Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Physiol ; 598(9): 1741-1752, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31106399

RESUMO

The left-right organizer (LRO) in the mouse consists of pit cells within the depression, located at the end of the developing notochord, also known as the embryonic node and crown cells lining the outer periphery of the node. Cilia on pit cells are posteriorly tilted, rotate clockwise and generate leftward fluid flow. Primary cilia on crown cells are required to interpret the directionality of fluid movement and initiate flow-dependent gene transcription. Crown cells express PC1-L1 and PC2, which may form a heteromeric polycystin channel complex on primary cilia. It is still only poorly understood how fluid flow activates the ciliary polycystin complex. Besides polycystin channels voltage gated channels like HCN4 and KCNQ1 have been implicated in establishing asymmetry. How this electrical network of ion channels initiates left-sided signalling cascades and differential gene expression is currently only poorly defined.


Assuntos
Padronização Corporal , Cílios , Animais , Camundongos , Transdução de Sinais , Canais de Cátion TRPP/metabolismo
2.
Sci Rep ; 7(1): 3756, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28623364

RESUMO

The peptide HsTX1[R14A] is a potent and selective blocker of the voltage-gated potassium channel Kv1.3, which is a highly promising target for the treatment of autoimmune diseases and other conditions. In order to assess the biodistribution of this peptide, it was conjugated with NOTA and radiolabelled with copper-64. [64Cu]Cu-NOTA-HsTX1[R14A] was synthesised in high radiochemical purity and yield. The radiotracer was evaluated in vitro and in vivo. The biodistribution and PET studies after intravenous and subcutaneous injections showed similar patterns and kinetics. The hydrophilic peptide was rapidly distributed, showed low accumulation in most of the organs and tissues, and demonstrated high molecular stability in vitro and in vivo. The most prominent accumulation occurred in the epiphyseal plates of trabecular bones. The high stability and bioavailability, low normal-tissue uptake of [64Cu]Cu-NOTA-HsTX1[R14A], and accumulation in regions of up-regulated Kv channels both in vitro and in vivo demonstrate that HsTX1[R14A] represents a valuable lead for conditions treatable by blockade of the voltage-gated potassium channel Kv1.3. The pharmacokinetics shows that both intravenous and subcutaneous applications are viable routes for the delivery of this potent peptide.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Peptídeos , Bloqueadores dos Canais de Potássio , Administração Intravenosa , Animais , Linhagem Celular , Injeções Subcutâneas , Masculino , Camundongos , Peptídeos/farmacocinética , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/farmacocinética , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Wistar
3.
Clin Immunol ; 180: 45-57, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28389388

RESUMO

Effector memory T lymphocytes (TEM cells) that lack expression of CCR7 are major drivers of inflammation in a number of autoimmune diseases, including multiple sclerosis and rheumatoid arthritis. The Kv1.3 potassium channel is a key regulator of CCR7- TEM cell activation. Blocking Kv1.3 inhibits TEM cell activation and attenuates inflammation in autoimmunity, and as such, Kv1.3 has emerged as a promising target for the treatment of TEM cell-mediated autoimmune diseases. The scorpion venom-derived peptide HsTX1 and its analog HsTX1[R14A] are potent Kv1.3 blockers and HsTX1[R14A] is selective for Kv1.3 over closely-related Kv1 channels. PEGylation of HsTX1[R14A] to create a Kv1.3 blocker with a long circulating half-life reduced its affinity but not its selectivity for Kv1.3, dramatically reduced its adsorption to inert surfaces, and enhanced its circulating half-life in rats. PEG-HsTX1[R14A] is equipotent to HsTX1[R14A] in preferential inhibition of human and rat CCR7- TEM cell proliferation, leaving CCR7+ naïve and central memory T cells able to proliferate. It reduced inflammation in an active delayed-type hypersensitivity model and in the pristane-induced arthritis (PIA) model of rheumatoid arthritis (RA). Importantly, a single subcutaneous dose of PEG-HsTX1[R14A] reduced inflammation in PIA for a longer period of time than the non-PEGylated HsTX1[R14A]. Together, these data indicate that HsTX1[R14A] and PEG-HsTX1[R14A] are effective in a model of RA and are therefore potential therapeutics for TEM cell-mediated autoimmune diseases. PEG-HsTX1[R14A] has the additional advantages of reduced non-specific adsorption to inert surfaces and enhanced circulating half-life.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Peptídeos/farmacologia , Polietilenoglicóis/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Venenos de Escorpião/farmacologia , Adulto , Alérgenos/imunologia , Animais , Artrite Experimental/induzido quimicamente , Artrite Experimental/patologia , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/patologia , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Hipersensibilidade Tardia/imunologia , Imunomodulação/efeitos dos fármacos , Leucócitos Mononucleares , Camundongos , Pessoa de Meia-Idade , Ovalbumina/imunologia , Peptídeos/química , Peptídeos/farmacocinética , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacocinética , Ratos , Ratos Endogâmicos Lew , Venenos de Escorpião/química , Venenos de Escorpião/farmacocinética , Baço/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Terpenos , Adulto Jovem
4.
Methods Mol Biol ; 1579: 231-244, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28299740

