerve terminals determined by immunoblotting. This study represents the first evidence for a link between an anaesthetic-sensitive 2-pore domain K(+) channel and presynaptic function, and provides further support for presynaptic mechanisms in determining volatile anaesthetic action. [ABSTRACT FROM AUTHOR]- Published
- 2007
- Full Text
- View/download PDF
108. The Presence of Arachidonic Acid-Activated K+ Channel, Trek-1, in Human Periodontal Ligament Fibroblasts.
- Author
-
Saeki, Yukikazu, Ohara, Akito, Nishikawa, Masanori, Yamamoto, Takahiro, and Yamamoto, Gaku
- Subjects
- *
ARACHIDONIC acid , *PERIODONTAL ligament , *FIBROBLASTS , *PHOSPHOLIPASE A2 , *MASTICATION , *ORAL habits , *PERIODONTAL disease , *ETIOLOGY of diseases , *PATCH-clamp techniques (Electrophysiology) - Abstract
Human periodontal ligament (PDL) fibroblasts expressed following two-pore-domain K+ channels, TWIK-2 > TREK-1 > TWIK-1 >> TASK-1 > TRAAK > TASK-2. TREK-2 message was not detectable. We found the presence of arachidonic acid-activated and mechanical stress-sensitive K+ channel, TREK-1, in the PDL fibroblasts by patch-clamp technique. It was also found the significant increase of intracellular concentration of arachidonic acid upon the application of cyclic stretch. Therefore, we suppose that the mechanical stretch due to the mastication activates phospholipase A2 to release arachidonic acid (AA) from membrane, then, the released AA activates TREK-1. Thus, TREK-1 K+ channels may play a protective role to maintain the negative membrane potential of PDL fibroblasts against the environmental stimuli. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
109. Evidence for two-pore domain potassium channels in rat cerebral arteries.
- Author
-
Bryan Jr., Robert M., Junping You, Phillips, Sharon C., Andresen, Jon J., Lloyd, Eric E., Rogers, Paul A., Dryer, Stuart E., and Marrelli, Sean P.
- Subjects
- *
VASCULAR smooth muscle , *MUSCLE cells , *ARACHIDONIC acid , *CEREBRAL arteries , *POTASSIUM channels , *RATS , *ANIMAL models in research - Abstract
Little is known about the presence and function of two-pore domain K+ (K2P) channels in vascular smooth muscle cells (VSMCs). Five members of the K2P channel family are known to be directly activated by arachidonic acid (AA). The purpose of this study was to determine 1) whether AA-sensitive K2P channels are expressed in cerebral VSMCs and 2) whether AA dilates the rat middle cerebral artery (MCA) by increasing K+ currents in VSMCs via an atypical K+ channel. RT-PCR revealed message for the following AA-sensitive K2P channels in rat MCA: tandem of P domains in weak inward rectifier K+ (TWIK-2), TWIK-related K+ (TREK-1 and TREK-2), TWIK-related AA-stimulated K+ (TRAAK), and TWIK-related halothane-inhibited K+ (THIK-1) channels. However, in isolated VSMCs, only message for TWIK-2 was found. Western blotting showed that TWIK-2 is present in MCA, and immunohistochemistry further demonstrated its presence in VSMCs. AA (10–100 μM) dilated MCAs through an endothelium-independent mechanism. AA-induced dilation was not affected by inhibition of cyclooxygenase, epoxygenase, or lipoxygenase or inhibition of classical K+ channels with 10 mM TEA, 3 mM 4-aminopyridine, 10 μM glibenclamide, or 100 μM Ba2+. AA-induced dilations were blocked by 50 mM K+, indicating involvement of a K+ channel. AA (10 μM) increased whole cell K+ currents in dispersed cerebral VSMCs. AA-induced currents were not affected by inhibitors of the AA metabolic pathways or blockade of classical K+ channels. We conclude that AA dilates the rat MCA and increases K+ currents in VSMCs via an atypical K+ channel that is likely a member of the K2P channel family. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
110. Interaction between the cellular prion (PrPC) and the 2P domain K+ channel TREK-1 protein
- Author
-
Azzalin, Alberto, Ferrara, Valentina, Arias, Agustina, Cerri, Silvia, Avella, Debora, Pisu, Maria Bonaria, Nano, Rosanna, Bernocchi, Graziella, Ferretti, Luca, and Comincini, Sergio
- Subjects
- *
PROTEINS , *BIOMOLECULES , *MAMMALS , *ION channels - Abstract
Abstract: The cellular prion protein (PrPC) is a highly conserved protein throughout the evolution of mammals and therefore is thought to play important cellular functions. Despite decades of intensive researches, the physiological function of PrPC remains enigmatic. Differently, in particular pathological contexts, generally referred as transmissible spongiform encephalopathies, a conformational isoform of PrPC, i.e., PrPSc, is considered the causative agent of these diseases. In this study, we investigated putative PrPC cellular functions through the identification of PrPC protein interactants. Using a bacterial two-hybrid approach, we identified a novel interaction between PrPC and a two-pore potassium channel protein, TREK-1. This interaction was further verified in transfected eukaryotic cells using co-immunoprecipitation and confocal microscopic analysis of the fluorescent transfected proteins. Importantly, in the cerebellar cortex, the endogenous PrPC and TREK-1 proteins exhibited co-localization signals in correspondence of the Purkinje cells. Furthermore, a deletion mapping study defined the carboxyl-terminal regions of the two proteins as the possible determinants of the PrPC–TREK-1 interaction. Our results indicated a novel PrPC interacting protein and suggested that this complex might be relevant in modulating a variety of electrophysiological-dependent cellular responses. [Copyright &y& Elsevier]
- Published
- 2006
- Full Text
- View/download PDF
111. Single-channel Recordings of TREK-1 K+ Channels in Periodontal Ligament Fibroblasts.
- Author
-
Ohara, A., Saeki, Y., Nishikawa, M., Yamamoto, Y., and Yamamoto, G.
- Subjects
PERIODONTAL ligament ,FIBROBLASTS ,MASTICATORY muscles ,ARACHIDONIC acid ,DENTISTRY ,TEETH - Abstract
The periodontal ligament (PDL) works as a suspensory ligament when external mechanical stress is placed on the teeth. PDL fibroblasts, the principal cells in the PDL, are responsible for many PDL functions. We hypothesized that mechanosensitive ion channels are present in human PDL fibroblasts, which are capable of responding to mechanical stress during normal function of the tissue. Using patch-clamp techniques, we detected mechanosensitive TREK-1 K
+ channels (a member of the two-pore-domain K+ channel family), whose single-channel conductance was 104 pS in symmetrical K+ -rich solutions. The open probability of the channel was low in the quiescent state, but it was strongly increased by the induction of membrane stretch. Arachidonic acid also enhanced the channel activity. RT-PCR and immunocytochemical observations showed the expression of TREK-1 K+ channels in PDL fibroblasts. The results suggest that the activation of TREK-1 K+ channels by masticatory stress contributes to the hyperpolarization of PDL fibroblasts. [ABSTRACT FROM AUTHOR]- Published
- 2006
- Full Text
- View/download PDF
112. TREK-1, a K+ channel involved in polymodal pain perception.
- Author
-
Alloui, Abdelkrim, Zimmermann, Katharina, Mamet, Julien, Duprat, Fabrice, Noël, Jacques, Chemin, Jean, Guy, Nicolas, Blondeau, Nicolas, Voilley, Nicolas, Rubat-Coudert, Catherine, Borsotto, Marc, Romey, Georges, Heurteaux, Catherine, Reeh, Peter, Eschalier, Alain, and Lazdunski, Michel
- Subjects
- *
PAIN perception , *POTASSIUM channels , *ION channels , *PAIN , *HYPERALGESIA , *INFLAMMATION - Abstract
The TREK-1 channel is a temperature-sensitive, osmosensitive and mechano-gated K+ channel with a regulation by Gs and Gq coupled receptors. This paper demonstrates that TREK-1 qualifies as one of the molecular sensors involved in pain perception. TREK-1 is highly expressed in small sensory neurons, is present in both peptidergic and nonpeptidergic neurons and is extensively colocalized with TRPV1, the capsaicin-activated nonselective ion channel. Mice with a disrupted TREK-1 gene are more sensitive to painful heat sensations near the threshold between anoxious warmth and painful heat. This phenotype is associated with the primary sensory neuron, as polymodal C-fibers were found to be more sensitive to heat in single fiber experiments. Knockout animals are more sensitive to low threshold mechanical stimuli and display an increased thermal and mechanical hyperalgesia in conditions of inflammation. They display a largely decreased pain response induced by osmotic changes particularly in prostaglandin E2-sensitized animals. TREK-1 appears as an important ion channel for polymodal pain perception and as an attractive target for the development of new analgesics. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
113. Desensitization of mechano-gated K2p channels.
- Author
-
Honoré, Eric, Patel, Amanda Jane, Chemin, Jean, Suchyna, Thomas, and Sachs, Frederick
- Subjects
- *
CYTOSKELETON , *CYTOPLASM , *SURGICAL excision , *OPERATIVE surgery , *ACIDOSIS , *ACID-base imbalances , *BIOLOGICAL membranes - Abstract
The neuronal mechano-gated K2P channels TREK-1 and TRAAK show pronounced desensitization within 100 ms of membrane stretch. Desensitization persists in the presence of cytoskeleton disrupting agents, upon patch excision, and when channels are expressed in membrane blebs. Mechanosensitive currents evoked with a variety of complex stimulus protocols were globally fit to a four-state cyclic kinetic model in detailed balance, without the need to introduce adaptation of the stimulus. However, we show that patch stress can be a complex function of time and stimulation history. The kinetic model couples desensitization to activation, so that gentle conditioning stimuli do not cause desensitization. Prestressing the channels with pressure, amphipaths, intracellular acidosis, or the E306A mutation reduces the peak-to-steady-state ratio by changing the preexponential terms of the rate constants, increasing the steady-state current amplitude. The mechanical responsivity can be accounted for by a change of in-plane area of ≃2 nm² between the closed and open conformations. Desensitization and its regulation by chemical messengers is predicted to condition the physiological role of K2P channels. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
114. Sodium permeable and 'hypersensitive' TREK‐1 channels cause ventricular tachycardia
- Author
-
Sven Zumhagen, Steffen Just, Marcus Schewe, Wendy González, Thomas Baukrowitz, Susanne Rinné, Rémi Peyronnet, Birgit Stallmeyer, Beatriz Ortiz-Bonnin, Jens Kockskämper, Daniel Bustos, Niels Decher, Corinna Friedrich, Eric Schulze-Bahr, Gunnar Seemann, Peter Kohl, Aytug K. Kiper, Commission of the European Communities, and British Heart Foundation
- Subjects
0301 basic medicine ,Tachycardia ,medicine.medical_specialty ,endocrine system ,Sodium ,Mutant ,chemistry.chemical_element ,RVOT ,Ventricular tachycardia ,arrhythmia ,Cardiovascular System ,03 medical and health sciences ,Potassium Channels, Tandem Pore Domain ,Cardiac Conduction System Disease ,Internal medicine ,medicine ,Ventricular outflow tract ,TREK‐1 ,Humans ,Research Articles ,Ions ,Chemistry ,K2P ,Point mutation ,Cardiac muscle ,Arrhythmias, Cardiac ,11 Medical And Health Sciences ,06 Biological Sciences ,medicine.disease ,Potassium channel ,030104 developmental biology ,medicine.anatomical_structure ,Cardiology ,Molecular Medicine ,two‐pore domain K+ channel ,Genetics, Gene Therapy & Genetic Disease ,medicine.symptom ,human activities ,Research Article - Abstract
In a patient with right ventricular outflow tract (RVOT) tachycardia, we identified a heterozygous point mutation in the selectivity filter of the stretch‐activated K2P potassium channel TREK‐1 ( KCNK2 or K2P2.1). This mutation introduces abnormal sodium permeability to TREK‐1. In addition, mutant channels exhibit a hypersensitivity to stretch‐activation, suggesting that the selectivity filter is directly involved in stretch‐induced activation and desensitization. Increased sodium permeability and stretch‐sensitivity of mutant TREK‐1 channels may trigger arrhythmias in areas of the heart with high physical strain such as the RVOT. We present a pharmacological strategy to rescue the selectivity defect of the TREK‐1 pore. Our findings provide important insights for future studies of K2P channel stretch‐activation and the role of TREK‐1 in mechano‐electrical feedback in the heart. ![][1] A point mutation in the selectivity filter of the stretch‐activated K2P potassium channel TREK‐1 was identified in a patient with right ventricular outflow tract tachycardia. The mutation most likely causes arrhythmias through abnormal sodium permeability and hypersensitivity to stretch‐activation. [1]: /embed/graphic-1.gif
- Published
- 2017
115. Serum sortilin-derived propeptides concentrations are decreased in major depressive disorder patients
- Author
-
Christelle Devader, Massimo Gennarelli, Morgane Roulot, Sébastien Moreno, Chiara Congiu, Marc Borsotto, Jean Mazella, Catherine Heurteaux, Alessandra Minelli, and Marco Bortolomasi
- Subjects
Adult ,Male ,0301 basic medicine ,medicine.medical_specialty ,TREK-1 ,Affect (psychology) ,Cohort Studies ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Propeptide ,Internal medicine ,Diagnosis ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Dosing ,Swimming ,Depression (differential diagnoses) ,Depressive Disorder, Major ,Depression ,Biomarker ,Middle Aged ,medicine.disease ,Sortilin ,Pathophysiology ,Diagnostic and Statistical Manual of Mental Disorders ,Mice, Inbred C57BL ,Clinical Psychology ,Psychiatry and Mental Health ,Adaptor Proteins, Vesicular Transport ,Psychiatry and Mental health ,030104 developmental biology ,Endocrinology ,Cohort ,Major depressive disorder ,Biomarker (medicine) ,Antidepressant ,Female ,Psychology ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Background Despite intense research on mechanisms underlying the depressive pathophysiology, reliable biomarkers to assess antidepressant treatment response are still lacking. Since the sortilin-derived propeptide (PE) displays potent antidepressant activities and can be measured in the blood of rodents, we wondered whether in human its seric level can vary between patients affected by major depressive disorder (MDD) and healthy controls and after antidepressant treatment. Methods By using a specific dosing method, characterized by structure-recognition analysis with various synthesized PE analogues, we conducted a translational study to test whether blood levels of PE are under pathophysiological regulation and could serve as biomarkers of the depression state. Results The serum concentration of PE, a peptide displaying potent antidepressant activities in rodents, is decreased in patients affected by major depressive disorder (MDD) when compared to healthy non-psychiatric controls cohort ( p =0.035). Interestingly, pharmacological antidepressant treatments restore normal PE levels. Limitations The limitation of the study concerns the relatively small patient samples that could negatively affect the likelihood that a nominally statistically significant finding actually reflects a true effect. Conclusions The longitudinal quantification of the serum PE concentration could assist psychiatrists in the diagnosis of antidepressant response efficacy, and the need to modify the therapeutic strategy.
- Published
- 2017
- Full Text
- View/download PDF
116. Inhibition of the human two-pore domain potassium channel, TREK-1, by fluoxetine and its metabolite norfluoxetine.
- Author
-
Kennard, Louise E., Chumbley, Justin R., Ranatunga, Kishani M., Armstrong, Stephanie J., Veale, Emma L., and Mathie, Alistair
- Subjects
- *
FLUOXETINE , *SEROTONIN uptake inhibitors , *ANTIDEPRESSANTS , *CHEMICAL reactions , *POTASSIUM , *DRUGS - Abstract
1. Block of the human two-pore domain potassium (2-PK) channel TREK-1 by fluoxetine (Prozac) and its active metabolite, norfluoxetine, was investigated using whole-cell patch-clamp recording of currents through recombinant channels in tsA 201 cells. 2. Fluoxetine produced a concentration-dependent inhibition of TREK-1 current that was reversible on wash. The IC50 for block was 19 microM. Block by fluoxetine was voltage-independent. Fluoxetine (100 microM) produced an 84% inhibition of TREK-1 currents, but only a 31% block of currents through a related 2-PK channel, TASK-3. 3. Norfluoxetine was a more potent inhibitor of TREK-1 currents with an IC50 of 9 microM. Block by norfluoxetine was also voltage-independent. 4. Truncation of the C-terminus of TREK-1 (delta89) resulted in a loss of channel function, which could be restored by intracellular acidification or the mutation E306A. The mutation E306A alone increased basal TREK-1 current and resulted in a loss of the slow phase of TREK-1 activation. 5. Progressive deletion of the C-terminus of TREK-1 had no effect on the inhibition of the channel by fluoxetine. The E306A mutation, on the other hand, reduced the magnitude of fluoxetine inhibition, with 100 microM producing only a 40% inhibition. 6. It is concluded that fluoxetine and norfluoxetine are potent inhibitors of TREK-1. Block of TREK-1 by fluoxetine may have important consequences when the drug is used clinically in the treatment of depression. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
117. Modulation of native TREK-1 and Kv1.4 K+ channels by polyunsaturated fatty acids and lysophospholipids.
- Author
-
Danthi, S., Enyeart, J. A., and Enyeart, J. J.
- Subjects
- *
ARACHIDONIC acid , *FATTY acids , *PHOSPHOLIPIDS , *BIOCHEMICAL mechanism of action , *LINOLEIC acid , *ESSENTIAL fatty acids , *ADRENAL cortex , *ANIMAL experimentation , *BIOLOGICAL transport , *CATTLE , *COMPARATIVE studies , *CYTOLOGICAL techniques , *RESEARCH methodology , *MEDICAL cooperation , *POTASSIUM , *RESEARCH , *RESEARCH funding , *UNSATURATED fatty acids , *EVALUATION research , *IN vitro studies , *PHYSIOLOGY - Abstract
The modulation of TREK-1 leak and Kv1.4 voltage-gated K+ channels by fatty acids and lysophospholipids was studied in bovine adrenal zona fasciculata (AZF) cells. In whole-cell patch-clamp recordings, arachidonic acid (AA) (1–20 µM) dramatically and reversibly increased the activity of bTREK-1, while inhibiting bKv1.4 current by mechanisms that occurred with distinctly different kinetics. bTREK-1 was also activated by the polyunsaturated cis fatty acid linoleic acid but not by the trans polyunsaturated fatty acid linolelaidic acid or saturated fatty acids. Eicosatetraynoic acid (ETYA), which blocks formation of active AA metabolites, failed to inhibit AA activation of bTREK-1, indicating that AA acts directly. Compared to activation of bTREK-1, inhibition of bKv1.4 by AA was rapid and accompanied by a pronounced acceleration of inactivation kinetics. Cis polyunsaturated fatty acids were much more effective than trans or saturated fatty acids at inhibiting bKv1.4. ETYA also effectively inhibited bKv1.4, but less potently than AA. bTREK-1 current was markedly increased by lysophospholipids including lysophosphatidyl choline (LPC) and lysophosphatidyl inositol (LPI). At concentrations from 1–5 µM, LPC produced a rapid, transient increase in bTREK-1 that peaked within one minute and then rapidly desensitized. The transient lysophospholipid-induced increases in bTREK-1 did not require the presence of ATP or GTP in the pipette solution. These results indicate that the activity of native leak and voltage-gated K+ channels are directly modulated in reciprocal fashion by AA and other cis unsaturated fatty acids. They also show that lysophospholipids enhance bTREK-1, but with a strikingly different temporal pattern. The modulation of native K+ channels by these agents differs from their effects on the same channels expressed in heterologous cells, highlighting the critical importance of auxiliary subunits and signaling. Finally, these results reveal that AZF cells express thousands of bTREK-1 K+ channels that lie dormant until activated by metabolites including phospholipase A2 (PLA2)-generated fatty acids and lysophospholipids. These metabolites may alter the electrical and secretory properties of AZF cells by modulating bTREK-1 and bKv1.4 K+ channels. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
118. Cold transduction in rat trigeminal ganglia neurons in vitro
- Author
-
Thut, P. D., Wrigley, D., and Gold, M. S.
- Subjects
- *
SENSORY neurons , *NOCICEPTORS - Abstract
Three sub-populations of sensory neurons may be distinguished based on responses to a decrease in temperature: one has a relatively low threshold for activation (cool fibers), a second has a high threshold for activation (cold nociceptors), and the third is unresponsive to a decrease in temperature. Results from several recent studies suggest that the ability to detect a decrease in temperature reflects an intrinsic property(ies) of sensory neurons and therefore may be characterized via the study of the sensory neuron cell body in vitro. However, while three unique ionic mechanisms of cold transduction have recently been identified (i.e. activation of the transient receptor potential channel M8 [TRPM8] or an epithelial Na+ channel [ENaC] or inhibition of two pore K+ channel [TREK-1]), the possibility that these “mechanisms” may be differentially distributed among sensory neurons in a manner consistent with predictions based on in vivo observations has not been investigated. To investigate this possibility, we have characterized the influence of cooling on isolated trigeminal ganglion (TG) neurons from adult rats in vitro with Ca2+ microfluorimetry in combination with a series of pharmacological interventions. We report that neurons responded to a decrease in temperature from approximately 34 °C to approximately 12 °C in one of two ways: 1) with a low threshold (30.1±0.6 °C) for activation demonstrating an increase in fluorescence with a minimal decrease in bath temperature (12.3%); 2) with a high threshold for activation (21.5±0.6 °C), demonstrating an increase in fluorescence only after a substantial decrease in bath temperature (13.3%); 74.4% did not respond to a decrease in temperature with an increase [Ca2+]i. These responses also were distinguishable on the basis of their rate of activation and degree of desensitization in response to prolonged application of a cold stimulus: low threshold responses were associated with a rapid (τ=12.0±5.7 s) increase in [Ca2+]i and a time constant of desensitization of 85.8±20.7 s while high threshold responses were associated with a slow (τ=38.1±8.2 s) increase in [Ca2+]i and demonstrated little desensitization over 4 min of stimulation. We refer to low threshold and high threshold cold responsive TG neurons as LTcool and HTcool neurons, respectively. LTcool and HTcool neurons were distributed among two distinct subpopulations of TG neurons distinguishable on the basis of cell body size and isolectin B4 staining. Both ENaC and TRPM8 appear to contribute to cold transduction, but neither is sufficient to account for all aspects of cold transduction in either population of TG neurons. Furthermore, inhibition of Ba2+ and/or Gd3+ sensitive two-pore K+ channels (i.e. TREK-1 and TRAAK) was insufficient to account for cold transduction in HTcool or LTcool neurons. Our results suggest that cold transduction in sensory neurons is a complex process involving the activation and inhibition of several different ion channels. In addition, there appear to be both similarities and differences between mechanisms underlying cold transduction in LTcool and HTcool neurons. Identification of specific mechanisms underlying cold transduction in LTcool and HTcool neurons may enable the development of novel therapeutic interventions for the treatment of pathological conditions such as cold allodynia. [Copyright &y& Elsevier]
- Published
- 2003
- Full Text
- View/download PDF
119. Trek-like Potassium Channels in Rat Cardiac Ventricular Myocytes Are Activated by Intracellular ATP.
- Author
-
Tan, J. H. C., Liu, W., and Saint, D. A.
- Subjects
POTASSIUM channels ,BIOLOGICAL membranes ,RATS ,MUSCLE cells ,ADENOSINE triphosphate ,PHOSPHORYLATION - Abstract
Large (111±3.0 pS) K+ channels were recorded in membrane patches from adult rat ventricular myocytes using patch-clamp techniques. The channels were not blocked by 4-AP (5 mM), intracellular TEA (5 mM) or glybenclamide (100 mM). Applying stretch to the membrane (as pipette suction) increased channel open probability (Po) in both cell-attached and isolated patches (typically, Po ~ 0.005 with no pressure; ~0.328 with 90 cm H2O: Vm = 40 mV, pHi = 7.2). The channels were activated by a decrease in intracellular pH; decreasing pHi to 5.5 from 7.2 increased Po to 0.16 from approx. 0.005 (no suction, Vm held at 40 mV). These properties are consistent with those demonstrated for TREK-1, a member of the recently cloned tandem pore family. We confirmed, using RT-PCR, that TREK-1 is expressed in rat ventricle, suggesting that the channel being recorded is indeed TREK-1. However, we show also that the channels are activated by millimolar concentrations of intracellular ATP. At a pH of 6 with no ATP at the intracellular membrane face, Po was 0.048±0.023, whereas Po increased to 0.22±0.1 with 1 mM ATP, and to 0.348±0.13 with 3 mM (n = 5; no membrane stretch applied). The rapid time course of the response and the fact that we see the effect in isolated patches appear to preclude phosphorylation. We conclude that intracellular ATP directly activates TREK-like channels, a property not previously described. [ABSTRACT FROM AUTHOR]
- Published
- 2002
- Full Text
- View/download PDF
120. The Knockdown of TREK-1 in Hippocampal Neurons Attenuate Lipopolysaccharide-Induced Depressive-Like Behavior in Mice
- Author
-
Ajung Kim, Jae Yong Park, Yeong Eun Kim, Seung-Chan Kim, Eun Mi Hwang, Hyun Gug Jung, and Seok-Geun Lee
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Genetically modified mouse ,medicine.medical_specialty ,endocrine system ,TREK-1 ,hippocampus ,Transgene ,Genetic Vectors ,Hippocampus ,Mice, Transgenic ,Biology ,Hippocampal formation ,Article ,Catalysis ,Inorganic Chemistry ,Mice ,03 medical and health sciences ,Potassium Channels, Tandem Pore Domain ,0302 clinical medicine ,Neurotrophic factors ,Internal medicine ,medicine ,Animals ,RNA, Small Interfering ,Physical and Theoretical Chemistry ,Receptor ,Molecular Biology ,Spectroscopy ,Depressive Disorder ,Gene knockdown ,Behavior, Animal ,Organic Chemistry ,General Medicine ,Dependovirus ,Tail suspension test ,Computer Science Applications ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Dentate Gyrus ,depression ,Cytokines ,neurotrophic factor, conditional knockdown ,Corticosterone ,human activities ,030217 neurology & neurosurgery - Abstract
TWIK-related potassium channel-1 (TREK-1) is broadly expressed in the brain and involved in diverse brain diseases, such as seizures, ischemia, and depression. However, the cell type-specific roles of TREK-1 in the brain are largely unknown. Here, we generated a Cre-dependent TREK-1 knockdown (Cd-TREK-1 KD) transgenic mouse containing a gene cassette for Cre-dependent TREK-1 short hairpin ribonucleic acid to regulate the cell type-specific TREK-1 expression. We confirmed the knockdown of TREK-1 by injecting adeno-associated virus (AAV) expressing Cre into the hippocampus of the mice. To study the role of hippocampal neuronal TREK-1 in a lipopolysaccharide (LPS)-induced depression model, we injected AAV-hSyn-BFP (nCTL group) or AAV-hSyn-BFP-Cre (nCre group) virus into the hippocampus of Cd-TREK-1 KD mice. Interestingly, the immobility in the tail suspension test after LPS treatment did not change in the nCre group. Additionally, some neurotrophic factors (BDNF, VEGF, and IGF-1) significantly increased more in the nCre group compared to the nCTL group after LPS treatment, but there was no difference in the expression of their receptors. Therefore, our data suggest that TREK-1 in the hippocampal neurons has antidepressant effects, and that Cd-TREK-1 KD mice are a valuable tool to reveal the cell type-specific roles of TREK-1 in the brain.
- Published
- 2019
- Full Text
- View/download PDF
121. TREK-1 protects the heart against ischemia-reperfusion-induced injury and from adverse remodeling after myocardial infarction
- Author
-
Samuel Kamatham, Christopher M. Waters, Salvatore Mancarella, and Andreas Schwingshackl
- Subjects
0301 basic medicine ,Male ,Potassium Channels ,TREK-1 ,Physiology ,Clinical Biochemistry ,Medical Physiology ,Action Potentials ,Blood Pressure ,Inbred C57BL ,Cardiovascular ,Mice ,0302 clinical medicine ,Heart Rate ,Telemetry ,2.1 Biological and endogenous factors ,Myocytes, Cardiac ,Myocardial infarction ,Potassium channel ,Aetiology ,Cells, Cultured ,Tandem Pore Domain ,Cardioprotection ,Cultured ,Ischemia-reperfusion ,Heart Disease ,Cardiology ,Cardiac ,medicine.medical_specialty ,endocrine system ,Cells ,Ischemia ,Myocardial Reperfusion Injury ,QT interval ,Article ,03 medical and health sciences ,Potassium Channels, Tandem Pore Domain ,Physiology (medical) ,Internal medicine ,Heart rate ,medicine ,Animals ,Calcium Signaling ,Heart Disease - Coronary Heart Disease ,Myocytes ,business.industry ,Human Movement and Sports Sciences ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Blood pressure ,business ,human activities ,030217 neurology & neurosurgery ,Homeostasis - Abstract
The TWIK-related K+ channels (TREK-1) is a two-pore domain potassium channel that produces background leaky type potassium currents. TREK-1 has a protective role against ischemia-induced neuronal damage. TREK-1 is also expressed in the heart, but its role in myocardial ischemia-reperfusion (IR)-induced injury has not been examined. In the current study, we used a TREK-1 knockout (KO) mouse model to show that TREK-1 has a critical role in the cardiac I/R-induced injury and during remodeling after myocardial infarction (MI). At baseline, TREK-1 KO mice had similar blood pressure and heart rate as the wildtype (WT) mice. However, the lack of TREK-1 was associated with increased susceptibility to ischemic injury and compromised functional recovery following ex-vivo I/R-induced injury. TREK-1 deficiency increased infarct size following permanent coronary artery ligation, resulting in greater systolic dysfunction than the WT counterpart. Electrocardiographic (ECG) analysis revealed QT interval prolongation in TREK-1 KO mice, but normal heart rate (HR). Acutely isolated TREK-1 KO cardiomyocytes exhibited prolonged Ca(2+) transients duration associated with action potential duration (APD) prolongation. Our data suggest that TREK-1 has a protective effect against I/R-induced injury and influences the post-MI remodeling processes by regulating membrane potential and maintaining intracellular Ca(2+) homeostasis. These data suggest that TREK-1 activation could be an effective strategy to provide cardioprotection against ischemia-induced damage.
- Published
- 2019
122. Trabecular Meshwork TREK-1 Channels Function as Polymodal Integrators of Pressure and pH
- Author
-
David Križaj, Tam T. T. Phuong, and Oleg Yarishkin
- Subjects
Physiology and Pharmacology ,Patch-Clamp Techniques ,TREK-1 ,Intracellular pH ,K2p channels ,Mechanotransduction, Cellular ,Membrane Potentials ,03 medical and health sciences ,0302 clinical medicine ,Potassium Channels, Tandem Pore Domain ,Extracellular ,medicine ,Pressure ,Humans ,Patch clamp ,Mechanotransduction ,Cells, Cultured ,030304 developmental biology ,mechanotransduction ,Membrane potential ,0303 health sciences ,Chemistry ,trabecular meshwork ,General Medicine ,Hydrogen-Ion Concentration ,3. Good health ,medicine.anatomical_structure ,Biophysics ,Mechanosensitive channels ,Trabecular meshwork ,030217 neurology & neurosurgery ,Intracellular - Abstract
Purpose The concentration of protons in the aqueous humor (AH) of the vertebrate eye is maintained close to blood pH; however, pathologic conditions and surgery may shift it by orders of magnitude. We investigated whether and how changes in extra- and intracellular pH affect the physiology and function of trabecular meshwork (TM) cells that regulate AH outflow. Methods Electrophysiology, in conjunction with pharmacology, gene knockdown, and optical recording, was used to track the pH dependence of transmembrane currents and mechanotransduction in primary and immortalized human TM cells. Results Extracellular acidification depolarized the resting membrane potential by inhibiting an outward K+-mediated current, whereas alkalinization hyperpolarized the cells and augmented the outward conductance. Intracellular acidification with sodium bicarbonate hyperpolarized TM cells, whereas removal of intracellular protons with ammonium chloride depolarized the membrane potential. The effects of extra- and intracellular acid and alkaline loading were abolished by quinine, a pan-selective inhibitor of two-pore domain potassium (K2P) channels, and suppressed by shRNA-mediated downregulation of the mechanosensitive K2P channel TREK-1. Extracellular acidosis suppressed, whereas alkalosis facilitated, the amplitude of the pressure-evoked TREK-1-mediated outward current. Conclusions These results demonstrate that TM mechanotransduction mediated by TREK-1 channels is profoundly sensitive to extra- and intracellular pH shifts. Intracellular acidification might modulate aqueous outflow and IOP by stimulating TREK-1 channels.
- Published
- 2019
123. Involvement of TREK-1 Channel in Cell Viability of H9c2 Rat Cardiomyoblasts Affected by Bupivacaine and Lipid Emulsion
- Author
-
Marie Merci Nyiramana, Eun-Jin Kim, Jaehee Han, Ju-Tae Sohn, Jong Woo Kim, Jun Ho Yang, Eui-Jung Shin, Adrian S. Siregar, and Dawon Kang
- Subjects
0301 basic medicine ,Programmed cell death ,endocrine system ,TREK-1 ,Cell Survival ,viruses ,Pharmacology ,Article ,Cell Line ,03 medical and health sciences ,Potassium Channels, Tandem Pore Domain ,0302 clinical medicine ,030202 anesthesiology ,Animals ,Humans ,Viability assay ,Cytotoxicity ,lcsh:QH301-705.5 ,Membrane Potential, Mitochondrial ,chemistry.chemical_classification ,Membrane potential ,Cardiotoxicity ,Chemistry ,bupivacaine ,Depolarization ,General Medicine ,biochemical phenomena, metabolism, and nutrition ,Lipids ,030104 developmental biology ,lcsh:Biology (General) ,cardiomyoblast ,membrane potential ,human activities ,Myoblasts, Cardiac ,Intracellular ,lipid emulsion ,Polyunsaturated fatty acid - Abstract
Lipid emulsion (LE) therapy has been used to reduce overdose of bupivacaine (BPV)-induced cardiotoxicity. The TWIK-related potassium channel-1 (TREK-1) is inhibited by BPV and activated by polyunsaturated fatty acids, which are the main component in LE. These pharmacological properties inspired us to investigate whether the TREK-1 channel is associated with cell viability of H9c2 cardiomyoblasts affected by BPV and LE. Consistent with previous studies, BPV-induced cell death was reduced by LE treatment. The reduction in the TREK-1 expression level by BPV was alleviated by LE. The BPV cytotoxicity highly decreased in TREK-1 overexpressed cells but was the opposite in TREK-1 knocked-down cells. TREK-1 channel activators and inhibitors increased and decreased cell viability, respectively. BPV-induced depolarization of the plasma and mitochondrial membrane potential and increase in intracellular Ca2+ level were blocked by LE treatment. BPV-induced depolarization of membrane potential was reduced in TREK-1 overexpressed cells, indicating that TREK-1 channels mediate setting the resting membrane potentials as a background K+ channel in H9c2 cells. These results show that TREK-1 activity is involved in the BPV cytotoxicity and the antagonistic effect of LE in H9c2 cells and suggest that TREK-1 could be a target for action of BPV and LE.
