Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Psychiatry ; 26(10): 5516-5531, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34400772

RESUMO

Amyloidogenic processing of the amyloid precursor protein (APP) forms the amyloid-ß peptide (Aß) component of pathognomonic extracellular plaques of AD. Additional early cortical changes in AD include neuroinflammation and elevated iron levels. Activation of the innate immune system in the brain is a neuroprotective response to infection; however, persistent neuroinflammation is linked to AD neuropathology by uncertain mechanisms. Non-parametric machine learning analysis on transcriptomic data from a large neuropathologically characterised patient cohort revealed the acute phase protein lactoferrin (Lf) as the key predictor of amyloid pathology. In vitro studies showed that an interaction between APP and the iron-bound form of Lf secreted from activated microglia diverted neuronal APP endocytosis from the canonical clathrin-dependent pathway to one requiring ADP ribosylation factor 6 trafficking. By rerouting APP recycling to the Rab11-positive compartment for amyloidogenic processing, Lf dramatically increased neuronal Aß production. Lf emerges as a novel pharmacological target for AD that not only modulates APP processing but provides a link between Aß production, neuroinflammation and iron dysregulation.


Assuntos
Doença de Alzheimer , Lactoferrina , Proteínas de Fase Aguda , Doença de Alzheimer/genética , Secretases da Proteína Precursora do Amiloide/metabolismo , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Humanos
2.
J Cell Sci ; 129(10): 2016-29, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27068538

RESUMO

Cell migration is a fundamental feature of tumour metastasis and angiogenesis. It is regulated by a variety of signalling molecules including H2O2 and Ca(2+) Here, we asked whether the H2O2-sensitive transient receptor potential melastatin 2 (TRPM2) Ca(2+) channel serves as a molecular link between H2O2 and Ca(2+) H2O2-mediated activation of TRPM2 channels induced filopodia formation, loss of actin stress fibres and disassembly of focal adhesions, leading to increased migration of HeLa and prostate cancer (PC)-3 cells. Activation of TRPM2 channels, however, caused intracellular release of not only Ca(2+) but also of Zn(2+) Intriguingly, elevation of intracellular Zn(2+) faithfully reproduced all of the effects of H2O2, whereas Ca(2+) showed opposite effects. Interestingly, H2O2 caused increased trafficking of Zn(2+)-enriched lysosomes to the leading edge of migrating cells, presumably to impart polarisation of Zn(2+) location. Thus, our results indicate that a reciprocal interplay between Ca(2+) and Zn(2+) regulates actin remodelling and cell migration; they call for a revision of the current notion that implicates an exclusive role for Ca(2+) in cell migration.


Assuntos
Movimento Celular/genética , Neoplasias/genética , Neovascularização Patológica/genética , Canais de Cátion TRPM/genética , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/genética , Morte Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Peróxido de Hidrogênio/metabolismo , Lisossomos/genética , Lisossomos/metabolismo , Metástase Neoplásica , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/metabolismo , Pseudópodes/genética , Ativação Transcricional/genética , Zinco/metabolismo
3.
Biochem J ; 466(3): 537-46, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25562606

RESUMO

Reactive oxygen species (ROS) can cause pancreatic ß-cell death by activating transient receptor potential (melastatin) 2 (TRPM2) channels. Cell death has been attributed to the ability of these channels to raise cytosolic Ca2+. Recent studies however revealed that TRPM2 channels can also conduct Zn2+, but the physiological relevance of this property is enigmatic. Given that Zn2+ is cytotoxic, we asked whether TRPM2 channels can permeate sufficient Zn2+ to affect cell viability. To address this, we used the insulin secreting (INS1) ß-cell line, human embryonic kidney (HEK)-293 cells transfected with TRPM2 and pancreatic islets. H2O2 activation of TRPM2 channels increases the cytosolic levels of both Ca2+ and Zn2+ and causes apoptotic cell death. Interestingly, chelation of Zn2+ alone was sufficient to prevent ß-cell death. The source of the cytotoxic Zn2+ is intracellular, found largely sequestered in lysosomes. Lysosomes express TRPM2 channels, providing a potential route for Zn2+ release. Zn2+ release is potentiated by extracellular Ca2+ entry, indicating that Ca2+-induced Zn2+ release leads to apoptosis. Knockout of TRPM2 channels protects mice from ß-cell death and hyperglycaemia induced by multiple low-dose streptozotocin (STZ; MLDS) administration. These results argue that TRPM2-mediated, Ca2+-potentiated Zn2+ release underlies ROS-induced ß-cell death and Zn2+, rather than Ca2+, plays a primary role in apoptosis.


Assuntos
Células Secretoras de Insulina/metabolismo , Líquido Intracelular/metabolismo , Canais de Cátion TRPM/fisiologia , Zinco/metabolismo , Animais , Morte Celular/fisiologia , Células HEK293 , Humanos , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo
4.
J Biol Chem ; 288(3): 1568-81, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23223335

RESUMO

Cardiac ATP-sensitive potassium (K(ATP)) channels are key sensors and effectors of the metabolic status of cardiomyocytes. Alteration in their expression impacts their effectiveness in maintaining cellular energy homeostasis and resistance to injury. We sought to determine how activation of calcium/calmodulin-dependent protein kinase II (CaMKII), a central regulator of calcium signaling, translates into reduced membrane expression and current capacity of cardiac K(ATP) channels. We used real-time monitoring of K(ATP) channel current density, immunohistochemistry, and biotinylation studies in isolated hearts and cardiomyocytes from wild-type and transgenic mice as well as HEK cells expressing wild-type and mutant K(ATP) channel subunits to track the dynamics of K(ATP) channel surface expression. Results showed that activation of CaMKII triggered dynamin-dependent internalization of K(ATP) channels. This process required phosphorylation of threonine at 180 and 224 and an intact (330)YSKF(333) endocytosis motif of the K(ATP) channel Kir6.2 pore-forming subunit. A molecular model of the µ2 subunit of the endocytosis adaptor protein, AP2, complexed with Kir6.2 predicted that µ2 docks by interaction with (330)YSKF(333) and Thr-180 on one and Thr-224 on the adjacent Kir6.2 subunit. Phosphorylation of Thr-180 and Thr-224 would favor interactions with the corresponding arginine- and lysine-rich loops on µ2. We concluded that calcium-dependent activation of CaMKII results in phosphorylation of Kir6.2, which promotes endocytosis of cardiac K(ATP) channel subunits. This mechanism couples the surface expression of cardiac K(ATP) channels with calcium signaling and reveals new targets to improve cardiac energy efficiency and stress resistance.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Regulação da Expressão Gênica , Miócitos Cardíacos/enzimologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Complexo 2 de Proteínas Adaptadoras/química , Complexo 2 de Proteínas Adaptadoras/metabolismo , Animais , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/química , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Dinaminas/genética , Dinaminas/metabolismo , Endocitose , Ativação Enzimática , Células HEK293 , Humanos , Transporte de Íons , Camundongos , Camundongos Transgênicos , Modelos Moleculares , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Fosforilação , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/genética , Transdução de Sinais , Treonina/metabolismo
5.
Nature ; 451(7174): 69-72, 2008 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-18172497

RESUMO

Mammalian homologues of Drosophila melanogaster transient receptor potential (TRP) are a large family of multimeric cation channels that act, or putatively act, as sensors of one or more chemical factor. Major research objectives are the identification of endogenous activators and the determination of cellular and tissue functions of these channels. Here we show the activation of TRPC5 (canonical TRP 5) homomultimeric and TRPC5-TRPC1 heteromultimeric channels by extracellular reduced thioredoxin, which acts by breaking a disulphide bridge in the predicted extracellular loop adjacent to the ion-selectivity filter of TRPC5. Thioredoxin is an endogenous redox protein with established intracellular functions, but it is also secreted and its extracellular targets are largely unknown. Particularly high extracellular concentrations of thioredoxin are apparent in rheumatoid arthritis, an inflammatory joint disease that disables millions of people worldwide. We show that TRPC5 and TRPC1 are expressed in secretory fibroblast-like synoviocytes from patients with rheumatoid arthritis, that endogenous TRPC5-TRPC1 channels of the cells are activated by reduced thioredoxin, and that blockade of the channels enhances secretory activity and prevents the suppression of secretion by thioredoxin. The data indicate the presence of a previously unrecognized ion-channel activation mechanism that couples extracellular thioredoxin to cell function.


Assuntos
Canais de Cátion TRPC/agonistas , Canais de Cátion TRPC/metabolismo , Tiorredoxinas/farmacologia , Animais , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Linhagem Celular , Dissulfetos/química , Dissulfetos/metabolismo , Condutividade Elétrica , Humanos , Oxirredução/efeitos dos fármacos , Técnicas de Patch-Clamp , Coelhos , Canais de Cátion TRPC/química , Tiorredoxinas/química
6.
Am J Physiol Cell Physiol ; 305(1): C61-9, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23596170

RESUMO

Reactive oxygen species such as H2O2 elevates the cytosolic Ca²âº concentration ([Ca²âº]c) and causes cell death via poly(ADPR) polymerase (PARP) activation, which also represents the primary mechanism by which H2O2 activate the transient receptor potential melastatin-related 2 (TRPM2) channel as a Ca²âº-permeable channel present in the plasma membrane or an intracellular Ca²âº-release channel. The present study aimed to define the contribution and mechanisms of the TRPM2 channels in macrophage cells in mediating Ca²âº signaling and cell death during initial response to H2O2, using mouse peritoneal macrophage, RAW264.7, and differentiated THP-1 cells. H2O2 evoked robust increases in the [Ca²âº]c, and such Ca²âº responses were significantly greater at body temperature than room temperature. H2O2-induced Ca²âº responses were strongly inhibited by pretreatment with PJ-34, a PARP inhibitor, and largely prevented by removal of extracellular Ca²âº. Furthermore, H2O2-induced increases in the [Ca²âº]c were completely abolished in macrophage cells isolated from trpm2-/- mice. H2O2 reduced macrophage cell viability in a duration- and concentration-dependent manner. H2O2-induced cell death was significantly attenuated by pretreatment with PJ-34 and TRPM2 channel deficiency but remained significant and persistent. Taken together, these results show that the TRPM2 channel in macrophage cells functions as a cell surface Ca²âº-permeable channel that mediates Ca²âº influx and constitutes the principal Ca²âº signaling mechanism but has a limited, albeit significant, role in cell death during early exposure to H2O2.


Assuntos
Sinalização do Cálcio/fisiologia , Peróxido de Hidrogênio/farmacologia , Macrófagos Peritoneais/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Morte Celular , Linhagem Celular , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Mutação , Canais de Cátion TRPM/genética
7.
Mol Membr Biol ; 29(8): 321-32, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22881396

RESUMO

Voltage-gated ion (K(+), Na(+), Ca(2+)) channels contain a pore domain (PD) surrounded by four voltage sensing domains (VSD). Each VSD is made up of four transmembrane helices, S1-S4. S4 contains 6-7 positively charged residues (arginine/lysine) separated two hydrophobic residues, whereas S1-S3 contribute to two negatively charged clusters. These structures are conserved among all members of the voltage-gated ion channel family and play essential roles in voltage gating. The role of S4 charged residues in voltage gating is well established: During depolarization, they move out of the membrane electric field, exerting a mechanical force on channel gates, causing them to open. However, the role of the intervening hydrophobic residues in voltage sensing is unclear. Here we studied the role of these residues in the prototypical Shaker potassium channel. We have altered the physicochemical properties of both charged and hydrophobic positions of S4 and examined the effect of these modifications on the gating properties of the channel. For this, we have introduced cysteines at each of these positions, expressed the mutants in Xenopus oocytes, and examined the effect of in situ addition of charge, via Cd(2+), on channel gating by two-electrode voltage clamp. Our results reveal a face of the S4 helix (comprising residues L358, L361, R365 and R368) where introduction of charge at hydrophobic positions destabilises the closed state and removal of charges from charged positions has an opposite effect. We propose that hydrophobic residues play a crucial role in limiting gating to a physiological voltage range.


Assuntos
Interações Hidrofóbicas e Hidrofílicas , Superfamília Shaker de Canais de Potássio/química , Superfamília Shaker de Canais de Potássio/metabolismo , Sequência de Aminoácidos , Animais , Cádmio/farmacologia , Cisteína/genética , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Íons , Cinética , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Mutação/genética , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Transporte Proteico/efeitos dos fármacos , Xenopus
8.
J Biol Chem ; 286(27): 23789-98, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21602277

RESUMO

Zinc ion (Zn(2+)) is an endogenous allosteric modulator that regulates the activity of a wide variety of ion channels in a reversible and concentration-dependent fashion. Here we used patch clamp recording to study the effects of Zn(2+) on the melastatin transient receptor potential 2 (TRPM2) channel. Zn(2+) inhibited the human (h) TRPM2 channel currents, and the steady-state inhibition was largely not reversed upon washout and concentration-independent in the range of 30-1000 µM, suggesting that Zn(2+) induces channel inactivation. Zn(2+) inactivated the channels fully when they conducted inward currents, but only by half when they passed outward currents, indicating profound influence of the permeant ion on Zn(2+) inactivation. Alanine substitution scanning mutagenesis of 20 Zn(2+)-interacting candidate residues in the outer pore region of the hTRPM2 channel showed that mutation of Lys(952) in the extracellular end of the fifth transmembrane segment and Asp(1002) in the large turret strongly attenuated or abolished Zn(2+) inactivation, and mutation of several other residues dramatically changed the inactivation kinetics. The mouse (m) TRPM2 channels were also inactivated by Zn(2+), but the kinetics were remarkably slower. Reciprocal mutation of His(995) in the hTRPM2 channel and the equivalent Gln(992) in the mTRPM2 channel completely swapped the kinetics, but no such opposing effects resulted from exchanging another pair of species-specific residues, Arg(961)/Ser(958). We conclude from these results that Zn(2+) inactivates the TRPM2 channels and that residues in the outer pore are critical determinants of the inactivation.


Assuntos
Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/metabolismo , Zinco/metabolismo , Substituição de Aminoácidos , Animais , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Camundongos , Mutação de Sentido Incorreto , Canais de Cátion TRPM/genética , Zinco/farmacologia
9.
J Biol Chem ; 286(10): 8176-8187, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21205829

RESUMO

P2X(7) receptors are important in mediating the physiological functions of extracellular ATP, and altered receptor expression and function have a causative role in the disease pathogenesis. Here, we investigated the mechanisms determining the P2X(7) receptor function by following two human single-nucleotide polymorphism (SNP) mutations that replace His-155 and Ala-348 in the human (h) P2X(7) receptor with the corresponding residues, Tyr-155 and Thr-348, in the rat (r) P2X(7) receptor. H155Y and A348T mutations in the hP2X(7) receptor increased ATP-induced currents, whereas the reciprocal mutations, Y155H and T348A, in the rP2X(7) receptor caused the opposite effects. Such a functional switch is a compelling indication that these residues are critical for P2X(7) receptor function. Additional mutations of His-155 and Ala-348 in the hP2X(7) receptor to residues with diverse side chains revealed a different dependence on the side chain properties, supporting the specificity of these two residues. Substitutions of the residues surrounding His-155 and Ala-348 in the hP2X(7) receptor with the equivalent ones in the rP2X(7) receptor also affected ATP-induced currents but were not fully reminiscent of the H155Y and A348T effects. Immunofluorescence imaging and biotin labeling assays showed that H155Y in the hP2X(7) receptor increased and Y155H in the rP2X(7) receptor decreased cell-surface expression. Such contrasting effects were not obvious with the reciprocal mutations of residue 348. Taken together, our results suggest that residues at positions 155 and 348 contribute to P2X(7) receptor function via determining the surface expression and the single-channel function, respectively. Such interpretations are consistent with the locations of the residues in the structural model of the hP2X(7) receptor.


Assuntos
Regulação da Expressão Gênica/fisiologia , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Receptores Purinérgicos P2X7/metabolismo , Trifosfato de Adenosina/genética , Substituição de Aminoácidos , Animais , Células HEK293 , Humanos , Modelos Moleculares , Ratos , Receptores Purinérgicos P2X7/genética , Peixe-Zebra
10.
J Biol Chem ; 285(8): 5963-73, 2010 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-20026601

RESUMO

Pancreatic ATP-sensitive potassium (K(ATP)) channels control insulin secretion by coupling the excitability of the pancreatic beta-cell to glucose metabolism. Little is currently known about how the plasma membrane density of these channels is regulated. We therefore set out to examine in detail the endocytosis and recycling of these channels and how these processes are regulated. To achieve this goal, we expressed K(ATP) channels bearing an extracellular hemagglutinin epitope in human embryonic kidney cells and followed their fate along the endocytic pathway. Our results show that K(ATP) channels undergo multiple rounds of endocytosis and recycling. Further, activation of protein kinase C (PKC) with phorbol 12-myristate 13-acetate significantly decreases K(ATP) channel surface density by reducing channel recycling and diverting the channel to lysosomal degradation. These findings were recapitulated in the model pancreatic beta-cell line INS1e, where activation of PKC leads to a decrease in the surface density of native K(ATP) channels. Because sorting of internalized channels between lysosomal and recycling pathways could have opposite effects on the excitability of pancreatic beta-cells, we propose that PKC-regulated K(ATP) channel trafficking may play a role in the regulation of insulin secretion.


Assuntos
Endocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Lisossomos/metabolismo , Canais de Potássio/metabolismo , Proteína Quinase C/metabolismo , Carcinógenos/farmacologia , Linhagem Celular , Endocitose/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Humanos , Insulina/genética , Secreção de Insulina , Lisossomos/genética , Modelos Biológicos , Canais de Potássio/genética , Proteína Quinase C/genética , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Acetato de Tetradecanoilforbol/farmacologia
11.
Hum Mol Genet ; 18(13): 2400-13, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19357197

RESUMO

The ATP-sensitive potassium (K(ATP)) channel controls insulin secretion by coupling glucose metabolism to excitability of the pancreatic beta-cell membrane. The channel comprises four subunits each of Kir6.2 and the sulphonylurea receptor (SUR1), encoded by KCNJ11 and ABCC8, respectively. Mutations in these genes that result in reduced activity or expression of K(ATP) channels lead to enhanced beta-cell excitability, insulin hypersecretion and hypoglycaemia, and in humans lead to the clinical condition congenital hyperinsulinism (CHI). Here we have investigated the molecular basis of the focal form of CHI caused by one such mutation in Kir6.2, E282K. The study led to the discovery that Kir6.2 contains a di-acidic ER exit signal, (280)DLE(282), which promotes concentration of the channel into COPII-enriched ER exit sites prior to ER export via a process that requires Sar1-GTPase. The E282K mutation abrogates the exit signal, and thereby prevents the ER export and surface expression of the channel. When co-expressed, the mutant subunit was able to associate with the wild-type Kir6.2 and form functional channels. Thus unlike most mutations, the E282K mutation does not cause protein mis-folding. Since in focal CHI, maternal chromosome containing the K(ATP) channel genes is lost, beta-cells of the patient would lack wild-type Kir6.2 to rescue the mutant Kir6.2 subunit expressed from the paternal chromosome. The resultant absence of functional K(ATP) channels leads to insulin hypersecretion. Taken together, we conclude that surface expression of K(ATP) channels is critically dependent on the Sar1-GTPase-dependent ER exit mechanism and abrogation of the di-acidic ER exit signal leads to CHI.


Assuntos
Hiperinsulinismo Congênito/genética , Retículo Endoplasmático/metabolismo , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Mutação de Sentido Incorreto , Canais de Potássio Corretores do Fluxo de Internalização/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular , Hiperinsulinismo Congênito/metabolismo , Retículo Endoplasmático/genética , Humanos , Insulina/metabolismo , Secreção de Insulina , Canais KATP/genética , Canais KATP/metabolismo , Dados de Sequência Molecular , Proteínas Monoméricas de Ligação ao GTP/genética , Canais de Potássio Corretores do Fluxo de Internalização/química , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ligação Proteica , Transporte Proteico , Alinhamento de Sequência
12.
Mol Membr Biol ; 26(8): 435-47, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19878047

RESUMO

The hERG potassium channel is a member of the voltage gated potassium (Kv) channel family, comprising a pore domain and four voltage sensing domains (VSDs). Like other Kv channels, the VSD senses changes in membrane voltage and transmits the signal to gates located in the pore domain; the gates open at positive potentials (activation) and close at negative potentials, thereby controlling the ion flux. hERG, however, differs from other Kv channels in that it is activated slowly but inactivated rapidly - a property that is crucial for the role it plays in the repolarization of the cardiac action potential. Voltage-gating requires movement of gating charges across the membrane electric field, which is accomplished by the transmembrane movement of the fourth transmembrane segment, S4, of the VSD containing the positively charged arginine or lysine residues. Here we ask if the functional differences between hERG and other Kv channels could arise from differences in the transmembrane movement of S4. To address this, we have introduced single cysteine residues into the S4 region of the VSD, expressed the mutant channels in Xenopus oocytes and examined the effect of membrane impermeable para-chloromercuribenzene sulphonate on function by the two-electrode voltage clamp technique. Our results show that depolarization results in the accessibility of seven consecutive S4 residues, including the first two charged residues, K525 and R528, to extracellularly applied reagent. These data indicate that the extent of S4 movement in hERG is similar to other Kv channels, including the archabacterial KvAP and the Shaker channel of Drosophila.


Assuntos
Canais de Potássio Éter-A-Go-Go/química , Potenciais da Membrana/fisiologia , Aminoácidos , Animais , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/fisiologia , Humanos , Movimento (Física) , Mutagênese Sítio-Dirigida , Oócitos , Técnicas de Patch-Clamp , Estrutura Terciária de Proteína , Transfecção , Xenopus
13.
Biochim Biophys Acta Biomembr ; 1862(5): 183192, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31945320

RESUMO

Membrane proteins are traditionally extracted and purified in detergent for biochemical and structural characterisation. This process is often costly and laborious, and the stripping away of potentially stabilising lipids from the membrane protein of interest can have detrimental effects on protein integrity. Recently, styrene-maleic acid (SMA) co-polymers have offered a solution to this problem by extracting membrane proteins directly from their native membrane, while retaining their naturally associated lipids in the form of stable SMA lipid particles (SMALPs). However, the inherent nature and heterogeneity of the polymer renders their use challenging for some downstream applications - particularly mass spectrometry (MS). While advances in cryo-electron microscopy (cryo-EM) have enhanced our understanding of membrane protein:lipid interactions in both SMALPs and detergent, the resolution obtained with this technique is often insufficient to accurately identify closely associated lipids within the transmembrane annulus. Native-MS has the power to fill this knowledge gap, but the SMA polymer itself remains largely incompatible with this technique. To increase sample homogeneity and allow characterisation of membrane protein:lipid complexes by native-MS, we have developed a novel SMA-exchange method; whereby the membrane protein of interest is first solubilised and purified in SMA, then transferred into amphipols or detergents. This allows the membrane protein and endogenously associated lipids extracted by SMA co-polymer to be identified and examined by MS, thereby complementing results obtained by cryo-EM and creating a better understanding of how the lipid bilayer directly affects membrane protein structure and function.


Assuntos
Maleatos/química , Lipídeos de Membrana/isolamento & purificação , Proteínas de Membrana/isolamento & purificação , Poliestirenos/química , Microscopia Crioeletrônica/métodos , Detergentes , Escherichia coli/química , Proteínas de Escherichia coli/química , Bicamadas Lipídicas/química , Gotículas Lipídicas/química , Espectrometria de Massas/métodos , Lipídeos de Membrana/metabolismo , Proteínas de Membrana/química , Polímeros/química
14.
Trends Cell Biol ; 13(12): 639-47, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14624842

RESUMO

Transmembrane domain (TMD) proteins comprise a major group of proteins that perform a wide range of functions and act to translate extracellular signals to intracellular responses. They include G-protein coupled receptors (GPCRs), growth factor receptors, ion channels, transporters and metabolic enzymes. In this review, we focus on the current understanding of trafficking of mutant membrane proteins in human disease and speculate on therapeutic strategies.


Assuntos
Transporte Biológico , Proteínas de Membrana/metabolismo , Transporte Proteico , Membrana Celular/química , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Fibrose Cística/terapia , Diabetes Insípido/metabolismo , Diabetes Insípido/patologia , Diabetes Insípido/terapia , Humanos , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia , Retinose Pigmentar/terapia , Transdução de Sinais
15.
Sci Rep ; 9(1): 16876, 2019 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-31727906

RESUMO

Glucose and hypotonicity induced cell swelling stimulate insulin release from pancreatic ß-cells but the mechanisms are poorly understood. Recently, Piezo1 was identified as a mechanically-activated nonselective Ca2+ permeable cationic channel in a range of mammalian cells. As cell swelling induced insulin release could be through stimulation of Ca2+ permeable stretch activated channels, we hypothesised a role for Piezo1 in cell swelling induced insulin release. Two rat ß-cell lines (INS-1 and BRIN-BD11) and freshly-isolated mouse pancreatic islets were studied. Intracellular Ca2+ measurements were performed using the fura-2 Ca2+ indicator dye and ionic current was recorded by whole cell patch-clamp. Piezo1 agonist Yoda1, a competitive antagonist of Yoda1 (Dooku1) and an inactive analogue of Yoda1 (2e) were used as chemical probes. Piezo1 mRNA and insulin secretion were measured by RT-PCR and ELISA respectively. Piezo1 mRNA was detected in both ß-cell lines and mouse islets. Yoda1 evoked Ca2+ entry was inhibited by Yoda1 antagonist Dooku1 as well as other Piezo1 inhibitors gadolinium and ruthenium red, and not mimicked by 2e. Yoda1, but not 2e, stimulated Dooku1-sensitive insulin release from ß-cells and pancreatic islets. Hypotonicity and high glucose increased intracellular Ca2+ and enhanced Yoda1 Ca2+ influx responses. Yoda1 and hypotonicity induced insulin release were significantly inhibited by Piezo1 specific siRNA. Pancreatic islets from mice with haploinsufficiency of Piezo1 released less insulin upon exposure to Yoda1. The data show that Piezo1 channel agonist induces insulin release from ß-cell lines and mouse pancreatic islets suggesting a role for Piezo1 in cell swelling induced insulin release. Hence Piezo1 agonists have the potential to be used as enhancers of insulin release.


Assuntos
Cálcio/metabolismo , Glucose/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/metabolismo , Canais Iônicos/genética , Proteínas de Membrana/genética , Animais , Transporte Biológico/efeitos dos fármacos , Linhagem Celular Tumoral , Gadolínio/farmacologia , Regulação da Expressão Gênica , Glucose/metabolismo , Heterozigoto , Secreção de Insulina/genética , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/metabolismo , Mecanotransdução Celular , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pirazinas/farmacologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Rutênio Vermelho/farmacologia , Tiadiazóis/farmacologia , Técnicas de Cultura de Tecidos
16.
Methods Mol Biol ; 491: 63-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18998083

RESUMO

Macroscopic ion channel currents (I) are a product of the channel open probability (P (o)), the single channel current (i) and the number of channels present on the cell surface (N) at any given time (I = P (o) iN). Endocytosis has been shown to be one of the key determinants of cell surface channel density and the defects of this process have been linked to diseases relating to ion channel dysfunction. Chemiluminescence-based techniques provide a rapid method for the examination of the rates of endocytosis and steady-state cell surface density of ion channels and have previously been used to investigate the endocytosis of pancreatic ATP-sensitive potassium (K(ATP)) channels.


Assuntos
Membrana Celular/fisiologia , Clatrina/fisiologia , Endocitose/fisiologia , Canais KATP/fisiologia , Linhagem Celular , Humanos , Canais KATP/genética , Rim , Cinética , Luminescência
17.
Methods Mol Biol ; 491: 69-77, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18998084

RESUMO

Macroscopic ion channel currents (1) are a product of the channel open probability (P(o)), the single channel current (i) and the number of channels present on the cell surface (N) at any given time (I = P(o)iN). Intra-cellular trafficking pathways are proving to be of vital importance in regulating ion channel function since endocytosis, recycling and degradation all work in concert to maintain appropriate channel numbers on the cell surface. Immunofluorescence-based techniques provide a convenient and rapid method for the examination of these processes and have been used to investigate the intracellular trafficking of pancreatic ATP-sensitive potassium (K(ATP)) channels.


Assuntos
Membrana Celular/fisiologia , Endocitose/fisiologia , Imunofluorescência/métodos , Canais KATP/fisiologia , Comunicação Celular , Linhagem Celular , Meios de Cultura , Humanos , Rim , Potenciais da Membrana/fisiologia , Organelas/fisiologia
18.
Methods Mol Biol ; 491: 79-89, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18998085

RESUMO

Macroscopic ion channel currents (I) are a product of the channel open probability (P(o)), the single channel current (i) and the number of channels present on the cell surface (N) at any given time (I = P(o)iN). Endocytosis has been shown to be one of the key determinants of cell surface channel density and defects of this process have been linked to diseases relating to ion channel dysfunction. Biotinylation allows the selective labelling and isolation of surface exposed proteins which can then be identified by Western blotting.


Assuntos
Membrana Celular/fisiologia , Endocitose/fisiologia , Canais KATP/fisiologia , Biotinilação , Western Blotting/métodos , Linhagem Celular , Eletroforese em Gel de Poliacrilamida/métodos , Humanos , Indicadores e Reagentes , Canais KATP/isolamento & purificação , Rim/embriologia
19.
Nat Biotechnol ; 23(10): 1289-93, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16170312

RESUMO

Here we describe a strategy for generating ion-channel inhibitors. It takes advantage of antibody specificity combined with a pattern recognition approach that targets the third extracellular region (E3) of a channel. To test the concept, we first focused on TRPC5, a member of the transient receptor potential (TRP) calcium channel family, the study of which has been hindered by poor pharmacological tools. Extracellular application of E3-targeted anti-TRPC5 antibody led to a specific TRPC5 inhibitor, enabling TRPC5 to be distinguished from its closest family members, and TRPC5 function to be explored in a relatively intractable physiological system. E3 targeting was further applied to voltage-gated sodium channels, leading to discovery of a subtype-specific inhibitor of Na(V)1.5. These examples illustrate the potential power of E3 targeting as a systematic method for producing gene-type specific ion-channel inhibitors for use in routine assays on cells or tissues from a range of species and having therapeutic potential.


Assuntos
Anticorpos Monoclonais/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Engenharia de Proteínas/métodos , Canais de Cátion TRPC/efeitos dos fármacos , Canais de Cátion TRPC/fisiologia , Sítios de Ligação , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Ligação Proteica
20.
Expert Rev Mol Med ; 9(21): 1-17, 2007 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-17666135

RESUMO

ATP-sensitive potassium (KATP) channels play a key role in the regulation of insulin secretion by coupling glucose metabolism to the electrical activity of pancreatic beta-cells. To generate an electric signal of suitable magnitude, the plasma membrane of the beta-cell must contain an appropriate number of channels. An inadequate number of channels can lead to congenital hyperinsulinism, whereas an excess of channels can result in the opposite condition, neonatal diabetes. KATP channels are made up of four subunits each of Kir6.2 and the sulphonylurea receptor (SUR1), encoded by the genes KCNJ11 and ABCC8, respectively. Following synthesis, the subunits must assemble into an octameric complex to be able to exit the endoplasmic reticulum and reach the plasma membrane. While this biosynthetic pathway ensures supply of channels to the cell surface, an opposite pathway, involving clathrin-mediated endocytosis, removes channels back into the cell. The balance between these two processes, perhaps in conjunction with endocytic recycling, would dictate the channel density at the cell membrane. In this review, we discuss the molecular signals that contribute to this balance, and how an imbalance could lead to a disease state such as neonatal diabetes.


Assuntos
Diabetes Mellitus/metabolismo , Insulina/metabolismo , Erros Inatos do Metabolismo/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Diabetes Mellitus/genética , Endocitose , Humanos , Hiperinsulinismo/genética , Hiperinsulinismo/metabolismo , Recém-Nascido , Erros Inatos do Metabolismo/genética , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/ultraestrutura , Transporte Proteico/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA