Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 72
Filtrer
1.
Sci Rep ; 11(1): 14632, 2021 07 16.
Article de Anglais | MEDLINE | ID: mdl-34272451

RÉSUMÉ

The voltage-dependent potassium channel Kv1.3 plays essential roles in the immune system, participating in leukocyte activation, proliferation and apoptosis. The regulatory subunit KCNE4 acts as an ancillary peptide of Kv1.3, modulates K+ currents and controls channel abundance at the cell surface. KCNE4-dependent regulation of the oligomeric complex fine-tunes the physiological role of Kv1.3. Thus, KCNE4 is crucial for Ca2+-dependent Kv1.3-related leukocyte functions. To better understand the role of KCNE4 in the regulation of the immune system, we manipulated its expression in various leukocyte cell lines. Jurkat T lymphocytes exhibit low KCNE4 levels, whereas CY15 dendritic cells, a model of professional antigen-presenting cells, robustly express KCNE4. When the cellular KCNE4 abundance was increased in T cells, the interaction between KCNE4 and Kv1.3 affected important T cell physiological features, such as channel rearrangement in the immunological synapse, cell growth, apoptosis and activation, as indicated by decreased IL-2 production. Conversely, ablation of KCNE4 in dendritic cells augmented proliferation. Furthermore, the LPS-dependent activation of CY15 cells, which induced Kv1.3 but not KCNE4, increased the Kv1.3-KCNE4 ratio and increased the expression of free Kv1.3 without KCNE4 interaction. Our results demonstrate that KCNE4 is a pivotal regulator of the Kv1.3 channelosome, which fine-tunes immune system physiology by modulating Kv1.3-associated leukocyte functions.


Sujet(s)
Canal potassique Kv1.3/physiologie , Leucocytes/physiologie , Canaux potassiques voltage-dépendants/physiologie , Animaux , Lignée cellulaire , Membrane cellulaire/métabolisme , Cellules dendritiques/métabolisme , Techniques de knock-down de gènes , Humains , Immunité , Synapses immunologiques/physiologie , Interleukine-2/métabolisme , Ouverture et fermeture des portes des canaux ioniques , Cellules Jurkat , Souris
2.
Neuropharmacology ; 185: 108399, 2021 03 01.
Article de Anglais | MEDLINE | ID: mdl-33400937

RÉSUMÉ

Midbrain dopamine neurons (DANs) regulate various brain functions such as motor control and motivation. Alteration of spiking activities of these neurons could contribute to severe brain disorders including Parkinson's disease and depression. Previous studies showed important roles of somatodendritic voltage-gated K+ channels (Kv) of DANs in governing neuronal excitability and dopamine release. However, it remains largely unclear about the biophysical properties and the function of Kv channels distributed at DAN axons. We performed whole-cell recordings from the axons of DANs in acute mouse midbrain and striatal slices. We detected both rapidly activating/inactivating Kv current (i.e. A-current) and rapidly activating but slowly inactivating current (i.e. D-current) in DAN axons. Pharmacological experiments with channel blockers revealed that these currents are predominantly mediated by Kv1.4 and Kv1.2 subunits, respectively. Blocking these currents could substantially prolong axonal action potentials (APs) via a reduction of their repolarization slope. Together, our results show that Kv channels mediating A- and D-currents shape AP waveforms in midbrain DAN axons, through this regulation they may control dopamine release at the axonal terminals. Therefore, these axonal Kv channels could be drug targets for brain disorders with abnormal dopamine release.


Sujet(s)
Potentiels d'action/physiologie , Axones/physiologie , Neurones dopaminergiques/physiologie , Canal potassique Kv1.3/physiologie , Canal potassique Kv1.4/physiologie , Mésencéphale/physiologie , Potentiels d'action/effets des médicaments et des substances chimiques , Animaux , Axones/effets des médicaments et des substances chimiques , Neurones dopaminergiques/effets des médicaments et des substances chimiques , Femelle , Protéines KChIP/antagonistes et inhibiteurs , Protéines KChIP/physiologie , Canal potassique Kv1.3/antagonistes et inhibiteurs , Canal potassique Kv1.4/antagonistes et inhibiteurs , Mâle , Mésencéphale/effets des médicaments et des substances chimiques , Souris , Souris transgéniques , Inhibiteurs des canaux potassiques/pharmacologie
3.
Cell Physiol Biochem ; 54(5): 842-852, 2020 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-32886870

RÉSUMÉ

BACKGROUND/AIMS: Kv1.3 channel is the only voltage-dependent potassium channel in plasma membrane of human lymphocytes. Bearing in mind a rather steep voltage-dependence of Kv1.3 activation and inactivation, its modulation by B and T cells activation and by co-culture with stromal bone-marrow cells was addressed. METHODS: Patch-clamp technique in the whole cell mode was applied to human resting and activated human B and T cells, in monoculture and co-culture with stromal OP9 cells. RESULTS: Polyclonal activation of B and T cells in monoculture caused Kv1.3 current in B cells to activate at more negative and in T cells at more positive potentials, whereas the inactivation of Kv1.3 current in resting T cells occurred at more negative voltages. Co-culture with OP9 cells abolished the shift of voltage dependence upon the polyclonal activation but fixed the substantial difference between B and T cells, resting or activated, with both activation and inactivation negatively shifted by 15 mV for T lymphocytes. However, activated B cells displayed an incomplete inactivation, which was augmented by the co-culture. Neither activation nor co-culture caused substantial changes in the Kv1.3 current density. CONCLUSION: The combination of activation and inactivation processes yields the fraction of steady-state Kv1.3 current (window current), which was higher in activated B cells, partly due to an incomplete inactivation. A relatively smaller window current in resting B cells and resting T cells in co-culture correlated with a more depolarized resting membrane potential. Rather than insignificant changes in the Kv1.3 channels functional expression, the modulation of their voltage dependence by activation and co-culture with bone-marrow stromal cells was essential for the control of membrane potential.