- Published
- 2019
- Full Text
- View/download PDF
124. Deficiency of TREK-1 potassium channel exacerbates blood-brain barrier damage and neuroinflammation after intracerebral hemorrhage in mice
- Author
-
Li Xu, Wei Wang, Yue Wan, Yongkang Fang, Minjie Xie, Yeye Tian, Qibao Huang, Dengji Pan, and Suiqiang Zhu
- Subjects
Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,TREK-1 ,Immunology ,Central nervous system ,Blood–brain barrier ,lcsh:RC346-429 ,Cerebral edema ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,Potassium Channels, Tandem Pore Domain ,0302 clinical medicine ,medicine ,Animals ,lcsh:Neurology. Diseases of the nervous system ,Neuroinflammation ,Cerebral Hemorrhage ,Evans Blue ,Inflammation ,Mice, Knockout ,Intracerebral hemorrhage ,Microglia ,business.industry ,Research ,General Neuroscience ,medicine.disease ,Secondary injury ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,chemistry ,Blood-Brain Barrier ,business ,human activities ,030217 neurology & neurosurgery - Abstract
Background Intracerebral hemorrhage (ICH) is a devastating medical emergency with high mortality and severe neurological deficit. ICH-related poor outcomes are due to a combination of pathological processes that could be complicated by secondary insults. TWIK-related K+ channel 1 (TREK-1) is a two-pore-domain potassium channel that is highly expressed in the mammalian nervous system. Previous studies have shown that TREK-1 channels play important roles in various central nervous system diseases. However, its role in the secondary injuries after intracerebral hemorrhage remains unknown. In this study, we explored the function of TREK-1 in secondary blood-brain barrier injuries and neuroinflammation after intracerebral hemorrhage in mice. Methods Adult male TREK-1−/− mice and WT mice were subjected to a collagenase-induced ICH model. Immunostaining, western blot, and enzyme-linked immunosorbent assay were used to assess inflammatory infiltration and neuronal death. Blood-brain barrier compromise was assessed using electron microscopy and Evans Blue dye injection on days 1 and 3 after intracerebral hemorrhage. Magnetic resonance imaging and behavioral assessments were conducted to evaluate the neurologic damage and recovery after intracerebral hemorrhage. Results Genetic deficiency of TREK-1 channel exacerbated blood-brain barrier impairment and promoted cerebral edema after intracerebral hemorrhage. Meanwhile, TREK-1 deficiency aggravated focal inflammatory featured by the increased recruitment of microglia and neutrophils, the enhanced secretion of proinflammatory factors interleukin-1 beta (IL-1β), tumor necrosis factor alpha (TNF-α), and cell adhesion molecules (CAMs). Furthermore, TREK-1 deficiency promoted neuronal injury and neurological impairment. Conclusions These results establish the first in vivo evidence for the protective role of TREK-1 in blood-brain barrier injury and neuroinflammation after intracerebral hemorrhage. TREK-1 may thereby be harnessed to a potential therapeutical target for the treatment of intracerebral hemorrhage. Electronic supplementary material The online version of this article (10.1186/s12974-019-1485-5) contains supplementary material, which is available to authorized users.
- Published
- 2019
- Full Text
- View/download PDF
125. Role of TREK-1 in Health and Disease, Focus on the Central Nervous System
- Author
-
Catherine Heurteaux, Marc Borsotto, Jean Mazella, Alaeddine Djillani, Signalisation calcique et interactions cellulaires dans le foie, Université Paris-Sud - Paris 11 (UP11)-Institut National de la Santé et de la Recherche Médicale (INSERM), Institut de pharmacologie moléculaire et cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS), Mazella, Jean, Université Nice Sophia Antipolis (1965 - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA)
- Subjects
0301 basic medicine ,Nervous system ,endocrine system ,TREK-1 ,[SDV]Life Sciences [q-bio] ,[SDV.NEU.NB]Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,neurological disorders ,Central nervous system ,Review ,Disease ,arrhythmia ,Neuroprotection ,03 medical and health sciences ,0302 clinical medicine ,Smooth muscle ,medicine ,Endocrine system ,modulators ,Pharmacology (medical) ,ComputingMilieux_MISCELLANEOUS ,Ion channel ,Pharmacology ,K2P ,business.industry ,potassium ,lcsh:RM1-950 ,[SDV.NEU.NB] Life Sciences [q-bio]/Neurons and Cognition [q-bio.NC]/Neurobiology ,Depolarization ,3. Good health ,[SDV] Life Sciences [q-bio] ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,ion channel ,business ,human activities ,Neuroscience - Abstract
TREK-1 is the most studied background K2P channel. Its main role is to control cell excitability and maintain the membrane potential below the threshold of depolarization. TREK-1 is multi-regulated by a variety of physical and chemical stimuli which makes it a very promising and challenging target in the treatment of several pathologies. It is mainly expressed in the brain but also in heart, smooth muscle cells, endocrine pancreas, and prostate. In the nervous system, TREK-1 is involved in many physiological and pathological processes such as depression, neuroprotection, pain, and anesthesia. These properties explain why many laboratories and pharmaceutical companies have been focusing their research on screening and developing highly efficient modulators of TREK-1 channels. In this review, we summarize the different roles of TREK-1 that have been investigated so far in attempt to characterize pharmacological tools and new molecules to modulate cellular functions controlled by TREK-1.
- Published
- 2019
- Full Text
- View/download PDF
126. Piezo1 and Piezo2 foster mechanical gating of K2P channels.
- Author
-
Glogowska, Edyta, Arhatte, Malika, Chatelain, Franck C., Lesage, Florian, Xu, Aimin, Grashoff, Carsten, Discher, Dennis E., Patel, Amanda, and Honoré, Eric
- Abstract
Mechanoelectrical transduction is mediated by the opening of different types of force-sensitive ion channels, including Piezo1/2 and the TREK/TRAAK K 2P channels. Piezo1 curves the membrane locally into an inverted dome that reversibly flattens in response to force application. Moreover, Piezo1 forms numerous preferential interactions with various membrane lipids, including cholesterol. Whether this structural architecture influences the functionality of neighboring membrane proteins is unknown. Here, we show that Piezo1/2 increase TREK/TRAAK current amplitude, slow down activation/deactivation, and remove inactivation upon mechanical stimulation. These findings are consistent with a mechanism whereby Piezo1/2 cause a local depletion of membrane cholesterol associated with a prestress of TREK/TRAAK channels. This regulation occurs in mouse fibroblasts between endogenous Piezo1 and TREK-1/2, both channel types acting in concert to delay wound healing. In conclusion, we demonstrate a community effect between different structural and functional classes of mechanosensitive ion channels. [Display omitted] • TREK/TRAAK mechanical activation is upregulated and kinetics are delayed by Piezo1/2 • Piezo1 opening is required for enhancing TREK-1 current amplitude • Local depletion of membrane cholesterol contributes to the upregulation of TREK-1 • Piezo1 upregulates TREK-1/2 in mouse fibroblasts causing a delay in wound healing Glogowska et al. show that Piezo1/2 enhance the mechanical activation of K 2P channels, slow down activation/deactivation, and remove inactivation. Piezo1/2 cause a prestress of TREK/TRAAK attributable to a local depletion of membrane cholesterol. A native regulation between Piezo1 and TREK-1/2 occurs in mouse gingival fibroblasts. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
127. Two-Pore-Domain Potassium (K 2P -) Channels: Cardiac Expression Patterns and Disease-Specific Remodelling Processes.
- Author
-
Wiedmann, Felix, Frey, Norbert, and Schmidt, Constanze
- Subjects
- *
ION channels , *HEART failure , *MEMBRANE potential , *HEART fibrosis , *CARDIAC hypertrophy , *STRAINS & stresses (Mechanics) - Abstract
Two-pore-domain potassium (K2P-) channels conduct outward K+ currents that maintain the resting membrane potential and modulate action potential repolarization. Members of the K2P channel family are widely expressed among different human cell types and organs where they were shown to regulate important physiological processes. Their functional activity is controlled by a broad variety of different stimuli, like pH level, temperature, and mechanical stress but also by the presence of lipids or pharmacological agents. In patients suffering from cardiovascular diseases, alterations in K2P-channel expression and function have been observed, suggesting functional significance and a potential therapeutic role of these ion channels. For example, upregulation of atrial specific K2P3.1 (TASK-1) currents in atrial fibrillation (AF) patients was shown to contribute to atrial action potential duration shortening, a key feature of AF-associated atrial electrical remodelling. Therefore, targeting K2P3.1 (TASK-1) channels might constitute an intriguing strategy for AF treatment. Further, mechanoactive K2P2.1 (TREK-1) currents have been implicated in the development of cardiac hypertrophy, cardiac fibrosis and heart failure. Cardiovascular expression of other K2P channels has been described, functional evidence in cardiac tissue however remains sparse. In the present review, expression, function, and regulation of cardiovascular K2P channels are summarized and compared among different species. Remodelling patterns, observed in disease models are discussed and compared to findings from clinical patients to assess the therapeutic potential of K2P channels. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
128. 大鼠局灶性脑缺血再灌注后海马 CA1 区TREK-1、GFAP 表达变化.
- Author
-
袁建清, 廖春英, and 黄樱
- Abstract
目的 观察新型双孔钾离子通道亚单元TREK-1及胶质纤维酸性蛋白( GFAP)在大鼠局灶性脑缺血再灌注后海马CA1区的表达变化. 方法 建立大鼠大脑中动脉阻塞( MCAO)再灌注模型,应用免疫荧光组化和激光扫描共聚焦观察正常大鼠及MCAO 再灌注后3、7、30天海马CA1区TREK-1和GFAP表达情况,同时应用Western blotting法检测大鼠MCAO再灌注后3、7、30天海马CA1区TREK-1、GFAP蛋白表达. 结果 成年大鼠海马CA1区可见TREK-1和GFAP双标阳性的细胞,而未见有TREK-1和NEUN双标阳性的细胞;MCAO再灌注后3、7、30天海马CA1区TREK-1和GFAP表达逐步上调,与正常大鼠比较差异均有统计学意义(P均<0.05). 结论 大鼠局灶性脑缺血再灌注后海马CA1区TREK-1、GFAP表达均升高. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
129. Swelling-activated and arachidonic acid-induced currents are TREK-1 in rat bladder smooth muscle cells
- Author
-
Mitsuko Fukasaku, Osamu Yamaguchi, and Junko Kimura
- Subjects
Male ,rat urinary bladder ,medicine.medical_specialty ,K+ channel ,TREK-1 ,Voltage clamp ,Urinary Bladder ,030232 urology & nephrology ,Biology ,perforated patch clamp ,Membrane Potentials ,Rats, Sprague-Dawley ,smooth muscle ,03 medical and health sciences ,chemistry.chemical_compound ,Potassium Channels, Tandem Pore Domain ,0302 clinical medicine ,Internal medicine ,medicine ,arachidonic acid ,Animals ,Myocyte ,494.9 ,Reversal potential ,Urinary bladder ,Forskolin ,Colforsin ,Muscle, Smooth ,General Medicine ,Hydrogen-Ion Concentration ,Potassium channel ,Rats ,medicine.anatomical_structure ,Endocrinology ,chemistry ,Biophysics ,Tetradecanoylphorbol Acetate ,Original Article ,Arachidonic acid ,Mechanosensitive channels ,030217 neurology & neurosurgery - Abstract
Using the perforated patch voltage clamp, we investigated swelling-activated ionic channels (SACs) in rat urinary bladder smooth muscle cells. Hypo-osmotic (60%) bath solution increased a membrane current which was inhibited by the SAC inhibitor, gadolinium. The reversal potential of the hypotonicity-induced current shifted in the positive direction by increasing external K(+) concentration. The hypotonicity-induced current was inhibited by extracellular acidic pH, phorbol ester and forskolin. These pharmacological properties are identical to those of arachidonic acid-induced current present in these cells, suggesting the presence of TREK-1, a four-transmembrane two pore domain K(+) channel. Using RT-PCR we screened rat bladder smooth muscles and cerebellum for expression of TREK-1, TREK-2 and TRAAK mRNAs. Only TREK-1 mRNA was expressed in the bladder, while all three were expressed in the cerebellum. We conclude that a mechanosensitive K(+) channel is present in rat bladder myocytes, which is activated by arachidonic acid and most likely is TREK-1. This K(+) channel may have an important role in the regulation of bladder smooth muscle tone during urine storage.
- Published
- 2016
130. TREK-1 potassium channels participate in acute and long-lasting nociceptive hypersensitivity induced by formalin in rats.
