Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Type of study
Publication year range
1.
Biochem Pharmacol ; 80(6): 858-66, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20488163

ABSTRACT

Kv1.3 plays a crucial role in the activation and proliferation of T-lymphocytes and macrophages. While Kv1.3 is responsible for the voltage-dependent potassium current in T-cells, in macrophages this K(+) current is generated by the association of Kv1.3 and Kv1.5. Patients with autoimmune diseases show a high number of effector memory T cells that are characterized by a high expression of Kv1.3 and Kv1.3 antagonists ameliorate autoimmune disorders in vivo. Diclofenac is a non-steroidal anti-inflammatory drug (NSAID) used in patients who suffer from painful autoimmune diseases such as rheumatoid arthritis. In this study, we show that diclofenac impairs immune response via a mechanism that involves Kv1.3. While diclofenac inhibited Kv1.3 expression in activated macrophages and T-lymphocytes, Kv1.5 remained unaffected. Diclofenac also decreased iNOS levels in Raw 264.7 cells, impairing their activation in response to lipopolysaccharide (LPS). LPS-induced macrophage migration and IL-2 production in stimulated Jurkat T-cells were also blocked by pharmacological doses of diclofenac. These effects were mimicked by Margatoxin, a specific Kv1.3 inhibitor, and Charybdotoxin, which blocks both Kv1.3 and Ca(2+)-activated K(+) channels (K(Ca)3.1). Because Kv1.3 is a very good target for autoimmune therapies, the effects of diclofenac on Kv1.3 are of high pharmacological relevance.


Subject(s)
Diclofenac/pharmacology , Drug Delivery Systems/methods , Immunologic Factors/pharmacology , Kv1.3 Potassium Channel/metabolism , Leukocytes/immunology , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/pathology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Cell Migration Inhibition/drug effects , Cell Migration Inhibition/immunology , Cells, Cultured , Humans , Jurkat Cells , Kv1.3 Potassium Channel/antagonists & inhibitors , Leukocytes/drug effects , Macrophages/cytology , Macrophages/immunology , Macrophages/pathology , Mice
2.
J Gen Physiol ; 135(2): 135-47, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20100893

ABSTRACT

Voltage-dependent potassium (K(v)) channels play a pivotal role in the modulation of macrophage physiology. Macrophages are professional antigen-presenting cells and produce inflammatory and immunoactive substances that modulate the immune response. Blockage of K(v) channels by specific antagonists decreases macrophage cytokine production and inhibits proliferation. Numerous pharmacological agents exert their effects on specific target cells by modifying the activity of their plasma membrane ion channels. Investigation of the mechanisms involved in the regulation of potassium ion conduction is, therefore, essential to the understanding of potassium channel functions in the immune response to infection and inflammation. Here, we demonstrate that the biophysical properties of voltage-dependent K(+) currents are modified upon activation or immunosuppression in macrophages. This regulation is in accordance with changes in the molecular characteristics of the heterotetrameric K(v)1.3/K(v)1.5 channels, which generate the main K(v) in macrophages. An increase in K(+) current amplitude in lipopolysaccharide-activated macrophages is characterized by a faster C-type inactivation, a greater percentage of cumulative inactivation, and a more effective margatoxin (MgTx) inhibition than control cells. These biophysical parameters are related to an increase in K(v)1.3 subunits in the K(v)1.3/K(v)1.5 hybrid channel. In contrast, dexamethasone decreased the C-type inactivation, the cumulative inactivation, and the sensitivity to MgTx concomitantly with a decrease in K(v)1.3 expression. Neither of these treatments apparently altered the expression of K(v)1.5. Our results demonstrate that the immunomodulation of macrophages triggers molecular and biophysical consequences in K(v)1.3/K(v)1.5 hybrid channels by altering the subunit stoichiometry.


Subject(s)
Immunomodulation , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Macrophages/immunology , Animals , Cell Line , Dexamethasone/pharmacology , Humans , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.5 Potassium Channel/antagonists & inhibitors , Lipopolysaccharides/immunology , Macrophages/drug effects , Membrane Potentials , Mice , Rats , Scorpion Venoms/pharmacology
3.
J Cell Physiol ; 217(3): 667-73, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18668522

ABSTRACT

Surface expression of voltage-dependent K(+) channels (Kv) has a pivotal role in leukocyte physiology. Although little is known about the physiological role of lipid rafts, these microdomains concentrate signaling molecules and their ion channel substrates. Kv1.3 associates with Kv1.5 to form functional channels in macrophages. Different isoform stoichiometries lead to distinct heteromeric channels which may be further modulated by targeting the complex to different membrane surface microdomains. Kv1.3 targets to lipid rafts, whereas Kv1.5 localization is under debate. With this in mind, we wanted to study whether heterotetrameric Kv1.5-containing channels target to lipid rafts. While in transfected HEK-293 cells, homo- and heterotetrameric channels targeted to rafts, Kv1.5 did not target to rafts in macrophages. Therefore, Kv1.3/Kv1.5 hybrid channels are mostly concentrated in non-raft microdomains. However, LPS-induced activation, which increases the Kv1.3/Kv1.5 ratio and caveolin, targeted Kv1.5 back to lipid rafts. Moreover, Kv1.5 did not localize to low-buoyancy fractions in L6E9 skeletal myoblasts, which also coexpress both channels, heart membranes or cardiomyocyes. Coexpression of a Cav3(DGV)-mutant confined Kv1.5 to Cav3(DGV)-vesicles of HEK cells. Contrarily, coexpression of Kvbeta2.1 impaired the Kv1.5 targeting to raft microdomains in HEK cells. Our results indicate that Kv1.5 partnership interactions are underlying mechanisms governing channel targeting to lipid rafts.


Subject(s)
Kv1.5 Potassium Channel/metabolism , Membrane Microdomains/metabolism , Animals , Caveolin 3/metabolism , Cell Line , Cell Separation , Cholesterol/metabolism , Humans , Kv1.3 Potassium Channel/metabolism , Macrophages/metabolism , Mice , Muscle, Skeletal/cytology , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Protein Subunits/metabolism , Protein Transport , Rats , Rats, Wistar
4.
Biochem Biophys Res Commun ; 369(4): 1094-7, 2008 May 16.
Article in English | MEDLINE | ID: mdl-18331828

ABSTRACT

Voltage-dependent K(+) channels (Kv) are involved in myocyte proliferation and differentiation by triggering changes in membrane potential and regulating cell volume. Since Kv7 channels may participate in these events, the purpose of this study was to investigate whether skeletal muscle Kv7.1 and Kv7.5 were involved during proliferation and myogenesis. Here we report that, while myotube formation did not regulate Kv7 channels, Kv7.5 was up-regulated during cell cycle progression. Although, Kv7.1 mRNA also increased during the G(1)-phase, pharmacological evidence mainly involves Kv7.5 in myoblast growth. Our results indicate that the cell cycle-dependent expression of Kv7.5 is involved in skeletal muscle cell proliferation.


Subject(s)
Cell Cycle , Cell Differentiation , KCNQ Potassium Channels/metabolism , KCNQ1 Potassium Channel/metabolism , Muscle Development , Myoblasts, Skeletal/cytology , Animals , Cell Line , Cell Proliferation , KCNQ Potassium Channels/genetics , KCNQ1 Potassium Channel/genetics , Muscle Development/genetics , Myoblasts, Skeletal/metabolism , Rats
5.
Biochim Biophys Acta ; 1783(5): 728-36, 2008 May.
Article in English | MEDLINE | ID: mdl-18230363

ABSTRACT

Voltage-dependent K(+) channels (Kv) are involved in the proliferation of many types of cells, but the mechanisms by which their activity is related to cell growth remain unclear. Kv antagonists inhibit the proliferation of mammalian cells, which is of physiological relevance in skeletal muscle. Although myofibres are terminally differentiated, some resident myoblasts may re-enter the cell cycle and proliferate. Here we report that the expression of Kv1.5 is cell-cycle dependent during myoblast proliferation. In addition to Kv1.5 other Kv, such as Kv1.3, are also up-regulated. However, pharmacological evidence mainly implicates Kv1.5 in myoblast growth. Thus, the presence of S0100176, a Kv antagonist, but not margatoxin and dendrotoxin, led to cell cycle arrest during the G(1)-phase. The use of selective cell cycle blockers showed that Kv1.5 was transiently accumulated during the early G(1)-phase. Furthermore, while myoblasts treated with S0100176 expressed low levels of cyclin A and D(1), the expression of p21(cip-1) and p27(kip1), two cyclin-dependent kinase inhibitors, increased. Our results indicate that the cell cycle-dependent expression of Kv1.5 is involved in skeletal muscle cell proliferation.


Subject(s)
Cell Cycle , Kv1.5 Potassium Channel/metabolism , Myoblasts, Skeletal/metabolism , Animals , Cell Cycle/genetics , Cell Line , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Gene Expression , Kv1.3 Potassium Channel/genetics , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/genetics , Kv1.5 Potassium Channel/physiology , Myoblasts, Skeletal/cytology , Rats
6.
J Biol Chem ; 283(13): 8756-64, 2008 Mar 28.
Article in English | MEDLINE | ID: mdl-18218624

ABSTRACT

Kv1.3 activity is determined by raft association. In addition to Kv1.3, leukocytes also express Kv1.5, and both channels control physiological responses. Because the oligomeric composition may modify the channel targeting to the membrane, we investigated heterotetrameric Kv1.3/Kv1.5 channel traffic and targeting in HEK cells. Kv1.3 and Kv1.5 generate multiple heterotetramers with differential surface expression according to the subunit composition. FRET analysis and pharmacology confirm the presence of functional hybrid channels. Raft association was evaluated by cholesterol depletion, caveolae colocalization, and lateral diffusion at the cell surface. Immunoprecipitation showed that both Kv1.3 and heteromeric channels associate with caveolar raft domains. However, homomeric Kv1.3 channels showed higher association with caveolin traffic. Moreover, FRAP analysis revealed higher mobility for hybrid Kv1.3/Kv1.5 than Kv1.3 homotetramers, suggesting that heteromers target to distinct surface microdomains. Studies with lipopolysaccharide-activated macrophages further supported that different physiological mechanisms govern Kv1.3 and Kv1.5 targeting to rafts. Our results implicate the traffic and localization of Kv1.3/Kv1.5 heteromers in the complex regulation of immune system cells.


Subject(s)
Cell Membrane/metabolism , Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Animals , Cells, Cultured , Electrophysiology , Humans , Kv1.3 Potassium Channel/genetics , Kv1.5 Potassium Channel/genetics , Macrophages/metabolism , Mice , Patch-Clamp Techniques , Protein Binding , Protein Transport , Rats
7.
Cardiovasc Res ; 76(3): 430-41, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-17900547

ABSTRACT

OBJECTIVE: Cellular cardiomyoplasty using skeletal myoblasts is a promising therapy for myocardial infarct repair. Once transplanted, myoblasts grow, differentiate and adapt their electrophysiological properties towards more cardiac-like phenotypes. Voltage-dependent Na(+) channels (Na(v)) are the main proteins involved in the propagation of the cardiac action potential, and their phenotype affects cardiac performance. Therefore, we examined the expression of Na(v) during proliferation and differentiation in skeletal myocytes. METHODS AND RESULTS: We used the rat neonatal skeletal myocyte cell line L6E9. Proliferation of L6E9 cells induced Na(v)1.4 and Na(v)1.5, although neither protein has an apparent role in cell growth. During myogenesis, Na(v)1.5 was largely induced. Electrophysiological and pharmacological properties, as well as mRNA expression, indicate that cardiac-type Na(v)1.5 accounts for almost 90% of the Na(+) current in myotubes. Unlike in proliferation, this protein plays a pivotal role in myogenesis. The adoption of a cardiac-like phenotype is further supported by the increase in Na(v)1.5 colocalization in caveolae. Finally, we demonstrate that the treatment of myoblasts with neuregulin further increased Na(v)1.5 in skeletal myocytes. CONCLUSION: Our results indicate that skeletal myotubes adopt a cardiac-like phenotype in cell culture conditions and that the expression of Na(v)1.5 acts as an underlying molecular mechanism.


Subject(s)
Cardiomyoplasty/methods , Muscle Proteins/metabolism , Myoblasts, Skeletal/metabolism , Phenotype , Sodium Channels/metabolism , Action Potentials/physiology , Animals , Biopsy , Caveolae/metabolism , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Cells, Cultured , Humans , Muscle Development/physiology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Myocardial Infarction/therapy , NAV1.5 Voltage-Gated Sodium Channel , Neuregulin-1/pharmacology , Patch-Clamp Techniques , Rats
8.
Biochem Biophys Res Commun ; 352(4): 913-8, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17157812

ABSTRACT

Voltage-dependent K(+) (Kv) channels are involved in the immune response. Kv1.3 is highly expressed in activated macrophages and T-effector memory cells of autoimmune disease patients. Macrophages are actively involved in T-cell activation by cytokine production and antigen presentation. However, unlike T-cells, macrophages express Kv1.5, which is resistant to Kv1.3-drugs. We demonstrate that mononuclear phagocytes express different Kv1.3/Kv1.5 ratios, leading to biophysically and pharmacologically distinct channels. Therefore, Kv1.3-based treatments to alter physiological responses, such as proliferation and activation, are impaired by Kv1.5 expression. The presence of Kv1.5 in the macrophagic lineage should be taken into account when designing Kv1.3-based therapies.


Subject(s)
Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Macrophages/drug effects , Macrophages/metabolism , Animals , Biophysical Phenomena , Biophysics , Cells, Cultured , Mice , Scorpion Venoms/pharmacology
9.
Recent Pat Anticancer Drug Discov ; 2(3): 212-23, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18221064

ABSTRACT

Potassium channels constitute a large and heterogeneous family with more than eighty genes which encode membrane proteins that control membrane potential. In addition to nerve and cardiac action potential, these proteins are involved in a number of physiological processes including volume regulation, apoptosis, immunomodulation and differentiation. Many potassium channels have been related to proliferation and cell-cycle progression in mammalian cell lines and certain potassium channels show impaired expression in cancer cells and tumours. In addition, in some cases a correlation has been established between the protein expression levels and the grade of malignancy of the tumour. Many drugs have been found to inhibit both K+ channel activity and cell-cycle progression. Since potassium channels may play a pivotal role in tumour cell proliferation, these proteins should be taken into account when designing new cancer treatment strategies. The increasing list of recent patents, covered in this review, shows the relevance of this emergent subject.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism , Potassium Channels/drug effects , Animals , Cell Proliferation/drug effects , Drug Design , Humans , Neoplasms/pathology , Patents as Topic , Potassium Channel Blockers/pharmacology , Potassium Channels/physiology
10.
J Biol Chem ; 281(49): 37675-85, 2006 Dec 08.
Article in English | MEDLINE | ID: mdl-17038323

ABSTRACT

Voltage-dependent K(+) (Kv) currents in macrophages are mainly mediated by Kv1.3, but biophysical properties indicate that the channel composition could be different from that of T-lymphocytes. K(+) currents in mouse bone marrow-derived and Raw-264.7 macrophages are sensitive to Kv1.3 blockers, but unlike T-cells, macrophages express Kv1.5. Because Shaker subunits (Kv1) may form heterotetrameric complexes, we investigated whether Kv1.5 has a function in Kv currents in macrophages. Kv1.3 and Kv1.5 co-localize at the membrane, and half-activation voltages and pharmacology indicate that K(+) currents may be accounted for by various Kv complexes in macrophages. Co-expression of Kv1.3 and Kv1.5 in human embryonic kidney 293 cells showed that the presence of Kv1.5 leads to a positive shift in K(+) current half-activation voltages and that, like Kv1.3, Kv1.3/Kv1.5 heteromers are sensitive to r-margatoxin. In addition, both proteins co-immunoprecipitate and co-localize. Fluorescence resonance energy transfer studies further demonstrated that Kv1.5 and Kv1.3 form heterotetramers. Electrophysiological and pharmacological studies of different ratios of Kv1.3 and Kv1.5 co-expressed in Xenopus oocytes suggest that various hybrids might be responsible for K(+) currents in macrophages. Tumor necrosis factor-alpha-induced activation of macrophages increased Kv1.3 with no changes in Kv.1.5, which is consistent with a hyperpolarized shift in half-activation voltage and a lower IC(50) for margatoxin. Taken together, our results demonstrate that Kv1.5 co-associates with Kv1.3, generating functional heterotetramers in macrophages. Changes in the oligomeric composition of functional Kv channels would give rise to different biophysical and pharmacological properties, which could determine specific cellular responses.


Subject(s)
Kv1.3 Potassium Channel/metabolism , Kv1.5 Potassium Channel/metabolism , Macrophages/metabolism , Animals , Base Sequence , Cell Line , DNA Primers/genetics , Female , Humans , In Vitro Techniques , Kv1.3 Potassium Channel/chemistry , Kv1.3 Potassium Channel/genetics , Kv1.5 Potassium Channel/chemistry , Kv1.5 Potassium Channel/genetics , Macrophages/ultrastructure , Membrane Potentials , Mice , Mice, Inbred BALB C , Microscopy, Immunoelectron , Oocytes/metabolism , Protein Structure, Quaternary , Protein Subunits , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection , Xenopus laevis
11.
Cancer Detect Prev ; 30(4): 375-85, 2006.
Article in English | MEDLINE | ID: mdl-16971052

ABSTRACT

BACKGROUND: Potassium channels (KCh) are the most diverse and ubiquitous class of ion channels. KCh control membrane potential and contribute to nerve and cardiac action potentials and neurotransmitter release. KCh are also involved in insulin release, differentiation, activation, proliferation, apoptosis, and several other physiological functions. The aim of this review is to provide an updated overview of the KCh role during the cell growth. Their potential use as pharmacological targets in cancer therapies is also discussed. METHODS: We searched PubMed (up to 2005) and identified relevant articles. Reprints were mainly obtained by on line subscription. Additional sources were identified through cross-referencing and obtained from Library services. RESULTS: KCh are responsible for some neurological and cardiovascular diseases and for a new medical discipline, channelopathies. Their role in congenital deafness, multiple sclerosis, episodic ataxia, LQT syndrome and diabetes has been proven. Furthermore, a large body of information suggests that KCh play a role in the cell cycle progression, and it is now accepted that cells require KCh to proliferate. Thus, KCh expression has been studied in a number of tumours and cancer cells. CONCLUSIONS: Cancer is far from being considered a channelopathy. However, it seems appropriate to take into account the involvement of KCh in cancer progression and pathology when developing new strategies for cancer therapy.


Subject(s)
Neoplasms/therapy , Potassium Channels/metabolism , Animals , Cell Proliferation , Humans , Ion Channel Gating , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...