Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 194
Filter
1.
Adv Pharmacol ; 77: 65-104, 2016.
Article in English | MEDLINE | ID: mdl-27451095

ABSTRACT

Endothelial calcium/calmodulin-gated K channels of small (KCa2.3) and intermediate conductance (KCa3.1) produce membrane hyperpolarization and endothelium-dependent hyperpolarization (EDH)-mediated vasodilation. Dysfunctions of the two channels and ensuing EDH impairments are found in several cardiovascular pathologies such as diabetes, atherosclerosis, postangioplastic neointima formation, but also inflammatory disease, cancer, and organ fibrosis. Moreover, KCa3.1 plays an important role in endothelial barrier dysfunction, edema formation in cardiac and pulmonary disease, and in ischemic stroke. Concerning KCa2.3, genome-wide association studies revealed an association of KCa2.3 channels with atrial fibrillation in humans. Accordingly, both channels are considered potential drug targets for cardio- and cerebrovascular disease states. In this chapter, we briefly review the function of the two channels in EDH-type vasodilation and systemic circulatory regulation and then highlight their pathophysiological roles in ischemic stroke as well as in pulmonary and brain edema. Finally, the authors summarize recent advances in the pharmacology of the channels and explore potential therapeutic utilities of novel channel modulators.


Subject(s)
Cardiovascular Diseases/drug therapy , Endothelium, Vascular/metabolism , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Genome-Wide Association Study , Humans , Vasodilation
2.
Cell Death Dis ; 7: e2174, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-27054329

ABSTRACT

Among the strategies adopted by glioma to successfully invade the brain parenchyma is turning the infiltrating microglia/macrophages (M/MΦ) into allies, by shifting them toward an anti-inflammatory, pro-tumor phenotype. Both glioma and infiltrating M/MΦ cells express the Ca(2+)-activated K(+) channel (KCa3.1), and the inhibition of KCa3.1 activity on glioma cells reduces tumor infiltration in the healthy brain parenchyma. We wondered whether KCa3.1 inhibition could prevent the acquisition of a pro-tumor phenotype by M/MΦ cells, thus contributing to reduce glioma development. With this aim, we studied microglia cultured in glioma-conditioned medium or treated with IL-4, as well as M/MΦ cells acutely isolated from glioma-bearing mice and from human glioma biopsies. Under these different conditions, M/MΦ were always polarized toward an anti-inflammatory state, and preventing KCa3.1 activation by 1-[(2-Chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34), we observed a switch toward a pro-inflammatory, antitumor phenotype. We identified FAK and PI3K/AKT as the molecular mechanisms involved in this phenotype switch, activated in sequence after KCa3.1. Anti-inflammatory M/MΦ have higher expression levels of KCa3.1 mRNA (kcnn4) that are reduced by KCa3.1 inhibition. In line with these findings, TRAM-34 treatment, in vivo, significantly reduced the size of tumors in glioma-bearing mice. Our data indicate that KCa3.1 channels are involved in the inhibitory effects exerted by the glioma microenvironment on infiltrating M/MΦ, suggesting a possible role as therapeutic targets in glioma.


Subject(s)
Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Macrophages/immunology , Microglia/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Chromones/pharmacology , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Humans , Interleukin-4/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/cytology , Morpholines/pharmacology , Phagocytosis/drug effects , Phenotype , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , RNA, Messenger/metabolism
3.
Internist (Berl) ; 55(4): 470-7, 2014 Apr.
Article in German | MEDLINE | ID: mdl-24577343

ABSTRACT

BACKGROUND: Integrated treatment pathways are an appropriate means for increasing the quality of treatment and outcome via process optimization. Taking the POLIKUM Health Centers as an example, we intend to demonstrate how the implementation can be effected for the indication of anemia. METHOD: The development and implementation were executed by an interdisciplinary workgroup in several workshops. In addition, the diagnoses and hemoglobin values of all patients with requests for hemograms were obtained and analyzed at two locations. RESULTS: Developing the pathway required significantly greater efforts than initially planned. The biggest challenge was to adequately map the complexity of the different forms of anemia and, concomitantly, to design a pathway that can actually be realized in everyday life. Moreover, evaluation of the patient data demonstrated that there are a large number of cases where existing anemias are not reflected in the respective diagnoses. CONCLUSION: While the ultimate effects of the new pathway cannot yet be assessed conclusively, it was possible to obtain valuable findings for practical use even at this point. Despite the limitations of the sample, the surprisingly high number of undetected anemias should give physicians cause for taking diagnostic measures even in patients with mild anemia.


Subject(s)
Anemia, Iron-Deficiency/therapy , Community Health Centers/organization & administration , Cooperative Behavior , Delivery of Health Care, Integrated/organization & administration , Health Plan Implementation/organization & administration , Interdisciplinary Communication , Internal Medicine/organization & administration , Algorithms , Anemia, Iron-Deficiency/diagnosis , Critical Pathways/organization & administration , Germany , Hemoglobinometry , Humans , Quality Improvement/organization & administration
4.
Cell Death Dis ; 4: e773, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23949222

ABSTRACT

Glioblastoma multiforme (GBM) is a diffuse brain tumor characterized by high infiltration in the brain parenchyma rendering the tumor difficult to eradicate by neurosurgery. Efforts to identify molecular targets involved in the invasive behavior of GBM suggested ion channel inhibition as a promising therapeutic approach. To determine if the Ca(2+)-dependent K(+) channel KCa3.1 could represent a key element for GBM brain infiltration, human GL-15 cells were xenografted into the brain of SCID mice that were then treated with the specific KCa3.1 blocker TRAM-34 (1-((2-chlorophenyl) (diphenyl)methyl)-1H-pyrazole). After 5 weeks of treatment, immunofluorescence analyses of cerebral slices revealed reduced tumor infiltration and astrogliosis surrounding the tumor, compared with untreated mice. Significant reduction of tumor infiltration was also observed in the brain of mice transplanted with KCa3.1-silenced GL-15 cells, indicating a direct effect of TRAM-34 on GBM-expressed KCa3.1 channels. As KCa3.1 channels are also expressed on microglia, we investigated the effects of TRAM-34 on microglia activation in GL-15 transplanted mice and found a reduction of CD68 staining in treated mice. Similar results were observed in vitro where TRAM-34 reduced both phagocytosis and chemotactic activity of primary microglia exposed to GBM-conditioned medium. Taken together, these results indicate that KCa3.1 activity has an important role in GBM invasiveness in vivo and that its inhibition directly affects glioma cell migration and reduces astrocytosis and microglia activation in response to tumor-released factors. KCa3.1 channel inhibition therefore constitutes a potential novel therapeutic approach to reduce GBM spreading into the surrounding tissue.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cell Movement/drug effects , Gene Silencing/drug effects , Humans , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasm Invasiveness , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Potassium Channel Blockers/pharmacology , Pyrazoles/pharmacology , RNA, Small Interfering/metabolism , Xenograft Model Antitumor Assays
5.
Br J Pharmacol ; 168(2): 432-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22891655

ABSTRACT

BACKGROUND AND PURPOSE: The K(Ca) 3.1 channel is a potential target for therapy of immune disease. We identified a compound from a new chemical class of K(Ca) 3.1 inhibitors and assessed in vitro and in vivo inhibition of immune responses. EXPERIMENTAL APPROACH: We characterized the benzothiazinone NS6180 (4-[[3-(trifluoromethyl)phenyl]methyl]-2H-1,4-benzothiazin-3(4H)-one) with respect to potency and molecular site of action on K(Ca) 3.1 channels, selectivity towards other targets, effects on T-cell activation as well as pharmacokinetics and inflammation control in colitis induced by 2,4-dinitrobenzene sulfonic acid, a rat model of inflammatory bowel disease (IBD). KEY RESULTS: NS6180 inhibited cloned human K(Ca) 3.1 channels (IC(50) = 9 nM) via T250 and V275, the same amino acid residues conferring sensitivity to triarylmethanes such as like TRAM-34. NS6180 inhibited endogenously expressed K(Ca) 3.1 channels in human, mouse and rat erythrocytes, with similar potencies (15-20 nM). NS6180 suppressed rat and mouse splenocyte proliferation at submicrolar concentrations and potently inhibited IL-2 and IFN-γ production, while exerting smaller effects on IL-4 and TNF-α and no effect on IL-17 production. Antibody staining showed K(Ca) 3.1 channels in healthy colon and strong up-regulation in association with infiltrating immune cells after induction of colitis. Despite poor plasma exposure, NS6180 (3 and 10 mg·kg(-1) b.i.d.) dampened colon inflammation and improved body weight gain as effectively as the standard IBD drug sulfasalazine (300 mg·kg(-1) q.d.). CONCLUSIONS AND IMPLICATIONS: NS6180 represents a novel class of K(Ca) 3.1 channel inhibitors which inhibited experimental colitis, suggesting K(Ca) 3.1 channels as targets for pharmacological control of intestinal inflammation.