- Author
-
García, Guadalupe, Martínez-Rojas, Vladimir A., and Murbartián, Janet
- Subjects
- *
POTASSIUM channels , *RATS , *FORMALDEHYDE , *NEURALGIA , *DORSAL root ganglia , *VISCERAL pain - Abstract
[Display omitted] • Spadin increases the acute and long-lasting formalin-induced nociception in rats. • BL-1249 prevents the pronociceptive effect of spadin in formalin test. • S300A or S333A TREK-1 mutants prevent and reduced the established formalin-induced nociceptive hypersensitivity. • 1% formalin increases TREK-1 protein expression in DRG and dorsal spinal cord in rats. • S300A, S333A and BL-1249 reduced formalin-induced enhanced c-fos protein expression. TREK-1 channels are expressed in small nociceptive dorsal root ganglion (DRG) neurons where they participate in acute inflammatory and neuropathic pain. However, the role of TREK-1 in persistent pain is not well understood. The aim of this study was to investigate the local peripheral and spinal participation of TREK-1 in formalin-induced acute and long-lasting nociceptive hypersensitivity. Local peripheral or intrathecal pre-treatment with spadin, selective blocker of TREK-1, increased acute flinching behavior and secondary mechanical allodynia and hyperalgesia behavior observed 6 days after formalin injection. Local peripheral or intrathecal pre-treatment with BL-1249, selective opener of TREK-1, decreased long-lasting secondary mechanical allodynia and hyperalgesia induced by formalin. Pre-treatment with BL-1249 prevented the pro-nociceptive effect of spadin on acute nociception and long-lasting mechanical allodynia and hyperalgesia in rats. Pre-treatment with two recombinant channels that produce a high TREK-1 current, S300A and S333A (non-phosphorylated state of TREK-1), reduced formalin-induced acute pain and long-lasting mechanical allodynia and hyperalgesia. Besides, post-treatment with S300A, S333A or BL-1249 reversed long-lasting mechanical allodynia and hyperalgesia induced by formalin. Formalin increased TREK-1 expression at 1 and 6 days in DRG and dorsal spinal cord in rats, whereas that it increased c-fos expression at the DRG. Intrathecal repeated transfection of rats with S300A and S333A or injection with BL-1249 reduced formalin-induced enhanced c-fos expression. Data suggest that TREK-1 activity at peripheral and spinal sites reduces neuronal excitability in the process of acute and long-lasting nociception induced by formalin in rats. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
131. Towards Quantum-Chemical Modeling of the Activity of Anesthetic Compounds.
- Author
-
Cukras, Janusz and Sadlej, Joanna
- Subjects
- *
LIPID rafts , *INTERMOLECULAR interactions , *PERTURBATION theory , *MICROTUBULES , *ANESTHETICS , *MOLECULAR dynamics - Abstract
The modeling of the activity of anesthetics is a real challenge because of their unique electronic and structural characteristics. Microscopic approaches relevant to the typical features of these systems have been developed based on the advancements in the theory of intermolecular interactions. By stressing the quantum chemical point of view, here, we review the advances in the field highlighting differences and similarities among the chemicals within this group. The binding of the anesthetics to their partners has been analyzed by Symmetry-Adapted Perturbation Theory to provide insight into the nature of the interaction and the modeling of the adducts/complexes allows us to rationalize their anesthetic properties. A new approach in the frame of microtubule concept and the importance of lipid rafts and channels in membranes is also discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
132. Regulation of TWIK-related K+ channel 1 in the anterior hippocampus of patients with temporal lobe epilepsy with comorbid depression.
- Author
-
Li, Xiao-Li, Tang, Chong-Yang, Wang, Shu, Zhao, Meng, Wang, Xiong-Fei, Li, Tian-Fu, Qi, Xue-Ling, Luan, Guo-Ming, and Guan, Yu-Guang
- Subjects
- *
TEMPORAL lobe epilepsy , *HIPPOCAMPUS (Brain) , *VAGUS nerve , *MENTAL depression , *COMORBIDITY , *APATHY - Abstract
• 31% of TLE patients performed depression. • Hippocampal sclerosis is related to depression. • Immunoreactive TREK-1 neurons increase in depressed TLE hippocampi. • More activated microglia are observed in the depressed hippocampi. Epilepsy with comorbid depression has recently attracted increasing attention. Temporal lobe epilepsy (TLE) may represent an increased risk of developing depression, especially if the seizures do not generalize. The two-pore domain potassium channel-TWIK-related K+ channel (TREK-1) plays important roles in both epilepsy and depression. However, the changes in its expression in patients with epilepsy with comorbid depression remain unclear. In the present study, we analyzed depressive symptoms using neuropsychiatric scales in forty-two patients with drug-resistant TLE, who also underwent EEG in waking and sleeping states, as well as 3.0 T brain MRI. We tested for TREK-1 positive neurons and microglial cells in the anterior hippocampi of patients with drug-resistant TLE with and without comorbid depression (n=5/group). Approximately 31% of patients with TLE had comorbid depression (13/42). Meanwhile, the patients who had hippocampal sclerosis had much higher scores on the depression rating scale. The results indicated the contribution of hippocampal sclerosis to the development of depression. Immunostaining of TREK-1 channels was observed in neurons and glia in the anterior hippocampus. Increased immunoreactivity of TREK-1 neurons was observed in the hippocampi of patients with TLE with comorbid depression compared with nondepressed patients with TLE. TREK-1 was expressed in almost all microglia. Curiously, more activated TREK-1-positive microglia were observed in patients with TLE with depression than in those without depression. The results suggested that a change in TREK-1 immunoreactivity was involved, at least partly, in the development of depression as a comorbidity of TLE. Imbalance of the TREK-1 channel may be a potential target for the treatment of patients with epilepsy with comorbid depression. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
133. TREK-1 channels regulate pressure sensitivity and calcium signaling in trabecular meshwork cells
- Author
-
Mary Elizabeth Hartnett, Colin A. Bretz, Kenneth W. Olsen, Catherine Heurteaux, Monika Lakk, Tam T. T. Phuong, David Križaj, Jackson M Baumann, Oleg Yarishkin, Alan S. Crandall, Institut de pharmacologie moléculaire et cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA)
- Subjects
0301 basic medicine ,Adult ,genetic structures ,Mechanotransduction ,TREK-1 ,Physiology ,[SDV]Life Sciences [q-bio] ,Primary Cell Culture ,TRPV Cation Channels ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Sensitivity and Specificity ,Mechanotransduction, Cellular ,Membrane Potentials ,03 medical and health sciences ,Transient receptor potential channel ,0302 clinical medicine ,Potassium Channels, Tandem Pore Domain ,Trabecular Meshwork ,medicine ,Pressure ,Humans ,Calcium Signaling ,Research Articles ,ComputingMilieux_MISCELLANEOUS ,Calcium signaling ,Membrane potential ,Arachidonic Acid ,Inward-rectifier potassium ion channel ,Chemistry ,Middle Aged ,Potassium channel ,eye diseases ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,TRPV4 ,Calcium ion homeostasis ,Commentary ,Trabecular meshwork ,sense organs ,030217 neurology & neurosurgery ,Research Article - Abstract
The trabecular meshwork (TM) plays a fundamental role in intraocular pressure regulation, but its mechanotransduction pathway is poorly understood. Yarishkin et al. show that the mechanosensing channel TREK-1 regulates TM membrane potential, pressure sensitivity, calcium homeostasis, and impedance., Mechanotransduction by the trabecular meshwork (TM) is an essential component of intraocular pressure regulation in the vertebrate eye. This process is compromised in glaucoma but is poorly understood. In this study, we identify transient receptor potential vanilloid isoform 4 (TRPV4) and TWIK-related potassium channel-1 (TREK-1) as key molecular determinants of TM membrane potential, pressure sensitivity, calcium homeostasis, and transcellular permeability. We show that resting membrane potential in human TM cells is unaffected by “classical” inhibitors of voltage-activated, calcium-activated, and inwardly rectifying potassium channels but is depolarized by blockers of tandem-pore K+ channels. Using gene profiling, we reveal the presence of TREK-1, TASK-1, TWIK-2, and THIK transcripts in TM cells. Pressure stimuli, arachidonic acid, and TREK-1 activators hyperpolarize these cells, effects that are antagonized by quinine, amlodipine, spadin, and short-hairpin RNA–mediated knockdown of TREK-1 but not TASK-1. Activation and inhibition of TREK-1 modulates [Ca2+]TM and lowers the impedance of cell monolayers. Together, these results suggest that tensile homeostasis in the TM may be regulated by balanced, pressure-dependent activation of TRPV4 and TREK-1 mechanotransducers.
- Published
- 2018
- Full Text
- View/download PDF
134. Piezo1 and Piezo2 foster mechanical gating of K 2P channels.
- Author
-
Glogowska E, Arhatte M, Chatelain FC, Lesage F, Xu A, Grashoff C, Discher DE, Patel A, and Honoré E
- Subjects
- Animals, Cholesterol metabolism, Fibroblasts cytology, Fibroblasts metabolism, Gingiva cytology, Gingiva metabolism, HEK293 Cells, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Potassium Channels, Tandem Pore Domain genetics, Ion Channel Gating, Ion Channels physiology, Mechanotransduction, Cellular, Potassium Channels, Tandem Pore Domain metabolism
- Abstract
Mechanoelectrical transduction is mediated by the opening of different types of force-sensitive ion channels, including Piezo1/2 and the TREK/TRAAK K
2P channels. Piezo1 curves the membrane locally into an inverted dome that reversibly flattens in response to force application. Moreover, Piezo1 forms numerous preferential interactions with various membrane lipids, including cholesterol. Whether this structural architecture influences the functionality of neighboring membrane proteins is unknown. Here, we show that Piezo1/2 increase TREK/TRAAK current amplitude, slow down activation/deactivation, and remove inactivation upon mechanical stimulation. These findings are consistent with a mechanism whereby Piezo1/2 cause a local depletion of membrane cholesterol associated with a prestress of TREK/TRAAK channels. This regulation occurs in mouse fibroblasts between endogenous Piezo1 and TREK-1/2, both channel types acting in concert to delay wound healing. In conclusion, we demonstrate a community effect between different structural and functional classes of mechanosensitive ion channels., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
135. Prognostic significance of the TREK-1 K2P potassium channels in prostate cancer
- Author
-
Fang Ning Wan, Da Long Cao, Hai Liang Zhang, Guo Hai Shi, Yao Zhu, Gui Ming Zhang, Dingwei Ye, Xiao Jian Qin, and Bo Dai
- Subjects
Biochemical recurrence ,Oncology ,Male ,medicine.medical_specialty ,Pathology ,endocrine system ,TREK-1 ,Blotting, Western ,Transplantation, Heterologous ,Mice, Nude ,Kaplan-Meier Estimate ,medicine.disease_cause ,Prostate cancer ,Potassium Channels, Tandem Pore Domain ,Prostate ,Internal medicine ,Cell Line, Tumor ,medicine ,biochemical recurrence ,Animals ,Humans ,Aged ,Cell Proliferation ,Proportional Hazards Models ,Mice, Inbred BALB C ,business.industry ,Cell growth ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Cycle ,Cancer ,Prostatic Neoplasms ,Cell cycle ,Middle Aged ,medicine.disease ,prostate cancer ,Prognosis ,Immunohistochemistry ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,HEK293 Cells ,RNA Interference ,business ,Carcinogenesis ,human activities ,Research Paper - Abstract
// Gui-Ming Zhang 1, 2 , Fang-Ning Wan 1, 2 , Xiao-Jian Qin 1 , Da-Long Cao 1 , Hai-Liang Zhang 1 , Yao Zhu 1 , Bo Dai 1 , Guo-Hai Shi 1 , Ding-Wei Ye 1 1 Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China 2 Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China Correspondence to: Guo-Hai Shi, e-mail: guohaishi@126.com Ding-Wei Ye, e-mail: yedingwei1963@126.com Keywords: TREK-1, prostate cancer, biochemical recurrence, cell proliferation, cell cycle Received: March 05, 2015 Accepted: April 13, 2015 Published: April 21, 2015 ABSTRACT Background: TREK-1 channels belong to the two-pore domain potassium channel superfamily and play an important role in central nervous system diseases. However, few studies have examined their role in carcinogenesis. Methods: In this study, we assessed the expression of TREK-1 in 100 prostate cancer (PCa) tissues using immunohistochemistry and further analyzed its clinicopathological significance. Next, cell proliferation and cell cycle analysis were carried out on human PCa PC-3 cell lines where TREK-1 was stably knockdown. Results: We found that compared with normal prostate tissues, PCa tissues showed overexpressed TREK-1 levels and TREK-1 levels were positively associated with Gleason score and T staging. High level of TREK-1 expression was related to shorter castration resistance free survival (CRFS). Furthermore, knockdown of TREK-1 significantly inhibited PCa cell proliferation in vitro and in vivo , and induced a G1/S cell cycle arrest. Conclusion: Our results suggest that TREK-1 might be a biomarker in CRFS judgment of PCa, as well as a potential therapeutic target.