RESUMO

Matrix metalloproteinases (MMPs) represent more than 20 zinc-containing endopeptidases that cleave internal peptide bonds, leading to protein degradation. They play a critical role in many physiological cell functions, including tissue remodeling, embryogenesis, and angiogenesis. They are also involved in the pathogenesis of a vast array of diseases, including but not limited to systemic inflammation, various cancers, and cardiovascular, neurological, and autoimmune diseases. Here, we describe gel zymography to detect MMPs in cell and tissue samples and in cell culture supernatants.


Assuntos
Eletroforese em Gel de Poliacrilamida/métodos , Metaloproteinases da Matriz/análise , Animais , Células Cultivadas , Humanos
5.
Cell Death Dis ; 7(10): e2426, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27763639

RESUMO

Myoblasts are mononucleated precursors of myofibers; they persist in mature skeletal muscles for growth and regeneration post injury. During myotonic dystrophy type 1 (DM1), a complex autosomal-dominant neuromuscular disease, the differentiation of skeletal myoblasts into functional myotubes is impaired, resulting in muscle wasting and weakness. The mechanisms leading to this altered differentiation are not fully understood. Here, we demonstrate that the calcium- and voltage-dependent potassium channel, KCa1.1 (BK, Slo1, KCNMA1), regulates myoblast proliferation, migration, and fusion. We also show a loss of plasma membrane expression of the pore-forming α subunit of KCa1.1 in DM1 myoblasts. Inhibiting the function of KCa1.1 in healthy myoblasts induced an increase in cytosolic calcium levels and altered nuclear factor kappa B (NFκB) levels without affecting cell survival. In these normal cells, KCa1.1 block resulted in enhanced proliferation and decreased matrix metalloproteinase secretion, migration, and myotube fusion, phenotypes all observed in DM1 myoblasts and associated with disease pathogenesis. In contrast, introducing functional KCa1.1 α-subunits into DM1 myoblasts normalized their proliferation and rescued expression of the late myogenic marker Mef2. Our results identify KCa1.1 channels as crucial regulators of skeletal myogenesis and suggest these channels as novel therapeutic targets in DM1.


Assuntos
Diferenciação Celular , Movimento Celular , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Mioblastos Esqueléticos/citologia , Mioblastos Esqueléticos/metabolismo , Cálcio/metabolismo , Fusão Celular , Membrana Celular/metabolismo , Proliferação de Células , Células Cultivadas , Humanos , Espaço Intracelular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Distrofia Miotônica/patologia , NF-kappa B/metabolismo
6.
Sci Rep ; 6: 33808, 2016 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-27654170

RESUMO

Autoimmune diseases mediated by a type of white blood cell-T lymphocytes-are currently treated using mainly broad-spectrum immunosuppressants that can lead to adverse side effects. Antioxidants represent an alternative approach for therapy of autoimmune disorders; however, dietary antioxidants are insufficient to play this role. Antioxidant carbon nanoparticles scavenge reactive oxygen species (ROS) with higher efficacy than dietary and endogenous antioxidants. Furthermore, the affinity of carbon nanoparticles for specific cell types represents an emerging tactic for cell-targeted therapy. Here, we report that nontoxic poly(ethylene glycol)-functionalized hydrophilic carbon clusters (PEG-HCCs), known scavengers of the ROS superoxide (O2•-) and hydroxyl radical, are preferentially internalized by T lymphocytes over other splenic immune cells. We use this selectivity to inhibit T cell activation without affecting major functions of macrophages, antigen-presenting cells that are crucial for T cell activation. We also demonstrate the in vivo effectiveness of PEG-HCCs in reducing T lymphocyte-mediated inflammation in delayed-type hypersensitivity and in experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Our results suggest the preferential targeting of PEG-HCCs to T lymphocytes as a novel approach for T lymphocyte immunomodulation in autoimmune diseases without affecting other immune cells.

8.
Arthritis Res Ther ; 18(1): 103, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27165430

RESUMO

BACKGROUND: Fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA-FLS) contribute to joint inflammation and damage characteristic of the disease. RA-FLS express KCa1.1 (BK, Slo1, MaxiK, KCNMA1) as their major plasma membrane potassium channel. Blocking KCa1.1 reduces the invasive phenotype of RA-FLS and attenuates disease severity in animal models of RA. This channel has therefore emerged as a promising therapeutic target in RA. However, the pore-forming α subunit of KCa1.1 is widely distributed in the body, and blocking it induces severe side effects, thus limiting its value as a therapeutic target. On the other hand, KCa1.1 channels can also contain different accessory subunits with restricted tissue distribution that regulate channel kinetics and pharmacology. Identification of the regulatory subunits of KCa1.1 expressed by RA-FLS may therefore provide the opportunity for generating a selective target for RA treatment. METHODS: Highly invasive RA-FLS were isolated from patients with RA, and FLS from patients with osteoarthritis (OA) were used as minimally invasive controls. The ß subunit expression by FLS was assessed by quantitative reverse transcription polymerase chain reactions, Western blotting, and patch-clamp electrophysiology combined with pharmacological agents. FLS were sorted by flow cytometry on the basis of their CD44 expression level for comparison of their invasiveness and with their expression of KCa1.1 α and ß subunits. ß1 and ß3 subunit expression was reduced with small interfering RNA (siRNA) to assess their specific role in KCa1.1α expression and function and in FLS invasiveness. RESULTS: We identified functional ß1 and ß3b regulatory subunits in RA-FLS. KCa1.1 ß3b subunits were expressed by 70 % of the cells and were associated with highly invasive CD44(high) RA-FLS, whereas minimally invasive CD44(low) RA-FLS and OA-FLS expressed either ß1 subunit. Furthermore, we found that silencing the ß3 but not the ß1 subunit with siRNA reduced KCa1.1 channel density at the plasma membrane of RA-FLS and inhibited RA-FLS invasiveness. CONCLUSIONS: Our findings suggest the KCa1.1 channel composed of α and ß3b subunits as an attractive target for the therapy of RA.


Assuntos
Artrite Reumatoide/metabolismo , Fibroblastos/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/biossíntese , Sinoviócitos/metabolismo , Adulto , Idoso , Artrite Reumatoide/patologia , Western Blotting , Movimento Celular/fisiologia , Feminino , Fibroblastos/patologia , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Sinoviócitos/patologia
9.
Sci Rep ; 5: 15776, 2015 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-26508475

RESUMO

Botulinum neurotoxins (BoNTs) are the most toxic substances known. BoNT intoxicates cells in a highly programmed fashion initiated by binding to the cell surface, internalization and enzymatic cleavage of substrate, thus, inhibiting synaptic exocytosis. Over the past two decades, immunological significance of BoNT/A C-terminal heavy chain (HC) and light chain (LC) domains were investigated extensively leading to important findings. In the current work, we explored the significance of BoNT/A heavy chain N-terminal (HN) region as a vaccine candidate. Mice were immunized with recombinant HN519-845 generating antibodies (Abs) that were found to be protective against lethal dose of BoNT/A. Immuno-dominant regions of HN519-845 were identified and individually investigated for antibody response along with synthetic peptides within those regions, using in vivo protection assays against BoNT/A. Results were confirmed by patch-clamp analysis where anti-HN antibodies were studied for the ability to block toxin-induced channel formation. This data strongly indicated that HN519-593 is an important region in generating protective antibodies and should be valuable in a vaccine design. These results are the first to describe and dissect the protective activity of the BoNT/A HN domain.


Assuntos
Anticorpos Antibacterianos/imunologia , Formação de Anticorpos/imunologia , Toxinas Botulínicas Tipo A/imunologia , Neurotoxinas/imunologia , Toxinas Biológicas/imunologia , Sequência de Aminoácidos/fisiologia , Animais , Imunização/métodos , Camundongos , Fragmentos de Peptídeos/imunologia , Peptídeos/imunologia , Ligação Proteica/imunologia , Vacinação/métodos
10.
Mar Drugs ; 13(1): 529-42, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25603346

RESUMO

ShK, from the sea anemone Stichodactyla helianthus, is a 35-residue disulfide-rich peptide that blocks the voltage-gated potassium channel Kv1.3 at ca. 10 pM and the related channel Kv1.1 at ca. 16 pM. We developed an analog of this peptide, ShK-186, which is currently in Phase 1b-2a clinical trials for the treatment of autoimmune diseases such as multiple sclerosis and rheumatoid arthritis. While ShK-186 displays a >100-fold improvement in selectivity for Kv1.3 over Kv1.1 compared with ShK, there is considerable interest in developing peptides with an even greater selectivity ratio. In this report, we describe several variants of ShK that incorporate p-phophono-phenylalanine at the N-terminus coupled with internal substitutions at Gln16 and Met21. In addition, we also explored the combinatorial effects of these internal substitutions with an alanine extension at the C-terminus. Their selectivity was determined by patch-clamp electrophysiology on Kv1.3 and Kv1.1 channels stably expressed in mouse fibroblasts. The peptides with an alanine extension blocked Kv1.3 at low pM concentrations and exhibited up to 2250-fold selectivity for Kv1.3 over Kv1.1. Analogs that incorporates p-phosphono-phenylalanine at the N-terminus blocked Kv1.3 with IC50s in the low pM range and did not affect Kv1.1 at concentrations up to 100 nM, displaying a selectivity enhancement of >10,000-fold for Kv1.3 over Kv1.1. Other potentially important Kv channels such as Kv1.4 and Kv1.6 were only partially blocked at 100 nM concentrations of each of the ShK analogs.


Assuntos
Canal de Potássio Kv1.3/antagonistas & inibidores , Peptídeos/farmacologia , Anêmonas-do-Mar/química , Sequência de Aminoácidos , Animais , Cromatografia Líquida de Alta Pressão , Cromatografia de Fase Reversa , Relação Dose-Resposta a Droga , Concentração Inibidora 50 , Canal de Potássio Kv1.1/antagonistas & inibidores , Dados de Sequência Molecular , Técnicas de Patch-Clamp , Peptídeos/genética , Peptídeos/isolamento & purificação , Anêmonas-do-Mar/genética
11.
Arthritis Rheumatol ; 67(1): 96-106, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25252152

RESUMO

OBJECTIVE: Fibroblast-like synoviocytes (FLS) participate in joint inflammation and damage in rheumatoid arthritis (RA) and its animal models. The purpose of this study was to define the importance of KCa1.1 (BK, Maxi-K, Slo1, KCNMA1) channel expression and function in FLS and to establish these channels as potential new targets for RA therapy. METHODS: We compared KCa1.1 expression levels in FLS from rats with pristane-induced arthritis (PIA) and in FLS from healthy rats. We then used ex vivo functional assays combined with small interfering RNA-induced knockdown, overexpression, and functional modulation of KCa1.1 in PIA FLS. Finally, we determined the effectiveness of modulating KCa1.1 in 2 rat models of RA, moderate PIA and severe collagen-induced arthritis (CIA). RESULTS: We found that PIA FLS expressed the KCa1.1 channel as their major potassium channel, as has been found in FLS from patients with RA. In contrast, FLS from healthy rats expressed fewer of these channels. Inhibiting the function or expression of KCa1.1 ex vivo reduced proliferation and invasive properties of, as well as protease production by, PIA FLS, whereas opening native KCa1.1 or overexpressing the channel enhanced the invasiveness of both FLS from rats with PIA and FLS from healthy rats. Treatment with a KCa1.1 channel blocker at the onset of clinical signs stopped disease progression in the PIA and CIA models, reduced joint and bone damage, and inhibited FLS invasiveness and proliferation. CONCLUSION: Our results demonstrate a critical role of KCa1.1 channels in the regulation of FLS invasiveness and suggest that KCa1.1 channels represent potential therapeutic targets in RA.


Assuntos
Artrite Reumatoide/patologia , Artrite Reumatoide/prevenção & controle , Movimento Celular/fisiologia , Fibroblastos/patologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/antagonistas & inibidores , Membrana Sinovial/patologia , Animais , Artrite Experimental/patologia , Artrite Experimental/fisiopatologia , Artrite Reumatoide/induzido quimicamente , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Feminino , Fibroblastos/fisiologia , Indóis/farmacologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/efeitos dos fármacos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Membrana Sinovial/fisiopatologia , Terpenos/efeitos adversos
12.
Int Immunopharmacol ; 22(2): 427-43, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25027630

RESUMO

The Big Potassium (BK) ion channel is commonly known by a variety of names (Maxi-K, KCNMA1, slo, stretch-activated potassium channel, KCa1.1). Each name reflects a different physical property displayed by this single ion channel. This transmembrane channel is found on nearly every cell type of the body and has its own distinctive roles for that tissue type. The BKα channel contains the pore that releases potassium ions from intracellular stores. This ion channel is found on the cell membrane, endoplasmic reticulum, Golgi and mitochondria. Complex splicing pathways produce different isoforms. The BKα channels can be phosphorylated, palmitoylated and myristylated. BK is composed of a homo-tetramer that interacts with ß and γ chains. These accessory proteins provide a further modulating effect on the functions of BKα channels. BK channels play important roles in cell division and migration. In this review, we will focus on the biology of the BK channel, especially its role, and its immune response towards cancer. Recent proteomic studies have linked BK channels with various proteins. Some of these interactions offer further insight into the role that BK channels have with cancers, especially with brain tumors. This review shows that BK channels have a complex interplay with intracellular components of cancer cells and still have plenty of secrets to be discovered.


Assuntos
Imunoterapia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Neoplasias/terapia , Animais , Vacinas Anticâncer , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Alta/química , Neoplasias/metabolismo , Isoformas de Proteínas , Subunidades Proteicas
13.
Am J Transl Res ; 6(3): 188-205, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936214

RESUMO

Big Potassium (BK) ion channels have several splice variants. One splice variant initially described within human glioma cells is called the glioma BK channel (gBK). Using a gBK-specific antibody, we detected gBK within three human small cell lung cancer (SCLC) lines. Electrophysiology revealed that functional membrane channels were found on the SCLC cells. Prolonged exposure to BK channel activators caused the SCLC cells to swell within 20 minutes and resulted in their death within five hours. Transduction of BK-negative HEK cells with gBK produced functional gBK channels. Quantitative RT-PCR analysis using primers specific for gBK, but not with a lung-specific marker, Sox11, confirmed that advanced, late-stage human SCLC tissues strongly expressed gBK mRNA. Normal human lung tissue and early, lower stage SCLC resected tissues very weakly expressed this transcript. Immunofluorescence using the anti-gBK antibody confirmed that SCLC cells taken at the time of the autopsy intensely displayed this protein. gBK may represent a late-stage marker for SCLC. HLA-A*0201 restricted human CTL were generated in vitro using gBK peptide pulsed dendritic cells. The exposure of SCLC cells to interferon-γ (IFN-γ) increased the expression of HLA; these treated cells were killed by the CTL better than non-IFN-γ treated cells even though the IFN-γ treated SCLC cells displayed diminished gBK protein expression. Prolonged incubation with recombinant IFN-γ slowed the in vitro growth and prevented transmigration of the SCLC cells, suggesting IFN-γ might inhibit tumor growth in vivo. Immunotherapy targeting gBK might impede advancement to the terminal stage of SCLC via two pathways.

14.
PLoS One ; 8(11): e78712, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24244345

RESUMO

The voltage-gated potassium channel Kv1.3 is a well-established target for treatment of autoimmune diseases. ShK peptide from a sea anemone is one of the most potent blockers of Kv1.3 but its application as a therapeutic agent for autoimmune diseases is limited by its lack of selectivity against other Kv channels, in particular Kv1.1. Accurate models of Kv1.x-ShK complexes suggest that specific charge mutations on ShK could considerably enhance its specificity for Kv1.3. Here we evaluate the K18A mutation on ShK, and calculate the change in binding free energy associated with this mutation using the path-independent free energy perturbation and thermodynamic integration methods, with a novel implementation that avoids convergence problems. To check the accuracy of the results, the binding free energy differences were also determined from path-dependent potential of mean force calculations. The two methods yield consistent results for the K18A mutation in ShK and predict a 2 kcal/mol gain in Kv1.3/Kv1.1 selectivity free energy relative to wild-type peptide. Functional assays confirm the predicted selectivity gain for ShK[K18A] and suggest that it will be a valuable lead in the development of therapeutics for autoimmune diseases.


Assuntos
Simulação por Computador , Canal de Potássio Kv1.3 , Modelos Moleculares , Peptídeos , Bloqueadores dos Canais de Potássio , Substituição de Aminoácidos , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/genética , Doenças Autoimunes/metabolismo , Linhagem Celular , Feminino , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/química , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Camundongos , Mutação de Sentido Incorreto , Peptídeos/química , Peptídeos/farmacologia , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Endogâmicos Lew , Termodinâmica
15.
PLoS One ; 8(10): e76740, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146918

RESUMO

Natural killer (NK) cells are large granular lymphocytes that participate in both innate and adaptive immune responses against tumors and pathogens. They are also involved in other conditions, including organ rejection, graft-versus-host disease, recurrent spontaneous abortions, and autoimmune diseases such as multiple sclerosis. We demonstrate that human NK cells express the potassium channels Kv1.3 and KCa3.1. Expression of these channels does not vary with expression levels of maturation markers but varies between adherent and non-adherent NK cell subpopulations. Upon activation by mitogens or tumor cells, adherent NK (A-NK) cells preferentially up-regulate KCa3.1 and non-adherent (NA-NK) cells preferentially up-regulate Kv1.3. Consistent with this different phenotype, A-NK and NA-NK do not display the same sensitivity to the selective KCa3.1 blockers TRAM-34 and NS6180 and to the selective Kv1.3 blockers ShK-186 and PAP-1 in functional assays. Kv1.3 block inhibits the proliferation and degranulation of NA-NK cells with minimal effects on A-NK cells. In contrast, blocking KCa3.1 increases the degranulation and cytotoxicity of A-NK cells, but not of NA-NK cells. TRAM-34, however, does not affect their ability to form conjugates with target tumor cells, to migrate, or to express chemokine receptors. TRAM-34 and NS6180 also increase the proliferation of both A-NK and NA-NK cells. This results in a TRAM-34-induced increased ability of A-NK cells to reduce in vivo tumor growth. Taken together, our results suggest that targeting KCa3.1 on NK cells with selective blockers may be beneficial in cancer immunotherapy.


Assuntos
Citotoxicidade Imunológica , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Células Matadoras Naturais/imunologia , Animais , Receptor 1 de Quimiocina CX3C , Adesão Celular/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Interleucina-12/farmacologia , Interleucina-15/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Células K562 , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/fisiologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Canal de Potássio Kv1.3/metabolismo , Camundongos , Proteínas Associadas a Pancreatite , Fenótipo , Bloqueadores dos Canais de Potássio/farmacologia , Pirazóis/metabolismo , Receptores de Quimiocinas/metabolismo , Proteínas Recombinantes/farmacologia , Regulação para Cima/efeitos dos fármacos
16.
FEBS Lett ; 586(22): 3996-4001, 2012 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-23063513

RESUMO

ShK, a 35-residue peptide from a sea anemone, is a potent blocker of potassium channels. Here we describe a new ShK analogue with an additional C-terminus Lys residue and amide. ShK-K-amide is a potent blocker of Kv1.3 and, in contrast to ShK and ShK-amide, is selective for Kv1.3. To understand this selectivity, we created complexes of ShK-K-amide with Kv1.3 and Kv1.1 using docking and molecular dynamics simulations, then performed umbrella sampling simulations to construct the potential of mean force of the ligand and calculate the corresponding binding free energy for the most stable configuration. The results agree well with experimental data.


Assuntos
Venenos de Cnidários/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Canal de Potássio Kv1.3/fisiologia , Bloqueadores dos Canais de Potássio/farmacologia , Algoritmos , Amidas/química , Animais , Sítios de Ligação , Ligação Competitiva , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Venenos de Cnidários/química , Venenos de Cnidários/metabolismo , Relação Dose-Resposta a Droga , Humanos , Ativação do Canal Iônico/genética , Ativação do Canal Iônico/fisiologia , Canal de Potássio Kv1.1/genética , Canal de Potássio Kv1.1/metabolismo , Canal de Potássio Kv1.1/fisiologia , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/metabolismo , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos
17.
Toxicon ; 60(5): 840-50, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22659540

RESUMO

The polypeptide toxin ShK is a potent blocker of Kv1.3 potassium channels, which play a crucial role in the activation of human effector memory T-cells (T(EM)). Selective blockers constitute valuable therapeutic leads for the treatment of autoimmune diseases mediated by T(EM) cells, such as multiple sclerosis, rheumatoid arthritis, and type-1 diabetes. We have established a recombinant peptide expression system in order to generate isotopically-labelled ShK and various ShK analogues for in-depth biophysical and pharmacological studies. ShK was expressed as a thioredoxin fusion protein in Escherichia coli BL21 (DE3) cells and purified initially by Ni²âº iminodiacetic acid affinity chromatography. The fusion protein was cleaved with enterokinase and purified to homogeneity by reverse-phase HPLC. NMR spectra of ¹5N-labelled ShK were similar to those reported previously for the unlabelled synthetic peptide, confirming that recombinant ShK was correctly folded. Recombinant ShK blocked Kv1.3 channels with a K(d) of 25 pM and inhibited the proliferation of human and rat T lymphocytes with a preference for T(EM) cells, with similar potency to synthetic ShK in all assays. This expression system also enables the efficient production of ¹5N-labelled ShK for NMR studies of peptide dynamics and of the interaction of ShK with Kv1.3 channels.


Assuntos
Venenos de Cnidários/metabolismo , Modelos Moleculares , Bloqueadores dos Canais de Potássio/metabolismo , Conformação Proteica , Proteínas Recombinantes de Fusão/metabolismo , Tiorredoxinas/metabolismo , Cromatografia de Afinidade , Cromatografia Líquida de Alta Pressão , Venenos de Cnidários/química , Venenos de Cnidários/farmacologia , Primers do DNA/genética , Escherichia coli , Marcação por Isótopo , Isótopos de Nitrogênio/metabolismo , Ressonância Magnética Nuclear Biomolecular , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Dobramento de Proteína , Proteínas Recombinantes de Fusão/farmacologia , Linfócitos T/efeitos dos fármacos , Tiorredoxinas/farmacologia
18.
Mol Pharmacol ; 82(3): 372-82, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22622363

RESUMO

Blockade of Kv1.3 K(+) channels in T cells is a promising therapeutic approach for the treatment of autoimmune diseases such as multiple sclerosis and type 1 diabetes mellitus. Vm24 (α-KTx 23.1) is a novel 36-residue Kv1.3-specific peptide isolated from the venom of the scorpion Vaejovis mexicanus smithi. Vm24 inhibits Kv1.3 channels of human lymphocytes with high affinity (K(d) = 2.9 pM) and exhibits >1500-fold selectivity over other ion channels assayed. It inhibits the proliferation and Ca(2+) signaling of human T cells in vitro and reduces delayed-type hypersensitivity reactions in rats in vivo. Our results indicate that Vm24 has exceptional pharmacological properties that make it an excellent candidate for treatment of certain autoimmune diseases.


Assuntos
Imunossupressores/farmacologia , Canal de Potássio Kv1.3/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Linfócitos T/efeitos dos fármacos , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/metabolismo , Células COS , Sinalização do Cálcio/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Chlorocebus aethiops , Feminino , Células HEK293 , Humanos , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Canal de Potássio Kv1.3/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Peptídeos/farmacologia , Ratos , Ratos Endogâmicos Lew , Venenos de Escorpião/metabolismo , Escorpiões/metabolismo , Linfócitos T/metabolismo
19.
PMC Biophys ; 3(1): 14, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-21054864

RESUMO

The various lamellar phases of dipalmitoylphosphadtidylcholine bilayers with and without cholesterol were used to assess the versatility of the fluorescent probe merocyanine 540 through simultaneous measurements of emission intensity, spectral shape, and steady-state anisotropy. Induction of the crystalline phase (Lc') by pre-incubation at 4°C produced a wavelength dependence of anisotropy which was strong at 15 and 25°C, weak at 38°C, and minimal above the main transition (>~41.5°C) or after returning the temperature from 46 to 25°C. The profile of anisotropy values across this temperature range revealed the ability of the probe to detect crystalline, gel (Lß'), and liquid crystalline (Lα) phases. The temperature dependence of fluorescence intensity was additionally able to distinguish between the ripple (Pß') and gel phases. In contrast, the shape of the emission spectrum, quantified as the ratio of merocyanine monomer and dimer peaks (585 and 621 nm), was primarily sensitive to the crystalline and gel phases because dimer fluorescence requires a highly-ordered environment. This requirement also explained the diminution of anisotropy wavelength dependence above 25°C. Repetition of experiments with vesicles containing cholesterol allowed creation of a phase map. Superimposition of data from the three simultaneous measurements provided details about the various phase regions in the map not discernible from any one of the three alone. The results were applied to assessment of calcium-induced membrane changes in living cells.PACS Codes: 87.16.dt.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...