Sujet(s)
Lymphocytes B/métabolisme , Canal potassique Kv1.3/métabolisme , Lymphocytes T/métabolisme , Adulte , Moelle osseuse/métabolisme , Techniques de coculture , Femelle , Volontaires sains , Humains , Ouverture et fermeture des portes des canaux ioniques/physiologie , Canal potassique Kv1.3/physiologie , Activation des lymphocytes/physiologie , Lymphocytes/métabolisme , Mâle , Potentiels de membrane/physiologie , Techniques de patch-clamp , Cellules stromales/métabolisme
4.
Rev Neurol ; 70(2): 53-66, 2020 Jan 16.
Article de Espagnol | MEDLINE | ID: mdl-31930471

RÉSUMÉ

Nowadays, it is well accepted that obesity and metabolic syndrome are diseases that constitute a global public health issue. In consequence, the interest in the study of the effects these pathologies produce in the central nervous system has greatly increased in the last decades. One of the most overlooked topics in the literature is the impact they exert in sensory systems, among which is olfaction. The olfactory system is related to a number of vital functions, like the activation of defense mechanisms, contribution to appetitive and digestive reflexes, recognition of conspecifics, and even has socio-sexual implications. It has been discovered that the olfactory system also plays a crucial role in food intake, the choice of foods, appetite and satiety mechanisms; therefore, it is involved in obesity development. Clinical studies have proven that obese patients exhibit hyposmia more frequently than aged-matched healthy controls. Olfactory alterations have also been found in obese rodents or in animals with similar features of human metabolic syndrome. The causes of this association are still being investigated. This work reviews the studies that have tried to understand this association from a preclinical and clinical approach as well as those biological mechanisms that could be involved. The evidences here presented suggest that obesity and metabolic syndrome affect the adequate function of olfactory sensory system.


TITLE: Obesidad, síndrome metabólico y percepción olfativa.En la actualidad, la obesidad y el síndrome metabólico son enfermedades que representan un grave problema global de salud pública. A consecuencia de ello, en las últimas décadas ha aumentado el interés por estudiar los efectos de estas patologías sobre el funcionamiento del sistema nervioso central. Uno de los aspectos más ignorados en la bibliografía ha sido el impacto que tienen sobre los sistemas sensoriales, entre los que se encuentra el olfato. El sistema olfativo se relaciona con distintas funciones vitales, como activar mecanismos de defensa, contribuir a la inducción de reflejos apetitivos y digestivos, y reconocer individuos de su misma especie, e incluso tiene implicaciones sociosexuales. Se sabe que, además, desempeña un papel importante en la ingesta de alimentos, en la decisión de lo que se va a consumir, en los mecanismos de apetito y saciedad y, por ende, está involucrado en el desarrollo de obesidad. Estudios clínicos han demostrado que pacientes con obesidad presentan hiposmia con mayor frecuencia en comparación con sujetos delgados de la misma edad. También se han encontrado alteraciones en el olfato de roedores que presentan obesidad o rasgos similares a los del síndrome metabólico del humano. Las causas por las cuales existe esta asociación apenas están comenzando a investigarse; en este trabajo se revisan los estudios que han intentado entenderla desde un enfoque clínico y preclínico, así como los mecanismos biológicos que hasta el momento se han explorado en la bibliografía.


Sujet(s)
Syndrome métabolique X/complications , Syndrome métabolique X/physiopathologie , Obésité/complications , Obésité/physiopathologie , Troubles de l'olfaction/étiologie , Perception olfactive , Animaux , Système nerveux central/cytologie , Système nerveux central/physiopathologie , Modèles animaux de maladie humaine , Humains , Canal potassique Kv1.3/physiologie , Syndrome métabolique X/diagnostic , Syndrome métabolique X/épidémiologie , Obésité/diagnostic , Obésité/épidémiologie , Perception olfactive/physiologie
5.
Biochem Pharmacol ; 165: 214-220, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-30878554

RÉSUMÉ

Rheumatoid arthritis (RA) is a serious autoimmune disease that has severe impacts on both the wellbeing of patients and the economy of the health system. Similar to many autoimmune diseases, RA concurs with a long evolution, which eventually results in highly debilitating symptoms. Therapeutic treatments last for long periods during RA. However, their efficiency and side effects result in suboptimal conditions. Therefore, the need for specific, safer and nontoxic alternatives for the treatment of RA is essential. Kv1.3 is a voltage-gated potassium channel that has a crucial role in immune system response. The proliferation and activation of leukocytes are linked to differential expressions of this channel. The evidence is particularly relevant in the aggressive T effector memory (TEM) cells, which are the main actors in the development of autoimmune diseases. Blockage of Kv1.3 inhibits the reactivity of these cells. Furthermore, pharmacological inhibition of Kv1.3 ameliorates symptoms in animal models of autoimmune diseases, such as experimental autoimmune encephalomyelitis or induced psoriasis with no side effects. Kv1.3 is sensitive to several animal toxins and plant compounds, and several research groups have searched for new Kv1.3 blockers by improving these natural molecules. The research is mainly focused on enhancing the selectivity of the blockers, thereby reducing the potential for side effects on other related channel subunits. Higher selectivity means that treatments will potentially be less harmful. This leads to a lower discontinuation rate of the therapy than the current first-line treatment for RA. The molecular backgrounds of many autoimmune diseases implicate leukocyte Kv1.3 and suggests that a new medication for RA is feasible. Therapies could also be later repurposed to treat other immune system disorders.