- Published
- 2015
136. Effects of fluoxetine on protein expression of potassium ion channels in the brain of chronic mild stress rats
- Author
-
Xiaoliang Wang, Xianfang Rong, Weiping Wang, Chun-Lin Chen, and Ling Wang
- Subjects
Fluoxetine ,medicine.diagnostic_test ,TREK-1 ,Chemistry ,CMS ,Kv2.1 ,Depression ,lcsh:RM1-950 ,Anhedonia ,Hippocampus ,Pharmacology ,Protein expression ,Pathophysiology ,Potassium channel ,Potassium ion channel ,lcsh:Therapeutics. Pharmacology ,Western blot ,Mild stress ,medicine ,Rat ,Original Article ,General Pharmacology, Toxicology and Pharmaceutics ,medicine.symptom ,medicine.drug - Abstract
The purpose of this study is to investigate the expression of major potassium channel subtypes in the brain of chronical mild stress (CMS) rats and reveal the effects of fluoxetine on the expression of these channels. Rats were exposed to a variety of unpredictable stress for three weeks and induced anhedonia, lower sucrose preference, locomotor activity and lower body weight. The protein expressions were determined by Western blot. CMS significantly increased the expression of Kv2.1 channel in frontal cortex but not in hippocampus, and the expression level was normalized after fluoxetine treatment. The expression of TREK-1 channel was also obviously increased in frontal cortex in CMS rats. Fluoxetine treatment might prevent this increase. However, the expression of Kv3.1 and Kv4.2 channels was considerably decreased in hippocampus after CMS, and was not affected by fluoxetine. These results suggest that different subtypes of potassium channels are associated with the pathophysiology of depression and that the therapeutical effects of fluoxetine may relate to Kv2.1 and TREK-1 potassium channels., Graphical abstract This study indicates that different subtypes of potassium channels are associated with the pathogenesis of depression. Kv2.1 and TREK-1 channels are related to the function of frontal cortex and might be regulated by fluoxetine, while Kv3.1 and Kv4.2 are related to the functions of hippocampus and can not be regulated by fluoxetine. Therefore, Kv2.1 and TREK-1 channels might be new drug targets for antidepressant.
- Published
- 2015
137. Maternal separation induces changes in TREK-1 and 5HT1A expression in brain areas involved in the stress response in a sex-dependent way.
- Author
-
Francis-Oliveira, J., Shieh, I.C, Vilar Higa, G.S., Barbosa, M.A., and De Pasquale, R.
- Subjects
- *
DENTATE gyrus , *LIFE change events , *AMYGDALOID body , *IMMOBILIZATION stress , *MODERN society , *DEPRESSION in men , *DESPAIR - Abstract
• Postnatal stress induces behavioural despair in female, but not male juvenile rats. • MS increases TREK-1 in basolateral amygdala and prelimbic cortex. • 5HT 1A reduction in dentate gyrus is associated to behavioural despair in females. • TREK-1 increase in dentate gyrus is associated to behavioural despair in females. • More 5HT 1A expression in basolateral amygdala is linked to resilience in males. Depression is a prevalent disease in modern society, and has been linked to stressful events at early ages. Women are more susceptible to depression, and the neural basis for this are still under investigation. Serotonin is known to be involved in depression, and a decrease in 5HT 1A expression is observed on temporal and cortical areas in both men and women with depression. As knockout animals for TREK-1 are resilient to depression, this channel has emerged as a new potential pharmacological target for depression treatment. In this study, maternal separation (MS) was used to emulate early-life stress, and evaluate behaviour, as well as TREK-1 and 5HT 1A expression in the brain using immunohistochemistry. In juvenile females, 5HT 1A reduction coupled to increased TREK-1 in the dentate gyrus (DG) was associated with behavioural despair, as well as increased TREK-1 expression in basolateral amygdala (BLA) and prelimbic cortex (PL). In juvenile males, MS induced an increase in 5HT 1A in the BLA, and in TREK-1 in the PL, while no behavioural despair was observed. Anhedonia and anxiety-like behaviour were not induced by MS. We conclude stress-induced increase in TREK-1 in PL and GD is associated to depression, while 5HT 1A changes coupled to TREK-1 changes may be necessary to induce depression, with females being more vulnerable to MS effects than males. Thus, TREK-1 and 5HT 1A may be potential pharmacological targets for antidepressants development. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
138. Quercetin relieves D-amphetamine-induced manic-like behaviour through activating TREK-1 potassium channels in mice.
- Author
-
Ren K, Liu H, Guo B, Li R, Mao H, Xue Q, Yao H, Wu S, Bai Z, and Wang W
- Subjects
- Animals, Dextroamphetamine, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Potassium Channels, Tandem Pore Domain, Quercetin pharmacology
- Abstract
Background and Purpose: Quercetin is a well-known plant flavonoid with neuroprotective properties. Earlier work suggested it may relieve psychiatric disorders, cognition deficits and memory dysfunction through anti-oxidant and/or radical scavenging mechanisms. In addition, quercetin modulated the physiological function of some ion channels. However, the detailed ionic mechanisms of the bioeffects of quercetin remain unknown., Experimental Approach: Effects of quercetin on neuronal activities in the prefrontal cortex (PFC) and its ionic mechanisms were analysed by calcium imaging using mice bearing a green fluorescent protein, calmodulin, and M13 fusion protein and patch clamp in acute brain slices from C57BL/6 J mice and in HEK 293 cells. The possible ionic mechanism of action of quercetin on D-amphetamine-induced manic-like effects in mice was explored with c-fos staining and the open field behaviour test., Key Results: Quercetin reduced calcium influx triggered by PFC pyramidal neuronal activity. This effect involved increasing the rheobase of neuronal firing through decreasing membrane resistance following quercetin treatment. Spadin, a blocker of TREK-1 potassium channels, also blocked the effect of quercetin on the membrane resistance and neuronal firing. Further, spadin blocked the neuroprotective effects of quercetin. The effects of quercetin on TREK-1 channels could be mimicked by GF109203X, a protein kinase C inhibitor. In vivo, injection of quercetin relieved the manic hyperlocomotion in mice, induced by D-amphetamine. This action was partly alleviated by spadin., Conclusion and Implications: TREK-1 channels are a novel target for quercetin, by inhibiting PKC. This action could contribute to both the neuroprotective and anti-manic-like effects., (© 2021 The British Pharmacological Society.)
- Published
- 2021
- Full Text
- View/download PDF
139. Human adrenal glomerulosa cells express K2P and GIRK potassium channels that are inhibited by ANG II and ACTH.
- Author
-
Enyeart JJ and Enyeart JA
- Subjects
- Adolescent, Adult, Aldosterone biosynthesis, Arachidonic Acid pharmacology, Autopsy, Child, Colforsin pharmacology, Female, G Protein-Coupled Inwardly-Rectifying Potassium Channels metabolism, Gene Expression, Humans, Kv1.4 Potassium Channel antagonists & inhibitors, Kv1.4 Potassium Channel metabolism, Male, Membrane Potentials drug effects, Membrane Potentials physiology, Middle Aged, Nerve Tissue Proteins antagonists & inhibitors, Nerve Tissue Proteins metabolism, Patch-Clamp Techniques, Potassium Channels, Tandem Pore Domain antagonists & inhibitors, Potassium Channels, Tandem Pore Domain metabolism, Primary Cell Culture, Zona Glomerulosa cytology, Zona Glomerulosa drug effects, Adrenocorticotropic Hormone pharmacology, Angiotensin II pharmacology, G Protein-Coupled Inwardly-Rectifying Potassium Channels genetics, Kv1.4 Potassium Channel genetics, Nerve Tissue Proteins genetics, Potassium Channels, Tandem Pore Domain genetics, Zona Glomerulosa metabolism
- Abstract
In whole cell patch clamp recordings, it was discovered that normal human adrenal zona glomerulosa (AZG) cells express members of the three major families of K
+ channels. Among these are a two-pore (K2P) leak-type and a G protein-coupled, inwardly rectifying (GIRK) channel, both inhibited by peptide hormones that stimulate aldosterone secretion. The K2P current displayed properties identifying it as TREK-1 (KCNK2). This outwardly rectifying current was activated by arachidonic acid and inhibited by angiotensin II (ANG II), adrenocorticotrophic hormone (ACTH), and forskolin. The activation and inhibition of TREK-1 was coupled to AZG cell hyperpolarization and depolarization, respectively. A second K2P channel, TASK-1 (KCNK3), was expressed at a lower density in AZG cells. Human AZG cells also express inwardly rectifying K+ current(s) (KIR ) that include quasi-instantaneous and time-dependent components. This is the first report demonstrating the presence of KIR in whole cell recordings from AZG cells of any species. The time-dependent current was selectively inhibited by ANG II, and ACTH, identifying it as a G protein-coupled (GIRK) channel, most likely KIR 3.4 (KCNJ5). The quasi-instantaneous KIR current was not inhibited by ANG II or ACTH and may be a separate non-GIRK current. Finally, AZG cells express a voltage-gated, rapidly inactivating K+ current whose properties identified as KV 1.4 (KCNA4), a conclusion confirmed by Northern blot. These findings demonstrate that human AZG cells express K2P and GIRK channels whose inhibition by ANG II and ACTH is likely coupled to depolarization-dependent secretion. They further demonstrate that human AZG K+ channels differ fundamentally from the widely adopted rodent models for human aldosterone secretion.- Published
- 2021
- Full Text
- View/download PDF
140. Neuroglial potassium channels in health and disease
- Author
-
Perić, M., Andjus, Pavle R., Perić, M., and Andjus, Pavle R.