Subject(s)
Inflammatory Bowel Diseases/drug therapy , Potassium Channel Blockers/therapeutic use , Thiazines/therapeutic use , Animals , Dinitrofluorobenzene/analogs & derivatives , Disease Models, Animal , Erythrocytes/drug effects , Erythrocytes/physiology , Humans , Inflammation/drug therapy , Inflammatory Bowel Diseases/immunology , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Lymphocyte Activation/drug effects , Male , Mice , Mice, Knockout , Potassium Channel Blockers/blood , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Thiazines/blood , Thiazines/pharmacology
6.
Acta Physiol (Oxf) ; 203(1): 117-26, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21362152

ABSTRACT

AIM: Endothelial membrane hyperpolarization mediated by KCa3.1 and KCa2.3 channels has been demonstrated to initiate endothelium-derived hyperpolarizing factor (EDHF)-type vasodilations. Moreover, pharmacological potentiation of KCa3.1/KCa2.3 channels has been suggested to improve EDHF-type vasodilations. Herein, we determined whether the KCa3.1/KCa2.3 activator SKA-31 and its derivative SKA-20 improve endothelial dysfunction in KCa3.1-/- and NOS3-/- mice. METHODS: Membrane potentials were measured using patch-clamp electrophysiology on carotid artery (CA) endothelial cells (CAEC) from wild-type (wt) and KCa3.1-/- mice. Endothelium-dependent vasodilations were determined by pressure myography in CA. RESULTS: SKA-31 (1 µm) activated KCa3.1 and KCa2.3 channels and induced membrane hyperpolarization in CAEC of wt (ΔMP -45 mV). These responses were significantly reduced in CAEC of KCa3.1-/- (ΔMP -8 mV). SKA-31 (200 nm, 500 nm) and SKA-20 (300 nm) significantly enhanced EDHF vasodilations in wt. SKA-20 also improved vasodilations during NO synthesis. In KCa3.1-/-, the defective EDHF vasodilations were unchanged at 200 nm SKA-31, but were significantly improved at 500 nm. EDHF vasodilations were slightly enhanced at 300 nm SKA-20, but vasodilations during NO synthesis were unchanged. SKA-31 (500 nm) enhanced the impaired endothelium-dependent vasodilation in NOS3-/- mice twofold. Pharmacological inhibition of the soluble epoxide hydrolase by t-AUCB (1 µm) in contrast did not increase ACh-induced EDHF- or NO-mediated vasodilations in wt and KCa3.1-/-. CONCLUSION: Normal and defective endothelium-dependent vasodilations in murine carotid arteries can be improved by pharmacological enhancement of KCa3.1/KCa2.3 functions. These findings further support the concept that pharmacological activation of endothelial KCa2.3/KCa3.1 could offer a novel endothelium-specific antihypertensive strategy.


Subject(s)
Benzothiazoles/pharmacology , Endothelium, Vascular/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/drug effects , Vasodilation/drug effects , Animals , Carotid Arteries/drug effects , Carotid Arteries/metabolism , Endothelium, Vascular/metabolism , Mice , Mice, Knockout , Patch-Clamp Techniques , Vasodilation/physiology
7.
Xenobiotica ; 41(3): 198-211, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21070145

ABSTRACT

1. PAP-1 (5-(4-phenoxybutoxy)psoralen), a potent small-molecule blocker of the voltage-gated potassium Kv1.3 channel, is currently in preclinical development for psoriasis. This study was undertaken to identify the major phase I metabolites of PAP-1 in Sprague-Dawley (SD) rats. 2. Five phase I metabolites, that is 5-(oxybutyric-acid)psoralen (M1), 5-[4-(4-hydroxybutoxy)]psoralen (M2), 5-[4-(4-hydroxyphenoxy)butoxy]psoralen (M3), 5-[4-(3-hydroxyphenoxy)butoxy]psoralen (M4), and 8-hydroxyl-5-(4-phenoxybutoxy)psoralen (M5), were isolated from the bile of rats and identified by mass spectrometry and NMR spectroscopy. The last four metabolites are new compounds. 3. Incubation of PAP-1 with SD rat liver microsomes rendered the same five major metabolites in a nicotinamide adenine dinucleotide phosphate (NADPH)-dependent manner suggesting that cytochrome P450 (CYP) enzymes are involved in PAP-1 metabolism. Inhibitors of rat CYP1A1/2 (alpha-naphthoflavone) and CYP3A (ketoconazole) but not CYP2D6 (quinidine), CYP2E (diethyldithiocarbamate), or CYP2C9 (sulphaphenazole) blocked the metabolism of PAP-1 in rat microsomes. 4. Of the five metabolites M3, M4, and M5 were found to inhibit Kv1.3 currents with nanomolar IC50s, while M1 and M2 were inactive. Our results identified the Kv1.3-inactive M1 as the major phase I metabolite, and suggest that hydroxylation and O-dealkylation are the major pathways of PAP-1 metabolism. 5. We further conducted a 6-month repeat-dose toxicity study with PAP-1 at 50 mg/kg in both male and female Lewis rats and did not observe any toxic effects.


Subject(s)
Ficusin/toxicity , Kv1.3 Potassium Channel/metabolism , Metabolic Detoxication, Phase I , Potassium Channel Blockers/toxicity , Animals , Bile/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Feces/chemistry , Ficusin/chemistry , Ficusin/metabolism , Male , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Pancreatitis-Associated Proteins , Potassium Channel Blockers/chemistry , Potassium Channel Blockers/metabolism , Rats , Rats, Sprague-Dawley , Toxicity Tests, Chronic
8.
Curr Med Chem ; 17(26): 2882-96, 2010.
Article in English | MEDLINE | ID: mdl-20858170

ABSTRACT

Recent results using animal models of inflammatory skin conditions have shown that blockers of the voltage-gated potassium channel, Kv1.3 hold great promise for clinical utility. Kv1.3 blockers act as immunosuppressants by modulating the various subsets of inflammatory T and B cells involved in autoimmune disorders. While peptidic inhibitors based on naturally occurring venoms demonstrate potent and selective Kv1.3 blockade, these require parenteral administration and may face potential immunogenicity problems. Small molecule blockers show considerable diversity, however selectivity over other Kv1-family channels has been difficult to achieve. More recent advances have added to the evidence that Kv1.3 channels are a suitable therapeutic target and that the development of novel and selective agents will herald new drugs for inflammatory skin disorders.


Subject(s)
Drug Delivery Systems , Kv1.3 Potassium Channel/antagonists & inhibitors , Potassium Channel Blockers/therapeutic use , Psoriasis/drug therapy , Humans , Kv1.3 Potassium Channel/chemistry , Potassium Channel Blockers/chemistry , Psoriasis/immunology , Psoriasis/physiopathology
9.
Transplant Proc ; 41(6): 2601-6, 2009.
Article in English | MEDLINE | ID: mdl-19715983

ABSTRACT

Currently, there is an unmet clinical need for novel immunosuppressive agents for long-term prevention of kidney transplant rejection as alternatives to the nephrotoxic calcineurin inhibitor cyclosporine (CsA). Recent studies have shown that K(+) channels have a crucial role in T-lymphocyte activity. We investigated whether combined blockade of the T-cell K(+) channels K(Ca)3.1 and K(v)1.3, both of which regulate calcium signaling during lymphocyte activation, is effective in prevention of rejection of kidney allografts from Fisher rats to Lewis rats. All recipients were initially treated with CsA (5 mg/kg d) for 7 days. In rats with intact allograft function, treatment was continued for 10 days with either CsA (5 mg/kg d), or a combination of TRAM-34 (K(Ca)3.1 inhibitor; 120 mg/kg d) plus Stichodactyla helianthus toxin (ShK, K(v)1.3 inhibitor; 80 microg/kg 3 times daily), or vehicle alone. Kidney sections were stained with periodic acid-Schiff or hematoxylin-eosin and histochemically for markers of macrophages (CD68), T-lymphocytes (CD43), or cytotoxic T-cells (CD8). Our results showed that treatment with TRAM-34 and ShK reduced total interstitial mononuclear cell infiltration (-42%) and the number of CD43+ T-cells (-32%), cytotoxic CD8+ T-cells (-32%), and CD68+ macrophages (-26%) in allografts when compared to vehicle treatment alone. Efficacy of TRAM-34/ShK treatment was comparable with that of CsA. In addition, no visible organ damage or other discernible adverse effects were observed with this treatment. Thus, selective blockade of T-lymphocyte K(Ca)3.1 and K(v)1.3 channels may represent a novel alternative therapy for prevention of kidney allograft rejection.


Subject(s)
Graft Rejection/prevention & control , Immunosuppression Therapy/methods , Intermediate-Conductance Calcium-Activated Potassium Channels/immunology , Kidney Transplantation/immunology , Kv1.1 Potassium Channel/immunology , T-Lymphocytes/immunology , Animals , Cnidarian Venoms/pharmacology , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Kv1.1 Potassium Channel/antagonists & inhibitors , Lymphocyte Culture Test, Mixed , Male , Pyrazoles/pharmacology , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Spleen/cytology , Spleen/drug effects , Spleen/immunology
10.
Arterioscler Thromb Vasc Biol ; 28(6): 1084-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18309114

ABSTRACT

OBJECTIVE: We previously demonstrated that upregulation of intermediate-conductance Ca(2+)-activated K(+) channels (K(Ca)3.1) is necessary for mitogen-induced phenotypic modulation in isolated porcine coronary smooth muscle cells (SMCs). The objective of the present study was to determine the role of K(Ca)3.1 in the regulation of coronary SMC phenotypic modulation in vivo using a swine model of postangioplasty restenosis. METHODS AND RESULTS: Balloon angioplasty was performed on coronary arteries of swine using either noncoated or balloons coated with the specific K(Ca)3.1 blocker TRAM-34. Expression of K(Ca)3.1, c-jun, c-fos, repressor element-1 silencing transcription factor (REST), smooth muscle myosin heavy chain (SMMHC), and myocardin was measured using qRT-PCR in isolated medial cells 2 hours and 2 days postangioplasty. K(Ca)3.1, c-jun, and c-fos mRNA levels were increased 2 hours postangioplasty, whereas REST expression decreased. SMMHC expression was unchanged at 2 hours, but decreased 2 days postangioplasty. Use of TRAM-34 coated balloons prevented K(Ca)3.1 upregulation and REST downregulation at 2 hours, SMMHC and myocardin downregulation at 2 days, and attenuated subsequent restenosis 14 and 28 days postangioplasty. Immunohistochemical analysis demonstrated corresponding changes at the protein level. CONCLUSIONS: Blockade of K(Ca)3.1 by delivery of TRAM-34 via balloon catheter prevented smooth muscle phenotypic modulation and limited subsequent restenosis.


Subject(s)
Coronary Stenosis/prevention & control , Coronary Vessels/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Potassium Channel Blockers/pharmacology , Pyrazoles/pharmacology , Angioplasty, Balloon, Coronary/adverse effects , Animals , Cell Differentiation/drug effects , Coronary Stenosis/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Disease Models, Animal , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Phenotype , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Swine , Transcription Factors/metabolism
11.
Exp Biol Med (Maywood) ; 232(10): 1338-54, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17959847

ABSTRACT

The small molecule 5-(4-phenoxybutoxy)psoralen (PAP-1) is a selective blocker of the voltage-gated potassium channel Kv1.3 that is highly expressed in cell membranes of activated effector memory T cells (TEMs). The blockade of Kv1.3 results in membrane depolarization and inhibition of TEM proliferation and function. In this study, the in vitro effects of PAP-1 on T cells and the in vivo toxicity and pharmacokinetics (PK) were examined in rhesus macaques (RM) with the ultimate aim of utilizing PAP-1 to define the role of TEMs in RM infected with simian immunodeficiency virus (SIV). Electrophysiologic studies on T cells in RM revealed a Kv1.3 expression pattern similar to that in human T cells. Thus, PAP-1 effectively suppressed TEM proliferation in RM. When administered intravenously, PAP-1 showed a half-life of 6.4 hrs; the volume of distribution suggested extensive distribution into extravascular compartments. When orally administered, PAP-1 was efficiently absorbed. Plasma concentrations in RM undergoing a 30-day, chronic dosing study indicated that PAP-1 levels suppressive to TEMs in vitro can be achieved and maintained in vivo at a non-toxic dose. PAP-1 selectively inhibited the TEM function in vivo, as indicated by a modest reactivation of cytomegalovirus (CMV) replication. Immunization of these chronically treated RM with the live influenza A/PR8 (flu) virus suggested that the development of an in vivo, flu-specific, central memory response was unaffected by PAP-1. These RM remained disease-free during the entire course of the PAP-1 study. Collectively, these data provide a rational basis for future studies with PAP-1 in SIV-infected RM.


Subject(s)
Ficusin/pharmacology , Kv1.3 Potassium Channel/physiology , T-Lymphocytes/immunology , Animals , Antibody Formation/drug effects , Electrophysiology , Immunity, Cellular/drug effects , Immunologic Memory , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/drug effects , Macaca mulatta , Pancreatitis-Associated Proteins , T-Lymphocytes/drug effects
12.
Anal Bioanal Chem ; 374(4): 720-3, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12397501

ABSTRACT

The oxidation of thin aluminium layers in a microwave plasma has been investigated to determine the kinetics of oxide growth. Thin Al-coatings were oxidized by means of a variety of gas mixtures, characterized by different partial pressures of oxygen, in microwave-induced plasmas of different power. To study the whole kinetic process the Al-metal and the oxide formed were investigated by means of a combination of grazing incidence X-ray reflectometry (GIXR) and grazing incidence X-ray diffractometry (GIXRD). XPS and FTIR spectroscopy confirmed the formation of stoichiometric Al(2)O(3). The alumina formed is X-ray amorphous. Quantitative description of oxide formation was achieved indirectly by determination of the decrease in the integrated intensity of the Al(111)-peak and the total thickness of the whole coating. These values enabled calculation of kinetic data. It was found that oxide growth was a combination of two simultaneous processes - diffusion and sputter processes. The diffusion coefficient D (cm(2) s(-1)) and the sputter rate S (nm s(-1)) were determined. The effect of the composition of the gas mixture, microwave power, and concentration of activated oxygen species on the oxidation process will be discussed. For calculation of the activation energy, E(A), of this plasma-enhanced diffusion process the temperature-dependence of D was investigated.

13.
Nervenarzt ; 73(9): 820-9, 2002 Sep.
Article in German | MEDLINE | ID: mdl-12215872

ABSTRACT

Regarding the high prevalence of traumatic experiences in patients with borderline personality disorders (BPD), we review the available literature focussing on the hypothesis that BPD is a subtype of trauma associated disorders. The criteria of BPD, of complex post-traumatic stress disorders (PTSD), and of disorders of extreme stress not otherwise specified (DESNOS) substantially overlap. Research of the long-term course of BPD and PTSD, trauma research, and research of vulnerability in both disorders yielded converging results. Neuropsychological deficits in BPD and PTSD as well as psychoendocrinological and neuroimaging studies in BPD und PTSD also revealed common features. A pathogenetic specificity of individual etiologic factors does not appear to exist, however the assumption of a diathesis-stress model with traumatisation as a necessary but etiologically insufficient condition seems justified. Further research will have to prove BPD as a complex and early-onset post-traumatic stress disorder after multiple and/or chronic (type II) traumatic experiences during childhood and/or youth. Definitive conclusions require further research efforts.


Subject(s)
Borderline Personality Disorder/diagnosis , Stress Disorders, Post-Traumatic/diagnosis , Adolescent , Adult , Arousal/physiology , Borderline Personality Disorder/physiopathology , Borderline Personality Disorder/psychology , Brain/pathology , Brain/physiopathology , Child , Humans , Life Change Events , Personality Development , Research , Risk Factors , Stress Disorders, Post-Traumatic/physiopathology , Stress Disorders, Post-Traumatic/psychology
14.
Proc Natl Acad Sci U S A ; 98(24): 13942-7, 2001 Nov 20.
Article in English | MEDLINE | ID: mdl-11717451

ABSTRACT

Adoptive transfer experimental autoimmune encephalomyelitis (AT-EAE), a disease resembling multiple sclerosis, is induced in rats by myelin basic protein (MBP)-activated CD4(+) T lymphocytes. By patch-clamp analysis, encephalitogenic rat T cells stimulated repeatedly in vitro expressed a unique channel phenotype ("chronically activated") with large numbers of Kv1.3 voltage-gated channels (approximately 1500 per cell) and small numbers of IKCa1 Ca(2+)-activated K(+) channels (approximately 50-120 per cell). In contrast, resting T cells displayed 0-10 Kv1.3 and 10-20 IKCa1 channels per cell ("quiescent" phenotype), whereas T cells stimulated once or twice expressed approximately 200 Kv1.3 and approximately 350 IKCa1 channels per cell ("acutely activated" phenotype). Consistent with their channel phenotype, [(3)H]thymidine incorporation by MBP-stimulated chronically activated T cells was suppressed by the peptide ShK, a blocker of Kv1.3 and IKCa1, and by an analog (ShK-Dap(22)) engineered to be highly specific for Kv1.3, but not by a selective IKCa1 blocker (TRAM-34). The combination of ShK-Dap(22) and TRAM-34 enhanced the suppression of MBP-stimulated T cell proliferation. Based on these in vitro results, we assessed the efficacy of K(+) channel blockers in AT-EAE. Specific and simultaneous blockade of the T cell channels by ShK or by a combination of ShK-Dap(22) plus TRAM-34 prevented lethal AT-EAE. Blockade of Kv1.3 alone with ShK-Dap(22), but not of IKCa1 with TRAM-34, was also effective. When administered after the onset of symptoms, ShK or the combination of ShK-Dap(22) plus TRAM-34 greatly ameliorated the clinical course of both moderate and severe AT-EAE. We conclude that selective targeting of Kv1.3, alone or with IKCa1, may provide an effective new mode of therapy for multiple sclerosis.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Multiple Sclerosis/prevention & control , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , Potassium Channels/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , Calcium Channel Blockers/administration & dosage , Calcium Channel Blockers/pharmacokinetics , Calcium Channel Blockers/pharmacology , Cells, Cultured , Cnidarian Venoms/administration & dosage , Cnidarian Venoms/pharmacokinetics , Cnidarian Venoms/pharmacology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Guinea Pigs , Intermediate-Conductance Calcium-Activated Potassium Channels , Isotope Labeling , Kv1.3 Potassium Channel , Multiple Sclerosis/metabolism , Phenotype , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/pharmacokinetics , Potassium Channel Blockers/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Rats , Rats, Inbred Lew , Thymidine/metabolism , Tritium/metabolism
15.
Gen Physiol Biophys ; 20(1): 83-95, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11508824

ABSTRACT

The effects of gallamine on ionic currents in single intact Ranvier nodes of the toad Xenopus were investigated. The following fully reversible effects were observed: 1. With a test concentration of 1 mmol/l the current-voltage relation of steady-state potassium currents, IK ss exhibited a complete block of IK ss up to about V = 110 mV; with stronger depolarisations the block was incomplete. The peak sodium currents, in contrast, were not affected. 2. At the same test concentration the potassium permeability constant PK was reduced by 92% from its normal value, while the sodium permeability constant PNa decreased by only 8%. 3. Concentration-response relations of the block of PK yielded an apparent dissociation constant of 30 micromol/l and a steepness parameter of unity. Patch-clamp experiments on cloned Kv1.1, Kv1.2, Kv1.3 and Kv3.1 channels yielded apparent dissociation constants of 86, 19, >>100 and 121 micromol/l, respectively. Our findings show that gallamine is particularly well suited for separating potassium and sodium currents in axonal current ensembles. They also strongly suggest that potassium currents in Ranvier nodes of Xenopus are mainly carried by an ensemble of Kv1.1 and 1.2 channels.


Subject(s)
Gallamine Triethiodide/pharmacology , Neuromuscular Nondepolarizing Agents/pharmacology , Potassium Channels, Voltage-Gated/metabolism , Ranvier's Nodes/drug effects , Animals , Dose-Response Relationship, Drug , Electrophysiology , Models, Chemical , Neurons/physiology , Patch-Clamp Techniques , Potassium/metabolism , Sodium/metabolism , Time Factors , Xenopus
16.
J Biol Chem ; 276(46): 43145-51, 2001 Nov 16.
Article in English | MEDLINE | ID: mdl-11527975

ABSTRACT

Apamin-sensitive small conductance calcium-activated potassium channels (SKCa1-3) mediate the slow afterhyperpolarization in neurons, but the molecular identity of the channel has not been defined because of the lack of specific inhibitors. Here we describe the structure-based design of a selective inhibitor of SKCa2. Leiurotoxin I (Lei) and PO5, peptide toxins that share the RXCQ motif, potently blocked human SKCa2 and SKCa3 but not SKCa1, whereas maurotoxin, Pi1, Tskappa, and PO1 were ineffective. Lei blocked these channels more potently than PO5 because of the presence of Ala(1), Phe(2), and Met(7). By replacing Met(7) in the RXCQ motif of Lei with the shorter, unnatural, positively charged diaminobutanoic acid (Dab), we generated Lei-Dab(7), a selective SKCa2 inhibitor (K(d) = 3.8 nm) that interacts with residues in the external vestibule of the channel. SKCa3 was rendered sensitive to Lei-Dab(7) by replacing His(521) with the corresponding SKCa2 residue (Asn(367)). Intracerebroventricular injection of Lei-Dab(7) into mice resulted in no gross central nervous system toxicity at concentrations that specifically blocked SKCa2 homotetramers. Lei-Dab(7) will be a useful tool to investigate the functional role of SKCa2 in mammalian tissues.


Subject(s)
Peptides/chemistry , Potassium Channel Blockers , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Potassium Channels/chemistry , Scorpion Venoms/pharmacology , Alanine/pharmacology , Amino Acid Sequence , Animals , Arginine/chemistry , COS Cells , Cell Line , Cloning, Molecular , Dose-Response Relationship, Drug , Electrophysiology , Humans , Kinetics , Methionine/pharmacology , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Neurotoxins/pharmacology , PC12 Cells , Peptides/pharmacology , Phenylalanine/pharmacology , Protein Binding , Rats , Scorpion Venoms/chemistry , Sequence Homology, Amino Acid , Small-Conductance Calcium-Activated Potassium Channels , Threonine/chemistry , Transfection , Valine/chemistry , gamma-Aminobutyric Acid/pharmacology
17.
J Clin Immunol ; 21(4): 235-52, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11506193

ABSTRACT

The discovery of a diverse and unique set of ion channels in T lymphocytes has led to a rapidly growing body of knowledge about their functional roles in the immune system. Here we review the biophysical and molecular characterization of K+, Ca2+, and Cl- channels in T lymphocytes. Potent and specific blockers, especially of K+ channels, have provided molecular tools to elucidate the involvement of voltage- and calcium-activated potassium channels in T-cell activation and cell-volume regulation. Their unique and differential expression makes lymphocyte K+ channels excellent pharmaceutical targets for modulating immune system function. This review surveys recent progress at the biophysical, molecular, and functional roles of the ion channels found in T lymphocytes.


Subject(s)
Ion Channels/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Biophysical Phenomena , Biophysics , Calcium Signaling , Electrophysiology , Female , Humans , Ion Channels/chemistry , Ion Channels/genetics , Ion Channels/physiology , Maternal-Fetal Exchange , Models, Biological , Molecular Sequence Data , Potassium Channels/genetics , Potassium Channels/immunology , Potassium Channels/physiology , Pregnancy , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/physiology
19.
J Biol Chem ; 276(34): 32040-5, 2001 Aug 24.
Article in English | MEDLINE | ID: mdl-11425865

ABSTRACT

Selective and potent triarylmethane blockers of the intermediate conductance calcium-activated potassium channel, IKCa1, have therapeutic use in sickle cell disease and secretory diarrhea and as immunosuppressants. Clotrimazole, a membrane-permeant triarylmethane, blocked IKCa1 with equal affinity when applied externally or internally, whereas a membrane-impermeant derivative TRAM-30 blocked the channel only when applied to the cytoplasmic side, indicating an internal drug-binding site. Introduction of the S5-P-S6 region of the triarylmethane-insensitive small conductance calcium-activated potassium channel SKCa3 into IKCa1 rendered the channel resistant to triarylmethanes. Replacement of Thr(250) or Val(275) in IKCa1 with the corresponding SKCa3 residues selectively abolished triarylmethane sensitivity without affecting the affinity of the channel for tetraethylammonium, charybdotoxin, and nifedipine. Introduction of these two residues into SKCa3 rendered the channel sensitive to triarylmethanes. In a molecular model of IKCa1, Thr(250) and Val(275) line a water-filled cavity just below the selectivity filter. Structure-activity studies suggest that the side chain methyl groups of Thr(250) and Val(275) may lock the triarylmethanes in place via hydrophobic interactions with the pi-electron clouds of the phenyl rings. The heterocyclic moiety may project into the selectivity filter and obstruct the ion-conducting pathway from the inside.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Clotrimazole/metabolism , Potassium Channels/metabolism , Pyrazoles/metabolism , Amino Acid Sequence , Binding Sites , Calcium Channels/chemistry , Cytoplasm/metabolism , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels , Models, Molecular , Molecular Sequence Data , Potassium Channels/chemistry , Protein Conformation , Pyrazoles/antagonists & inhibitors , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...