Sujet(s)
Polyarthrite rhumatoïde/traitement médicamenteux , Canal potassique Kv1.3/antagonistes et inhibiteurs , Animaux , Polyarthrite rhumatoïde/immunologie , Humains , Canal potassique Kv1.3/physiologie , Lymphocytes T/immunologie
6.
Toxins (Basel) ; 11(2)2019 02 01.
Article de Anglais | MEDLINE | ID: mdl-30717088

RÉSUMÉ

It was recently discovered that Ssm Spooky Toxin (SsTx) with 53 residues serves as a key killer factor in red-headed centipede's venom arsenal, due to its potent blockage of the widely expressed KCNQ channels to simultaneously and efficiently disrupt cardiovascular, respiratory, muscular, and nervous systems, suggesting that SsTx is a basic compound for centipedes' defense and predation. Here, we show that SsTx also inhibits KV1.3 channel, which would amplify the broad-spectrum disruptive effect of blocking KV7 channels. Interestingly, residue R12 in SsTx extends into the selectivity filter to block KV7.4, however, residue K11 in SsTx replaces this ploy when toxin binds on KV1.3. Both SsTx and its mutant SsTx_R12A inhibit cytokines production in T cells without affecting the level of KV1.3 expression. The results further suggest that SsTx is a key molecule for defense and predation in the centipedes' venoms and it evolves efficient strategy to disturb multiple physiological targets.


Sujet(s)
Venins d'arthropode/pharmacologie , Canaux potassiques KNCQ/antagonistes et inhibiteurs , Canal potassique Kv1.3/antagonistes et inhibiteurs , Inhibiteurs des canaux potassiques/pharmacologie , Animaux , Arthropodes , Cellules CHO , Cricetulus , Cytokines/métabolisme , Cellules HEK293 , Humains , Canaux potassiques KNCQ/physiologie , Canal potassique Kv1.3/physiologie , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme
7.
Arthritis Res Ther ; 21(1): 6, 2019 01 07.
Article de Anglais | MEDLINE | ID: mdl-30612588

RÉSUMÉ

BACKGROUND: Fibroblast-like synoviocytes (FLS) and CCR7- effector memory T (TEM) cells are two of the major cell types implicated in the progression of rheumatoid arthritis (RA). In particular, FLS become highly invasive, whereas TEM cells proliferate and secrete proinflammatory cytokines, during RA. FLS and T cells may also interact and influence each other's phenotypes. Inhibition of the pathogenic phenotypes of both FLS and TEM cells can be accomplished by selectively blocking the predominant potassium channels that they upregulate during RA: KCa1.1 (BK, Slo1, MaxiK, KCNMA1) upregulated by FLS and Kv1.3 (KCNA3) upregulated by activated TEM cells. In this study, we investigated the roles of KCa1.1 and Kv1.3 in regulating the interactions between FLS and TEM cells and determined if combination therapies of KCa1.1- and Kv1.3-selective blockers are more efficacious than monotherapies in ameliorating disease in rat models of RA. METHODS: We used in vitro functional assays to assess the effects of selective KCa1.1 and Kv1.3 channel inhibitors on the interactions of FLS isolated from rats with collagen-induced arthritis (CIA) with syngeneic TEM cells. We also used flow cytometric analyses to determine the effects of KCa1.1 blockers on the expression of proteins used for antigen presentation on CIA-FLS. Finally, we used the CIA and pristane-induced arthritis models to determine the efficacy of combinatorial therapies of KCa1.1 and Kv1.3 blockers in reducing disease severity compared with monotherapies. RESULTS: We show that the interactions of FLS from rats with CIA and of rat TEM cells are regulated by KCa1.1 and Kv1.3. Inhibiting KCa1.1 on FLS reduces the ability of FLS to stimulate TEM cell proliferation and migration, and inhibiting Kv1.3 on TEM cells reduces TEM cells' ability to enhance FLS expression of KCa1.1 and major histocompatibility complex class II protein, as well as stimulates their invasion. Furthermore, we show that combination therapies of selective KCa1.1 and Kv1.3 blockers are more efficacious than monotherapies at reducing signs of disease in two rat models of RA. CONCLUSIONS: Our results demonstrate the importance of KCa1.1 and Kv1.3 in regulating FLS and TEM cells during RA, as well as the value of combined therapies targeting both of these cell types to treat RA.


Sujet(s)
Arthrite expérimentale/métabolisme , Fibroblastes/métabolisme , Canal potassique Kv1.3/physiologie , Sous-unités alpha des canaux potassiques calcium-dépendants de grande conductance/physiologie , Cellules synoviales/métabolisme , Lymphocytes T/métabolisme , Animaux , Arthrite expérimentale/imagerie diagnostique , Polyarthrite rhumatoïde/imagerie diagnostique , Polyarthrite rhumatoïde/métabolisme , Cellules cultivées , Femelle , Cellules HEK293 , Humains , Rats , Rats de lignée LEW
8.
Inflamm Res ; 67(10): 863-877, 2018 Oct.
Article de Anglais | MEDLINE | ID: mdl-30083976

RÉSUMÉ

OBJECTIVE: Kaliotoxin2 (KTX2) is a highly selective blocker of voltage-dependent potassium channels Kv1.3 containing 37 amino acid residues. It is purified from Androctonus australis scorpion venom. The binding of KTX2 to its targets is able to alter the neuronal excitability leading to neurological disorders, accompanied by an inflammatory response. In brain, activation of insulin receptor signaling pathway by insulin induces current suppression and concomitant tyrosine phosphorylation of Kv1.3 channel. The aim of this study is to evaluate the effect of insulin injected by i.c.v. route on the neuro-pathophysiological and systemic disorders induced by KTX2. MATERIALS AND METHODS: Tissue damage, inflammatory response and oxidative stress biomarkers were assessed in NMRI mice at 24 h after co-injection of KTX2 and insulin by intracerebroventricular route. RESULTS: Obtained results revealed that the central administration of insulin prevents cerebral cortex injury, brain edema and blood-brain barrier alteration induced by KTX2, these are accompanied by significant decrease of systemic disorders including serum cytokines, inflammatory and oxidative stress markers and tissue damage. CONCLUSION: These results indicate that insulin is able to reduce neuro-immunological effects and systemic disorders induced by KTX2. The neuroprotective effect of insulin may be due to its crucial role in the regulation of inflammation response and its properties to modulate the activity of Kv1.3 channels in brain.


Sujet(s)
Insuline/usage thérapeutique , Canal potassique Kv1.3/antagonistes et inhibiteurs , Neuroprotecteurs/usage thérapeutique , Syndromes neurotoxiques/traitement médicamenteux , Inhibiteurs des canaux potassiques/toxicité , Venins de scorpion/toxicité , Animaux , Encéphale/effets des médicaments et des substances chimiques , Encéphale/métabolisme , Cytokines/sang , Canal potassique Kv1.3/physiologie , Souris , Syndromes neurotoxiques/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques
9.
Microcirculation ; 24(4)2017 05.
Article de Anglais | MEDLINE | ID: mdl-28504408

RÉSUMÉ

The connection between metabolism and flow in the heart, metabolic dilation, is essential for cardiac function. We recently found redox-sensitive Kv1.5 channels play a role in coronary metabolic dilation; however, more than one ion channel likely plays a role in this process as animals null for these channels still showed limited coronary metabolic dilation. Accordingly, we examined the role of another Kv1 family channel, the energetically linked Kv1.3 channel, in coronary metabolic dilation. We measured myocardial blood flow (contrast echocardiography) during norepinephrine-induced increases in cardiac work (heart rate x mean arterial pressure) in WT, WT mice given correolide (preferential Kv1.3 antagonist), and Kv1.3-null mice (Kv1.3-/- ). We also measured relaxation of isolated small arteries mounted in a myograph. During increased cardiac work, myocardial blood flow was attenuated in Kv1.3-/- and in correolide-treated mice. In isolated vessels from Kv1.3-/- mice, relaxation to H2 O2 was impaired (vs WT), but responses to adenosine and acetylcholine were equivalent to WT. Correolide reduced dilation to adenosine and acetylcholine in WT and Kv1.3-/- , but had no effect on H2 O2 -dependent dilation in vessels from Kv1.3-/- mice. We conclude that Kv1.3 channels participate in the connection between myocardial blood flow and cardiac metabolism.


Sujet(s)
Circulation coronarienne , Canal potassique Kv1.3/physiologie , Myocarde/métabolisme , Animaux , Circulation coronarienne/effets des médicaments et des substances chimiques , Souris , Inhibiteurs des canaux potassiques/pharmacologie , Débit sanguin régional/effets des médicaments et des substances chimiques , Triterpènes/pharmacologie , Vasodilatation/effets des médicaments et des substances chimiques
10.
J Neurochem ; 140(3): 404-420, 2017 02.
Article de Anglais | MEDLINE | ID: mdl-27861889

RÉSUMÉ

Venom-derived ion channel inhibitors have strong channel selectivity, potency, and stability; however, tracking delivery to their target can be challenging. Herein, we utilized luminescent quantum dots (QDs) conjugated to margatoxin (MgTx) as a traceable vehicle to target a voltage-dependent potassium channel, Kv1.3, which has a select distribution and well-characterized role in immunity, glucose metabolism, and sensory ability. We screened both unconjugated (MgTx) and conjugated MgTx (QD-MgTx) for their ability to inhibit Shaker channels Kv1.1 to Kv1.7 using patch-clamp electrophysiology in HEK293 cells. Our data indicate that MgTx inhibits 79% of the outward current in Kv1.3-transfected cells and that the QD-MgTx conjugate is able to achieve a similar level of block, albeit a slightly reduced efficacy (66%) and at a slower time course (50% block by 10.9 ± 1.1 min, MgTx; vs. 15.3 ± 1.2 min, QD-MgTx). Like the unbound peptide, the QD-MgTx conjugate inhibits both Kv1.3 and Kv1.2 at a 1 nM concentration, whereas it does not inhibit other screened Shaker channels. We tested the ability of QD-MgTx to inhibit native Kv1.3 expressed in the mouse olfactory bulb (OB). In brain slices of the OB, the conjugate acted similarly to MgTx to inhibit Kv1.3, causing an increased action potential firing frequency attributed to decreased intraburst duration rather than interspike interval. Our data demonstrate a retention of known biophysical properties associated with block of the vestibule of Kv1.3 by QD-MgTx conjugate compared to that of MgTx, inferring QDs could provide a useful tool to deliver ion channel inhibitors to targeted tissues in vivo.