- Abstract
Astrocytes are glial cells that contain a set of significant roles in maintaining the optimal conditions for the intercellular communications and functions in the central nervous system (CNS). They are essential for the control of extracellular ion homeostasis, neurotransmitter concentration, water balance and the support of the blood-brain barrier. Glial cells show a hyperpolarized membrane potential with a high K+ conductance, implicating the importance of potassium channels in these cells. The inwardly rectifying potassium channel (Kir) expressed in glial cells has the basic function in setting the cell membrane potential which regulates the transmembrane gradients of glutamate and other neurotransmitters, as well as of transported molecules. Through the mechanism of potassium buffering, astrocytes take up excess K+, siphon it towards sites with lower concentrations providing the equilibrium K+ concentration in the extracellular environment. Kir4.1 has the major role in spatial buffering and it is mainly expressed in astrocytes in CNS. It was revealed that other glial cells also express Kir4.1 with the role in maintaining membrane potential, cellular development and physiology.Two-pore domain K+ channels (K2P) TWIK-1 and TREK-1 were showed to play a part in astrocytic K+ passive conductance, although recent knockdown studies challenge these findings. Alterations in the expression and function of potassium channels are shown in many deleterious neural conditions. A reduced expression of Kir4.1 has been shown in amyotrophic lateral sclerosis, Alzheimer;'s disease, and Huntington disease. Antibodies against Kir4.1 were found in the serum of patients with multiple sclerosis. Cells with a decreased Kir4.1 expression show a significant cell membrane depolarization and impaired K+ and glutamate uptake that lead to long term CNS dysfunction. Recent studies on K2Ps revealed an important role for TWIK-1 and TREK-1 in glutamatergic synaptic transmission and thus their poten
- Published
- 2017
141. Le récepteur 3 de la neurotensine/Sortiline dans la régulation de l’état dépressif
- Author
-
Moreno, Sébastien, Institut de pharmacologie moléculaire et cellulaire ( IPMC ), Université Nice Sophia Antipolis ( UNS ), Université Côte d'Azur ( UCA ) -Université Côte d'Azur ( UCA ) -Centre National de la Recherche Scientifique ( CNRS ), Université Côte d'Azur, Jean Mazella, Institut de pharmacologie moléculaire et cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA)
- Subjects
Trouble dépressif ,Neurotensine ,TREK-1 ,Depressive disorder ,[ SDV.BC ] Life Sciences [q-bio]/Cellular Biology ,NTSR3 ,[ SDV.MHEP ] Life Sciences [q-bio]/Human health and pathology ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Sortilin ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,Neurotensin ,Sortiline - Abstract
Major depressive disorder is a condition that affects 20% of the population and is the leading cause of morbidity and disability worldwide. Recently, the TREK-1 potassium channel has been shown to be a potential target in the treatment of depression. The deletion of this channel or its blocking by a derived peptide resulting from the maturation of Sortilin, propeptide (PE), or its synthetic analogue Spadin, results in a phenotype of resistance to depression in mice. Sortilin is a protein able to bind with TREK-1 but also with the neurotrophic factor BDNF, an important factor for neuronal viability and depressive state regulation. Sortilin is therefore involved in regulating the intracellular addressing of TREK-1 and BDNF. Initially, my work focused on the consequences of the deletion of the Sortilin gene (sort1-/-) on the TREK-1 and BDNF addressing, and the neurotensinergic system. The results showed a decrease in TREK-1 membrane expression at the cerebral level and an increase in BDNF. All of these changes lead the Sort1-/- mice to develop a phenotype of resistance to depression. In addition, these mice show an increase in brain neurotensin concentration and its receptor 2, leading to increased resistance to pain perception. In a second phase, I was interested in whether PE, a potential antidepressant, showed serum variations in depressed patients and could be an indicator of depressive syndrome. We showed that the serum PE level is significantly reduced in depressed people, a level restored after treatment with antidepressants. In conclusion, Sortilin plays a major key in the regulation of depressive disorder and also in nociception.; Le trouble dépressif majeur est une pathologie qui atteint 20% de la population et qui représente le premier facteur de morbidité et d’incapacité au niveau mondial. Récemment, le canal potassique TREK-1 est une cible potentielle avérée dans le traitement de la dépression. La délétion de ce canal ou son blocage par un peptide dérivé résultant de la maturation de la sortiline, le propeptide (PE) ou son analogue synthétique la Spadine, résulte en un phénotype de résistance à la dépression. La sortiline est une protéine capable de s’associer à TREK-1 mais également au facteur neurotrophique BDNF important pour la viabilité neuronale et la régulation de l’état dépressif. La sortiline est donc impliquée dans la régulation de l’adressage intracellulaire de TREK-1 et du BDNF. Mes travaux se sont d’abords focalisés sur les conséquences de la délétion du gène codant pour la sortiline (Sort1-/-) sur l’adressage de TREK-1 et du BDNF, et également sur le système neurotensinergique. Les résultats révèlent au niveau cérébral une diminution de l’expression membranaire de TREK-1 et une augmentation de BDNF. L’ensemble de ces modifications conduisent les souris Sort1-/- à développer un phénotype de résistance à la dépression. De plus, ces souris présentent une augmentation de la concentration en neurotensine cérébrale ainsi que de son récepteur 2, ce qui entraine une résistance à la douleur. Par la suite, nous avons montré une diminution de la concentration sérique du PE chez les personnes dépressives, un niveau restauré après traitement avec des antidépresseurs. En conclusion, la sortiline joue un rôle prépondérant dans la régulation du trouble dépressif et aussi dans la nociception.
- Published
- 2017
142. TREK-1 Channel Expression in Smooth Muscle as a Target for Regulating Murine Intestinal Contractility: Therapeutic Implications for Motility Disorders
- Author
-
Anthony Lewis, Mohsen Seifi, Maria Papanikolaou, Jerome D. Swinny, Ruolin Ma, and James Brown
- Subjects
0301 basic medicine ,TREK-1 ,Physiology ,Motility ,Ileum ,APC-PAID ,contractility ,lcsh:Physiology ,law.invention ,Contractility ,smooth muscle ,03 medical and health sciences ,Confocal microscopy ,law ,Physiology (medical) ,medicine ,K2P channels ,Original Research ,lcsh:QP1-981 ,colon ,Chemistry ,Smooth muscle contraction ,Riluzole ,Cell biology ,030104 developmental biology ,Flufenamic acid ,medicine.anatomical_structure ,motility ,Immunohistochemistry ,ileum ,medicine.drug - Abstract
Gastrointestinal (GI) motility disorders such as irritable bowel syndrome (IBS) can occur when coordinated smooth muscle contractility is disrupted. Potassium (K+) channels regulate GI smooth muscle tone and are key to GI tract relaxation, but their molecular and functional phenotypes are poorly described. Here we define the expression and functional roles of mechano-gated K2P channels in mouse ileum and colon. Expression and distribution of the K2P channel family were investigated using quantitative RT-PCR (qPCR), immunohistochemistry and confocal microscopy. The contribution of mechano-gated K2P channels to mouse intestinal muscle tension was studied pharmacologically using organ bath. Multiple K2P gene transcripts were detected in mouse ileum and colon whole tissue preparations. Immunohistochemistry confirmed TREK-1 expression was smooth muscle specific in both ileum and colon, whereas TREK-2 and TRAAK channels were detected in enteric neurons but not smooth muscle. In organ bath, mechano-gated K2P channel activators (Riluzole, BL-1249, flufenamic acid, and cinnamyl 1-3,4-dihydroxy-alpha-cyanocinnamate) induced relaxation of KCl and CCh pre-contracted ileum and colon tissues and reduced the amplitude of spontaneous contractions. These data reveal the specific expression of mechano-gated K2P channels in mouse ileum and colon tissues and highlight TREK-1, a smooth muscle specific K2P channel in GI tract, as a potential therapeutic target for combating motility pathologies arising from hyper-contractility.
- Published
- 2017
- Full Text
- View/download PDF
143. Involvement of intracellular transport in TREK-1c current run-up in 293T cells
- Author
-
Hiroko Matsuda, Naaz Andharia, Mikio Hayashi, Ancy Joseph, and Masayoshi Okada
- Subjects
0301 basic medicine ,intracellular transport ,TREK-1 ,microtubule-associated protein 2 ,Potassium ,Biophysics ,Intracellular Space ,chemistry.chemical_element ,two-pore-domain K+ channel ,Biology ,293T cell ,Biochemistry ,Cell membrane ,03 medical and health sciences ,0302 clinical medicine ,Potassium Channels, Tandem Pore Domain ,medicine ,Humans ,surface localization ,Cytoskeleton ,Protein Stability ,run-up ,HEK 293 cells ,Cell Membrane ,Cell biology ,Transport protein ,Electrophysiological Phenomena ,ezrin ,Cytoskeletal Proteins ,Kinetics ,Protein Transport ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,chemistry ,Current (fluid) ,human activities ,Microtubule-Associated Proteins ,030217 neurology & neurosurgery ,Intracellular transport ,Intracellular ,Research Paper - Abstract
The TREK-1 channel, the TWIK-1-related potassium (K+) channel, is a member of a family of 2-pore-domain K+ (K2P) channels, through which background or leak K+ currents occur. An interesting feature of the TREK-1 channel is the run-up of current: i.e. the current through TREK-1 channels spontaneously increases within several minutes of the formation of the whole-cell configuration. To investigate whether intracellular transport is involved in the run-up, we established 293T cell lines stably expressing the TREK-1c channel (K2P2.1) and examined the effects of inhibitors of membrane protein transport, N-methylmaleimide (NEM), brefeldin-A, and an endocytosis inhibitor, pitstop2, on the run-up. The results showing that NEM and brefeldin-A inhibited and pitstop2 facilitated the run-up suggest the involvement of intracellular protein transport. Correspondingly, in cells stably expressing the mCherry-TREK-1 fusion protein, NEM decreased and pitstop2 increased the cell surface localization of the fusion protein. Furthermore, the run-up was inhibited by the intracellular application of a peptide of the C-terminal fragment TREK335–360, corresponding to the interaction site with microtubule-associated protein 2 (Mtap2). This peptide also inhibited the co-immunoprecipitation of Mtap2 with anti-mCherry antibody. The extracellular application of an ezrin inhibitor (NSC668394) also suppressed the run-up and surface localization of the fusion protein. The co-application of these inhibitors abolished the TREK-1c current, suggesting that the additive effects of ezrin and Mtap2 enhance the surface expression of TREK-1c channels and the run-up. These findings clearly showed the involvement of intracellular transport in TREK-1c current run-up and its mechanism.
- Published
- 2017
144. Tandem pore TWIK-related potassium channels and neuroprotection
- Author
-
J. Antonio Lamas and Diego Fernández-Fernández
- Subjects
TREK-2 ,0301 basic medicine ,TREK-1 ,Review ,Neuroprotection ,lcsh:RC346-429 ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Developmental Neuroscience ,medicine ,lcsh:Neurology. Diseases of the nervous system ,TREK channels ,lysophospholipids ,Chemistry ,TRAAK ,neuroprotection ,free fatty acids ,riluzole ,medicine.disease ,Potassium channel ,Riluzole ,030104 developmental biology ,human activities ,Neuroscience ,030217 neurology & neurosurgery ,medicine.drug - Abstract
TWIK-related potassium channels (TREK) belong to a subfamily of the two-pore domain potassium channels family with three members, TREK1, TREK2 and TWIK-related arachidonic acid-activated potassium channels. The two-pore domain potassium channels is the last big family of channels being discovered, therefore it is not surprising that most of the information we know about TREK channels predominantly comes from the study of heterologously expressed channels. Notwithstanding, in this review we pay special attention to the limited amount of information available on native TREK-like channels and real neurons in relation to neuroprotection. Mainly we focus on the role of free fatty acids, lysophospholipids and other neuroprotective agents like riluzole in the modulation of TREK channels, emphasizing on how important this modulation may be for the development of new therapies against neuropathic pain, depression, schizophrenia, epilepsy, ischemia and cardiac complications.
- Published
- 2019
- Full Text
- View/download PDF
145. A regulatory domain in the K2P2.1 (TREK-1) carboxyl-terminal allows for channel activation by monoterpenes.
- Author
-
Arazi, Eden, Blecher, Galit, and Zilberberg, Noam
- Subjects
- *
PAIN perception , *MONOTERPENES , *ARACHIDONIC acid , *PHOSPHOLIPIDS , *CHANNEL flow - Abstract
Potassium K 2P ('leak') channels conduct current across the entire physiological voltage range and carry leak or 'background' currents that are, in part, time- and voltage-independent. K 2P 2.1 channels (i.e., TREK-1, KCNK2) are highly expressed in excitable tissues, where they play a key role in the cellular mechanisms of neuroprotection, anesthesia, pain perception, and depression. Here, we report for the first time that human K 2P 2.1 channel activity is regulated by monoterpenes (MTs). We found that cyclic, aromatic monoterpenes containing a phenol moiety, such as carvacrol, thymol and 4-IPP had the most profound effect on current flowing through the channel (up to a 6-fold increase). By performing sequential truncation of the carboxyl-terminal domain of the channel and testing the activity of several channel regulators, we identified two distinct regulatory domains within this portion of the protein. One domain, as previously reported, was needed for regulation by arachidonic acid, anionic phospholipids, and temperature changes. Within a second domain, a triple arginine residue motif (R344–346), an apparent PIP 2 -binding site, was found to be essential for regulation by holding potential changes and important for regulation by monoterpenes. • The human K 2P 2.1 channel activity is regulated by monoterpenes. • Aromatic monoterpenes containing a phenol moiety were found to be the most potent. • The carboxyl-terminal of the channel is essential for regulation by monoterpenes. • A three arginine residue motif is important for regulation by monoterpenes. • This motif is essential for regulation by holding potential changes. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
146. Circadian photoperiod alters TREK-1 channel function and expression in dorsal raphe serotonergic neurons via melatonin receptor 1 signaling.
- Author
-
Giannoni-Guzmán MA, Kamitakahara A, Magalong V, Levitt P, and McMahon DG
- Subjects
- Animals, Electrophysiology, Female, Humans, Male, Melatonin metabolism, Photoperiod, Potassium Channels, Tandem Pore Domain genetics, Receptors, Melatonin metabolism, Serotonin metabolism, Dorsal Raphe Nucleus metabolism, Potassium Channels, Tandem Pore Domain metabolism, Serotonergic Neurons metabolism
- Abstract
Seasonal day length has been linked to the prevalence of mood disorders, and however, the mechanisms underlying this relationship remain unknown. Previous work in our laboratory has shown that developmental exposure to seasonal photoperiods has enduring effects on the activity of mouse dorsal raphe serotonergic neurons, their intrinsic electrical properties, as well as on depression and anxiety-related behaviors. Here we focus on the possible ionic mechanisms that underlie the observed programming of the electrophysiological properties of serotonin neurons, focusing on the twin-pore K + channels TREK-1 and TASK-1 that set resting membrane potential and regulate excitability. Pharmacological inhibition of TREK-1 significantly increased spike frequency in Short and Equinox photoperiods, but did not further elevate the firing rate in slices from Long photoperiod mice, suggesting that TREK-1 function is reduced in Long photoperiods. In contrast, inhibition of TASK-1 resulted in increases in firing rates across all photoperiods, suggesting that it contributes to setting excitability, but is not regulated by photoperiod. We then quantified Kcnk2 mRNA levels specifically in dorsal raphe 5-HT neurons using triple-label RNAscope. We found that Long photoperiod significantly reduced levels of Kcnk2 in serotonin neurons co-expressing Tph2, and Pet-1. Photoperiodic effects on the function and expression of TREK-1 were blocked in melatonin 1 receptor knockout (MT-1KO) mice, consistent with previous findings that MT-1 signaling is necessary for photoperiodic programming of dorsal raphe 5-HT neurons. Taken together these results indicate that photoperiodic regulation of TREK-1 expression and function plays a key role in photoperiodic programming the excitability of dorsal raphe 5-HT neurons., (© 2020 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
147. Genetic and pharmacological inhibition of two-pore domain potassium channel TREK-1 alters depression-related behaviors and neuronal plasticity in the hippocampus in mice.
- Author
-
Wu F, Sun H, Gong W, Li X, Pan Z, Shan H, and Zhang Z
- Subjects
- Animals, Bridged Bicyclo Compounds administration & dosage, Depression drug therapy, Depression psychology, Hippocampus drug effects, Male, Mice, Mice, Inbred C57BL, Neuronal Plasticity drug effects, Oxazoles administration & dosage, Peptides administration & dosage, RNA, Small Interfering administration & dosage, Depression genetics, Hippocampus physiology, Neuronal Plasticity physiology, Potassium Channels, Tandem Pore Domain antagonists & inhibitors, Potassium Channels, Tandem Pore Domain genetics
- Abstract
Introduction: The two-pore domain potassium channel TREK-1 is a member of background K
+ channels that are thought to provide baseline regulation of membrane excitability. Recent studies have highlighted the putative role of TREK-1 in the action of antidepressants, and its antagonists might be potentially effective antidepressants. However, the mechanisms underlying the actions of TREK-1 are not yet fully understood., Methods: The expression of TREK-1 was examined in a mouse model of chronic unpredictable mild stress (CUMS) using immunoblotting. Neuron-specific genetic manipulation of TREK-1 was performed through adeno-associated virus. Behavioral tests were performed to evaluate depression-related behaviors. Electrophysiological recordings were used to evaluate synaptic plasticity. Golgi staining was used to examine neuroplasticity., Results: TREK-1 expression was increased in the mouse hippocampus after CUMS. Knockdown of TREK-1 in hippocampal neurons significantly attenuated depressive-like behaviors and prevented the decrease of CUMS-induced synaptic proteins in mice. Further examination indicated that neuron-specific knockdown of TREK-1 in the hippocampus prevented stress-induced impairment of glutamatergic synaptic transmission in the CA1 region. Moreover, chronic TREK-1 inhibition protected against CUMS-induced depressive-like behaviors and impairment of synaptogenesis in the hippocampus., Conclusion: Our results indicate a role for TREK-1 in the modulation of synaptic plasticity in a mouse model of depression. These findings will provide insight into the pathological mechanism of depression and further evidence for a novel target for antidepressant treatment., (© 2020 The Authors. CNS Neuroscience & Therapeutics published by John Wiley & Sons Ltd.)- Published
- 2021
- Full Text
- View/download PDF
148. Mechanosensitive TREK-1 two-pore-domain potassium (K 2P ) channels in the cardiovascular system.
- Author
-
Wiedmann F, Rinné S, Donner B, Decher N, Katus HA, and Schmidt C
- Subjects
- Animals, Anti-Arrhythmia Agents pharmacology, Arrhythmias, Cardiac metabolism, Biomechanical Phenomena physiology, Cardiomegaly metabolism, Drug Development, Heart Failure metabolism, Humans, Lipid Bilayers metabolism, Potassium Channels, Tandem Pore Domain genetics, Xenopus laevis, Anti-Arrhythmia Agents metabolism, Cardiovascular System metabolism, Myocytes, Cardiac metabolism, Potassium metabolism, Potassium Channels, Tandem Pore Domain metabolism
- Abstract
TWIK-related K
+ channel (TREK-1) two-pore-domain potassium (K2P ) channels mediate background potassium currents and regulate cellular excitability in many different types of cells. Their functional activity is controlled by a broad variety of different physiological stimuli, such as temperature, extracellular or intracellular pH, lipids and mechanical stress. By linking cellular excitability to mechanical stress, TREK-1 currents might be important to mediate parts of the mechanoelectrical feedback described in the heart. Furthermore, TREK-1 currents might contribute to the dysregulation of excitability in the heart in pathophysiological situations, such as those caused by abnormal stretch or ischaemia-associated cell swelling, thereby contributing to arrhythmogenesis. In this review, we focus on the functional role of TREK-1 in the heart and its putative contribution to cardiac mechanoelectrical coupling. Its cardiac expression among different species is discussed, alongside with functional evidence for TREK-1 currents in cardiomyocytes. In addition, evidence for the involvement of TREK-1 currents in different cardiac arrhythmias, such as atrial fibrillation or ventricular tachycardia, is summarized. Furthermore, the role of TREK-1 and its interaction partners in the regulation of the cardiac heart rate is reviewed. Finally, we focus on the significance of TREK-1 in the development of cardiac hypertrophy, cardiac fibrosis and heart failure., Competing Interests: Declaration of competing interest The authors declare that they have no competing financial interests or personal relationships that could be perceived to have influenced the work reported in this paper., (Copyright © 2020 Elsevier Ltd. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
149. Étude des mécanismes de libération du propeptide de la sortiline et de ses effets sur l’homéostasie glucidique
- Author
-
Hivelin, Céline, Institut de pharmacologie moléculaire et cellulaire ( IPMC ), Université Nice Sophia Antipolis ( UNS ), Université Côte d'Azur ( UCA ) -Université Côte d'Azur ( UCA ) -Centre National de la Recherche Scientifique ( CNRS ), Université Côte d'Azur, Thierry Coppola, Institut de pharmacologie moléculaire et cellulaire (IPMC), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA)
- Subjects
[SDV.SA]Life Sciences [q-bio]/Agricultural sciences ,Homéostasie glucidique ,Sécrétion d'insuline ,TREK-1 ,Insulin secretion ,Spadine ,Glucose homeostasis ,Sortilin ,[ SDV.SA ] Life Sciences [q-bio]/Agricultural sciences ,Sortiline - Abstract
In France, approximately 15% of the population is obese and this number keeps rising up every year. Obesity is a major cause of diabetes, inducing an increase of the number of fat-filled cells, called the adipocytes, and a peripheral insulin resistance. This increase of the number of adipocytes is associated with a decrease of sortilin expression, a transmembrane protein which is involved in the release of a propeptide (PE) in the blood circulation. Spadin, a synthetic PE analog, is known to modulate the potassium TREK-1 channel activity. Since, this channel is expressed in pancreatic beta cells which secrete insulin, a hormone involved in blood glucose regulation, spadin may play a role in glucose homeostasis. Consistent with this hypothesis, spadin improves glucose tolerance in mice, by stimulating insulin release. Spadin is a natural peptide derived from sortilin, which is known to control the glucose transporter Glut4 trafficking to the plasma membrane of adipocytes. This suggests that spadin may regulate glucose storage in adipocytes by affecting the sortilin function. However, my results show that spadin has no effect on glucose storage. In summary, spadin is involved in insulin secretion and glucose homeostasis and may be an alternative treatment against obesity and diabetes; En France, l’obésité touche 15% de la population et est en perpétuelle augmentation. Elle est une cause majeure du diabète de type 2. Elle se traduit par un accroissement du nombre de cellules de stockage du gras (adipocytes) et une résistance périphérique à l’insuline. Chez des individus obèses, l’augmentation du nombre d’adipocytes est associée à une diminution de l’expression de la sortiline, protéine transmembranaire dont le clivage entraine la formation du propeptide (PE) et sa libération dans la circulation sanguine. L’analogue synthétique du PE, la spadine, est connu pour moduler l’activité du canal potassique TREK-1. Ce canal étant exprimé dans la cellule bêta pancréatique qui sécrète l’insuline, une hormone participant à la régulation du taux de glucose dans le sang, il est possible que la spadine joue un rôle dans l’homéostasie du glucose. Mes résultats confirment cette hypothèse. En effet, la spadine améliore la tolérance au glucose des souris, en favorisant la libération d’insuline. La spadine est également connue pour interagir avec sortiline, indispensable au trafic du transporteur de glucose Glut4 vers la membrane des adipocytes. Cette interaction spadine-sortiline suggère que la spadine pourrait moduler l’entrée du glucose dans les adipocytes via la sortiline. Mes résultats montrent que la spadine ne modifie pas les capacités de stockage du glucose des adipocytes. En conclusion, la spadine joue un rôle dans la sécrétion de l’insuline et dans la régulation de la glycémie, ce qui peut présenter un intérêt pharmaceutique
- Published
- 2016
150. State-independent intracellular access of quaternary ammonium blockers to the pore of TREK-1
- Author
-
Stephen J. Tucker, Matthias R. Schmidt, Markus Rapedius, Mark S.P. Sansom, Thomas Baukrowitz, Chetan Sharma, and Phillip J. Stansfeld
- Subjects
Models, Molecular ,K2P channel ,TREK-1 ,Protein Conformation ,Xenopus ,homology modeling ,Intracellular Space ,Biophysics ,TWIK-1 ,Gating ,Biochemistry ,Ion ,Potassium Channels, Tandem Pore Domain ,Protein structure ,Potassium Channel Blockers ,Animals ,Humans ,Binding site ,Ion channel ,Helix bundle ,Chemistry ,Potassium channel ,Article Addendum ,Quaternary Ammonium Compounds ,TRAAK ,Structural Homology, Protein ,Covalent bond ,ion channel ,docking ,potassium channel - Abstract
We previously reported that TREK-1 gating by internal pH and pressure occurs close to or within the selectivity filter. These conclusions were based upon kinetic measurements of high-affinity block by quaternary ammonium (QA) ions that appeared to exhibit state-independent accessibility to their binding site within the pore. Intriguingly, recent crystal structures of two related K2P potassium channels were also both found to be open at the helix bundle crossing. However, this did not exclude the possibility of gating at the bundle crossing and it was suggested that side-fenestrations within these structures might allow state-independent access of QA ions to their binding site. In this addendum to our original study we demonstrate that even hydrophobic QA ions do not access the TREK-1 pore via these fenestrations. Furthermore, by using a chemically reactive QA ion immobilized within the pore via covalent cysteine modification we provide additional evidence that the QA binding site remains accessible to the cytoplasm in the closed state. These results support models of K2P channel gating which occur close to or within the selectivity filter and do not involve closure at the helix bundle crossing.
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.