Sujet(s)
Canal potassique Kv1.3/antagonistes et inhibiteurs , Canal potassique Kv1.3/physiologie , Neurotoxines/pharmacologie , Boîtes quantiques/administration et posologie , Potentiels d'action/effets des médicaments et des substances chimiques , Potentiels d'action/physiologie , Animaux , Femelle , Cellules HEK293 , Humains , Mâle , Souris , Souris de lignée C57BL , Neurotoxines/métabolisme , Bulbe olfactif/effets des médicaments et des substances chimiques , Bulbe olfactif/physiologie , Boîtes quantiques/métabolisme , Venins de scorpion/métabolisme , Venins de scorpion/pharmacologie
11.
Toxins (Basel) ; 8(4): 115, 2016 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-27104568

RÉSUMÉ

The Kv1.3 channel-acting scorpion toxins usually adopt the conserved anti-parallel ß-sheet domain as the binding interface, but it remains challenging to discover some highly selective Kv1.3 channel-acting toxins. In this work, we investigated the pharmacological profile of the Kv1.3 channel-acting BmKTX-D33H, a structural analogue of the BmKTX scorpion toxin. Interestingly, BmKTX-D33H, with its conserved anti-parallel ß-sheet domain as a Kv1.3 channel-interacting interface, exhibited more than 1000-fold selectivity towards the Kv1.3 channel as compared to other K⁺ channels (including Kv1.1, Kv1.2, Kv1.7, Kv11.1, KCa2.2, KCa2.3, and KCa3.1). As expected, BmKTX-D33H was found to inhibit the cytokine production and proliferation of both Jurkat cells and human T cells in vitro. It also significantly improved the delayed-type hypersensitivity (DTH) responses, an autoreactive T cell-mediated inflammation in rats. Amino acid sequence alignment and structural analysis strongly suggest that the "evolutionary" Gly11 residue of BmKTX-D33H interacts with the turret domain of Kv1 channels; it appears to be a pivotal amino acid residue with regard to the selectivity of BmKTX-D33H towards the Kv1.3 channel (in comparison with the highly homologous scorpion toxins). Together, our data indicate that BmKTX-D33H is a Kv1.3 channel-specific blocker. Finally, the remarkable selectivity of BmKTX-D33H highlights the great potential of evolutionary-guided peptide drug design in future studies.


Sujet(s)
Facteurs immunologiques/pharmacologie , Canal potassique Kv1.3/physiologie , Inhibiteurs des canaux potassiques/pharmacologie , Venins de scorpion/pharmacologie , Séquence d'acides aminés , Animaux , Maladies auto-immunes , Antigènes CD3 , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Cytokines/métabolisme , Femelle , Cellules HEK293 , Humains , Hypersensibilité retardée/induit chimiquement , Hypersensibilité retardée/traitement médicamenteux , Facteurs immunologiques/composition chimique , Facteurs immunologiques/usage thérapeutique , Cellules Jurkat , Ovalbumine , Inhibiteurs des canaux potassiques/composition chimique , Inhibiteurs des canaux potassiques/usage thérapeutique , Rats de lignée LEW , Venins de scorpion/composition chimique , Venins de scorpion/usage thérapeutique , Scorpions , Alignement de séquences , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme
12.
Int Immunopharmacol ; 31: 266-71, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26795234

RÉSUMÉ

OBJECTIVE: To investigate the effects of Ca(2+) activated potassium channel KCa3.1 and voltage-gated potassium channel Kv1.3 of B lymphocyte on inflammatory monocytes chemotaxis and the potential mechanisms. MATERIALS AND METHODS: Thanswell test was used to detect the inflammatory monocyte (Ly-6C(hi)) chemotaxis caused by the B lymphocyte. Enzyme-linked immunosorbent assay (ELISA) was applied to detecting the C-C motif ligand 7 (CCL7) in cultured media. Cell counting kit-8 (CCK) was used to detect the proliferation of B lymphocytes after activation and blockage of both KCa3.1 and Kv1.3 channels. Western blot was used to detect the expression of phosphorylated extracellular signal-regulated kinase (P-ERK) of the B lymphocytes. RESULTS: When activated, B lymphocytes significantly proliferated. After application of KCa3.1 channel-specific inhibitor TRAM-34 and potent Kv1.3 channel inhibitor ShK, both B lymphocytes proliferation and Ly-6C(hi) monocyte chemotaxis were significantly inhibited. The expression of chemotaxis related factor CCL7 decreased remarkably. CONCLUSION: The opening of KCa3.1 and Kv1.3 channels promote B lymphocyte activation, proliferation and Ly-6C(hi) monocyte chemotaxis. The increase of CCL7 secretion by B lymphocyte may explain the pro migration effects.


Sujet(s)
Lymphocytes B/immunologie , Chimiotaxie/immunologie , Venins de cnidaires/pharmacologie , Canaux potassiques calcium-dépendants de conductance intermédiaire/physiologie , Canal potassique Kv1.3/physiologie , Monocytes/effets des médicaments et des substances chimiques , Pyrazoles/pharmacologie , Animaux , Lymphocytes B/effets des médicaments et des substances chimiques , Calcium/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Chimiokine CCL7/génétique , Chimiokine CCL7/métabolisme , Chimiotaxie/effets des médicaments et des substances chimiques , Canaux potassiques calcium-dépendants de conductance intermédiaire/antagonistes et inhibiteurs , Canal potassique Kv1.3/antagonistes et inhibiteurs , Activation des lymphocytes/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Monocytes/immunologie
13.
Nanotechnology ; 26(50): 505103, 2015 Dec 18.
Article de Anglais | MEDLINE | ID: mdl-26584910

RÉSUMÉ

Superparamagnetic iron oxide nanoparticles (SPIONs) are promising nanomaterials in medical practice due to their special magnetic characteristics and nanoscale size. However, their potential impacts on immune cells are not well documented. This study aims to investigate the effects of Fe2O3 nanoparticles (Fe2O3-NPs) on the electrophysiology of Kv1.3 channels in Jurkat T cells. Using the whole-cell patch-clamp technique, we demonstrate that incubation of Jurkat cells with Fe2O3-NPs dose- and time-dependently decreased the current density and shifted the steady-state inactivation curve and the recovery curve of Kv1.3 channels to a rightward direction. Fe2O3-NPs increased the NADP level but decreased the NADPH level of Jurkat cells. Direct induction of NADPH into the cytosole of Jurkat cells via the pipette abolished the rightward shift of the inactivation curve. In addition, transmission electron microscopy showed that Fe2O3-NPs could be endocytosed by Jurkat cells with relatively low speed and capacity. Fe2O3-NPs did not significantly affect the viability of Jurkat cells, but suppressed the expressions of certain cytokines (TNFα, IFNγ and IL-2) and interferon responsive genes (IRF-1 and PIM-1), and the time courses of Fe2O3-NPs endocytosis and effects on the expressions of cytokines and interferon responsive genes were compatible. We conclude that Fe2O3-NPs can be endocytosed by Jurkat cells and act intracellularly. Fe2O3-NPs decrease the current density and delay the inactivation and recovery kinetics of Kv1.3 channels in Jurkat cells by oxidizing NADPH and therefore disrupting the redox activity of the Kvß2 auxiliary subunit, and as a result, lead to changes of the Kv1.3 channel function. These results suggest that iron oxide nanoparticles may affect T cell function by disturbing the activity of Kv1.3 channels. Further, the suppressing effects of Fe2O3-NPs on the expressions of certain inflammatory cytokines and interferon responsive genes suggest that iron oxide nanoparticles may exert modulatory effects on T cell immune activities and anti-inflammation effects.


Sujet(s)
Composés du fer III/administration et posologie , Canal potassique Kv1.3/métabolisme , Nanoparticules de magnétite/administration et posologie , Oxydoréduction , Canaux potassiques voltage-dépendants/métabolisme , Cytokines/métabolisme , Endocytose , Composés du fer III/composition chimique , Humains , Interférons/génétique , Cellules Jurkat , Canal potassique Kv1.3/physiologie , Nanoparticules de magnétite/composition chimique , Canaux potassiques de la superfamille Shaker
14.
Sheng Li Xue Bao ; 67(5): 505-12, 2015 Oct 25.
Article de Chinois | MEDLINE | ID: mdl-26490068

RÉSUMÉ

This study was aimed to investigate the effects of blockade of Ca(2+) activated channel KCa3.1 and voltage-gated potassium channel Kv1.3 of the monocytes/macrophages on inflammatory monocyte chemotaxis. Chemotaxis assay was used to test the inflammatory Ly-6C(hi) monocyte chemotaxis caused by the monocytes/macrophages. The proliferation of monocytes/macrophages was detected by cell counting kit-8 (CCK8). Enzyme-linked immunosorbent assay (ELISA) was applied to detect the C-C motif ligand 7 (CCL7) in cultured media. The results showed that the recruitment of Ly-6C(hi) monocyte induced by monocytes/macrophages was suppressed by the potent Kv1.3 blocker Stichodactyla helianthus neurotoxin (ShK) or the specific KCa3.1 inhibitor TRAM-34. Meanwhile, the proliferation of monocytes/macrophages was significantly inhibited by ShK. The response of Ly-6C(hi) monocyte pretreated with ShK or TRAM-34 to CCL2 was declined. These results suggest that KCa3.1 and Kv1.3 may play an important role in monocytes/macrophages' proliferation and migration.


Sujet(s)
Canal potassique Kv1.3/physiologie , Macrophages/cytologie , Monocytes/cytologie , Canaux potassiques calcium-dépendants de petite conductance/physiologie , Mouvement cellulaire , Prolifération cellulaire , Venins de cnidaires/pharmacologie , Test ELISA , Humains , Canal potassique Kv1.3/antagonistes et inhibiteurs , Structure tertiaire des protéines , Pyrazoles/pharmacologie , Canaux potassiques calcium-dépendants de petite conductance/antagonistes et inhibiteurs
15.
Biochim Biophys Acta ; 1848(11 Pt A): 2805-12, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26299819

RÉSUMÉ

Chlorpromazine often causes severe and persistent thrombocytopenia. Several clinical studies have suggested the presence of an as-yet-unknown mechanism in this drug-induced thrombocytopenia, by which the platelet production from megakaryocytes may directly be affected. As we previously demonstrated in rat peritoneal mast cells or adipocytes, chlorpromazine is amphiphilic and preferentially partitioned into the lipid bilayers of the plasma membrane. Therefore, it can induce some structural changes in the megakaryocyte membrane surface and thus affect the process of thrombopoiesis. In the present study, employing the standard patch-clamp whole-cell recording technique, we examined the effects of chlorpromazine on the membrane capacitance and Kv1.3-channel currents in rat megakaryocytes. By electron microscopic imaging of the cellular surface, we also examined the effects of chlorpromazine on the membrane micro-architecture of megakaryocytes. Chlorpromazine markedly decreased the membrane capacitance of megakaryocytes, indicating the decreased number of invaginated plasma membranes, which was not detected by the fluorescent imaging techniques. As shown by electron microscopy, chlorpromazine actually changed the membrane micro-architecture of megakaryocytes, and was likely to halt the process of pro-platelet formation in the cells. This drug persistently decreased the membrane capacitance and almost totally and irreversibly inhibited the Kv1.3-channel currents in megakaryocytes. This study demonstrated for the first time that chlorpromazine is likely to inhibit the process of thrombopoiesis persistently in megakaryocytes, as detected by the long-lasting decrease in the membrane capacitance and the irreversible suppression of the Kv1.3-channel currents. Chlorpromazine-induced changes in the membrane micro-architecture are thought to be responsible for its persistent effects.


Sujet(s)
Membrane cellulaire/effets des médicaments et des substances chimiques , Chlorpromazine/pharmacologie , Mégacaryocytes/effets des médicaments et des substances chimiques , Thrombopoïèse/effets des médicaments et des substances chimiques , Animaux , Membrane cellulaire/métabolisme , Membrane cellulaire/physiologie , Cellules cultivées , Antagonistes de la dopamine/pharmacologie , Capacité électrique , Ouverture et fermeture des portes des canaux ioniques/effets des médicaments et des substances chimiques , Ouverture et fermeture des portes des canaux ioniques/physiologie , Canal potassique Kv1.3/métabolisme , Canal potassique Kv1.3/physiologie , Mâle , Mégacaryocytes/métabolisme , Mégacaryocytes/ultrastructure , Microscopie électronique , Techniques de patch-clamp , Rats , Rat Wistar
16.
FASEB J ; 29(10): 4324-33, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26148969

RÉSUMÉ

Defensins form a major family of antimicrobial peptides. Recently, human ß-defensin 2 and fungal plectasin were shown to be immune-related potassium voltage-gated channel subfamily A member 3 (Kv1.3) channel inhibitors. This work continued to show that the human α-defensins human neutrophil peptide (HNP) 1 and human defensin (HD) 5 are selective Kv1.3 channel inhibitors with 50% inhibition concentration values of 102.0 ± 30.3 nM and 2.2 ± 0.2 µM, respectively. Furthermore, HNP1 was found to inhibit Kv1.3 currents and IL-2 secretion in human CD3(+) T cells. Despite the structural similarity between HNP1 and HD5, HNP1 could simultaneously bind to the S1-S2 linker and the pore region of the Kv1.3 channel as both a toxinlike blocker and a novel modifier, whereas HD5 could only bind to the channel pore region as a toxinlike blocker. As a channel modifier, HNP1 could shift the conductance-voltage relationship curve of the Kv1.3 channel by ∼9.5 mV in the positive direction and could increase the time constant for channel activation through the electrostatic repulsion between the cationic HNP1 anchored in the S1-S2 linker and the positively charged S4 domain of the Kv1.3 channel. Together, these findings reveal that human α-defensins are novel endogenous inhibitors of Kv1.3 channels with distinct interaction mechanisms, which facilitates future research into their adaptive immune functions.


Sujet(s)
Ouverture et fermeture des portes des canaux ioniques/effets des médicaments et des substances chimiques , Canal potassique Kv1.3/physiologie , Défensines-alpha/pharmacologie , Immunité acquise/effets des médicaments et des substances chimiques , Immunité acquise/physiologie , Séquence d'acides aminés , Cellules cultivées , Cellules HEK293 , Humains , Cinétique , Canal potassique Kv1.3/génétique , Canal potassique Kv1.3/métabolisme , Potentiels de membrane/effets des médicaments et des substances chimiques , Données de séquences moléculaires , Techniques de patch-clamp , Liaison aux protéines , Similitude de séquences d'acides aminés , Défensines-alpha/génétique , Défensines-alpha/métabolisme
17.
Mediators Inflamm ; 2015: 436572, 2015.
Article de Anglais | MEDLINE | ID: mdl-25866450

RÉSUMÉ

Delayed rectifier K(+)-channels (Kv1.3) are predominantly expressed in T lymphocytes. Based on patch-clamp studies, the channels play crucial roles in facilitating the calcium influx necessary to trigger lymphocyte activation and proliferation. Using selective channel inhibitors in experimental animal models, in vivo studies then revealed the clinically relevant relationship between the channel expression and the pathogenesis of autoimmune diseases. In renal diseases, in which "chronic inflammation" or "the overstimulation of cellular immunity" is responsible for the pathogenesis, the overexpression of Kv1.3-channels in lymphocytes promotes their cellular proliferation and thus contributes to the progression of tubulointerstitial fibrosis. We recently demonstrated that benidipine, a potent dihydropyridine calcium channel blocker, which also strongly and persistently inhibits the lymphocyte Kv1.3-channel currents, suppressed the proliferation of kidney lymphocytes and actually ameliorated the progression of renal fibrosis. Based on the recent in vitro evidence that revealed the pharmacological properties of the channels, the most recent studies have revealed novel therapeutic implications of targeting the lymphocyte Kv1.3-channels for the treatment of renal diseases.


Sujet(s)
Maladies du rein/étiologie , Canal potassique Kv1.3/physiologie , Lymphocytes/physiologie , Animaux , Dihydropyridines/pharmacologie , Humains , Maladies du rein/traitement médicamenteux , Canal potassique Kv1.3/antagonistes et inhibiteurs , Activation des lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes/immunologie
18.
Med Hypotheses ; 85(1): 61-3, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25847395

RÉSUMÉ

In the gastrointestinal mucosal immune system, T-lymphocytes are activated to produce pro-inflammatory cytokines, and the over-activation of the lymphocytes is primarily responsible for the development of inflammatory bowel disease (IBD). Despite our understanding of the molecular involvement in the activation of lymphocytes, we know little about the physiological involvement. Circulating T-lymphocytes or those derived from thymus predominantly express delayed rectifier K(+)-channels (Kv1.3) in their plasma membranes and these channels play crucial roles in inducing the lymphocyte activation and proliferation. In the pathogenesis of chronic renal failure, these channels, which are overexpressed in proliferating lymphocytes within the kidneys, are responsible for the progression of the disease. Since the over-activation of cellular immunity is also involved in the pathogenesis of IBD, T-lymphocytes in the gastrointestinal mucosa could share the same stimulatory mechanisms with those in the inflamed kidneys. Therefore, we hypothesize here that T-lymphocytes in the gastrointestinal mucosa would also be stimulated by the activation of the Kv1.3-channels expressed in their plasma membranes, and that the overexpression of the channels would contribute to the development of IBD. Our hypothesis is unique because it sheds light for the first time on a physiological mechanism by which T-lymphocytes are activated in the gut mucosal immune system. It is also important because our idea could have novel therapeutic implications for IBD, in which the over-activation of the lymphocytes is responsible for the pathogenesis.


Sujet(s)
Maladies inflammatoires intestinales/physiopathologie , Muqueuse intestinale/physiopathologie , Canal potassique Kv1.3/physiologie , Lymphocytes/anatomopathologie , Humains
19.
Naunyn Schmiedebergs Arch Pharmacol ; 388(6): 653-61, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25743574

RÉSUMÉ

Glucocorticoids are hormones released in response to stress that are involved in various physiological processes including immune functions. One immune-modulating mechanism is achieved by the Kv1.3 voltage-dependent potassium channel, which is expressed highly in lymphocytes including effector memory T lymphocytes (TEM). Although glucocorticoids are known to inhibit Kv1.3 function, the detailed inhibitory mechanism is not yet fully understood. Here we studied the rapid non-genomic effects of cortisone and hydrocortisone on the human Kv1.3 channel expressed in Xenopus oocytes. Both cortisone and hydrocortisone reduced the amplitude of the Kv1.3 channel current in a concentration-dependent manner. Both cortisone and hydrocortisone rapidly and irreversibly inhibited Kv1.3 currents, eliminating the possibility of genomic regulation. Inhibition rate was stable relative to the degree of depolarization. Kinetically, cortisone altered the activating gate of Kv1.3 and hydrocortisone interacted with this channel in an open state. These results suggest that cortisone and hydrocortisone inhibit Kv1.3 currents via a non-genomic mechanism, providing a mechanism for the immunosuppressive effects of glucocorticoids.


Sujet(s)
Cortisone/pharmacologie , Hydrocortisone/pharmacologie , Canal potassique Kv1.3/physiologie , Inhibiteurs des canaux potassiques/pharmacologie , Animaux , Humains , Canal potassique Kv1.3/génétique , Potentiels de membrane/effets des médicaments et des substances chimiques , Ovocytes/effets des médicaments et des substances chimiques , Ovocytes/physiologie , Xenopus
20.
Dan Med J ; 61(11): B4946, 2014 Nov.
Article de Anglais | MEDLINE | ID: mdl-25370966

RÉSUMÉ

Ulcerative Colitis (UC) is a chronic inflammatory bowel disease located in the mucosa of the large bowel. UC often affects young adults between 15 and 40 years of age with no pre-dominant sex. Over time, incidence rates are steadily increasing and the cause of the disease remains unknown. Symptoms are general discomfort and bloody diarrhea. UC is diagnosed by endoscopic examination of the large bowel, where different hallmarks are found. It is of great importance that attacks/relapses are treated medically, as flares may cause death due to inflammatory destruction of the mucosa and perforation of the colon leading to extreme infection of the abdominal cavity. UC often affects the social life of the patients, as they feel that they must be in the immediate vicinity of toilets. Therefore, many patients prefer to stay at home during active disease. For society, UC is a costly disease due to patients reporting in sick and expensive medications. When medical treatment fails, UC patients must undergo surgery and have their colon removed (colectomy). This PhD project focused on the immune system of the body. Specifically, we looked into T cells (the chairmen of the immune system) that we believe play an important role in disease activity. When T cells are activated in inflammatory diseases, they produce several signaling substances (cytokines) that attract and activate the other parts of the immune system. T cells regulate their effector functions through calcium regulation. Upon activation, calcium is released from intracellular stores, which causes calcium channels to be embedded in the cell membrane (CRAC channels). As long as the T cells are stimulated, the two potassium channels KV1.3 and KCa3.1 maintain the driving force for calcium influx, thus keeping the T cells activated. Our aims were to investigate whether the two potassium channels KV1.3 and KCa3.1 were upregulated in mucosal biopsies from patients with active UC and whether there were correlations between the expression of the channels and the disease severity assessed by endoscopic and histological evaluation. Moreover, we used a rat colitis model (dextran sodium sulphate-induced) to examine the effect of pharmacological inhibition of KV1.3 and KCa3.1 on inflammation. We found that the expression of T cell potassium channel, KV1.3, was increased in active UC and a higher expression correlated well with both the endoscopic and the histological degree of inflammation. This suggests KV1.3 to be involved in the inflammatory process of UC. We did not find an increase of the other potassium channel, KCa3.1, at the gene expression level, but the channels were definitely present in the infiltrating T cells as examined by immunostaining. Preliminary gene expression data showed similar changes of gene expression in biopsies from Crohns disease (CD) patients. In addition, we conducted first pilot studies investigating whether pharmacological blockade of the channels ameliorates colitis in the rat DSS-model. We found a tendency towards less endoscopic inflammation in the acute phase (at day 7 and 10). However, at study termination, the improvement of inflammation failed to reach a significant level, presumably because of insufficient compound absorption from the intestine (based on low plasma concentration and previously reported amelioration of colitis by inhibiting KCa3.1). Based on these findings in our target identification study, it is suggested that both KV1.3 and KCa3.1 play a role in the inflammation of UC and possibly of CD and represent new pharmacological targets.


Sujet(s)
Rectocolite hémorragique/immunologie , Canaux potassiques calcium-dépendants de conductance intermédiaire/physiologie , Canal potassique Kv1.3/physiologie , Lymphocytes T/immunologie , Adulte , Animaux , Rectocolite hémorragique/anatomopathologie , Côlon/physiopathologie , Maladie de Crohn/immunologie , Maladie de Crohn/anatomopathologie , Cytokines/métabolisme , Humains , Canaux potassiques calcium-dépendants de conductance intermédiaire/métabolisme , Canal potassique Kv1.3/métabolisme , Rats
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE