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










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-38854018

RESUMO

Targeted recruitment of E3 ubiquitin ligases to degrade traditionally undruggable proteins is a disruptive paradigm for developing new therapeutics. Two salient limitations are that <2% of the ~600 E3 ligases in the human genome have been exploited to produce proteolysis targeting chimeras (PROTACs), and the efficacy of the approach has not been demonstrated for a vital class of complex multi-subunit membrane proteins- ion channels. NEDD4-1 and NEDD4-2 are physiological regulators of myriad ion channels, and belong to the 28-member HECT (homologous to E6AP C-terminus) family of E3 ligases with widespread roles in cell/developmental biology and diverse diseases including various cancers, immunological and neurological disorders, and chronic pain. The potential efficacy of HECT E3 ligases for targeted protein degradation is unexplored, constrained by a lack of appropriate binders, and uncertain due to their complex regulation by layered intra-molecular and posttranslational mechanisms. Here, we identified a nanobody that binds with high affinity and specificity to a unique site on the N-lobe of the NEDD4-2 HECT domain at a location physically separate from sites critical for catalysis- the E2 binding site, the catalytic cysteine, and the ubiquitin exosite- as revealed by a 3.1 Å cryo-electron microscopy reconstruction. Recruiting endogenous NEDD4-2 to diverse ion channel proteins (KCNQ1, ENaC, and CaV2.2) using a divalent (DiVa) nanobody format strongly reduced their functional expression with minimal off-target effects as assessed by global proteomics, compared to simple NEDD4-2 overexpression. The results establish utility of a HECT E3 ligase for targeted protein downregulation, validate a class of complex multi-subunit membrane proteins as susceptible to this modality, and introduce endogenous E3 ligase recruitment with DiVa nanobodies as a general method to generate novel genetically-encoded ion channel inhibitors.

2.
Mol Ther Nucleic Acids ; 34: 102032, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37842167

RESUMO

Disease-causing premature termination codons (PTCs) individually disrupt the functional expression of hundreds of genes and represent a pernicious clinical challenge. In the heart, loss-of-function mutations in the hERG potassium channel account for approximately 30% of long-QT syndrome arrhythmia, a lethal cardiac disorder with limited treatment options. Premature termination of ribosomal translation produces a truncated and, for potassium channels, a potentially dominant-negative protein that impairs the functional assembly of the wild-type homotetrameric hERG channel complex. We used high-throughput flow cytometry and patch-clamp electrophysiology to assess the trafficking and voltage-dependent activity of hERG channels carrying patient PTC variants that have been corrected by anticodon engineered tRNA. Adenoviral-mediated expression of mutant hERG channels in cultured adult guinea pig cardiomyocytes prolonged action potential durations, and this deleterious effect was corrected upon adenoviral delivery of a human ArgUGA tRNA to restore full-length hERG protein. The results demonstrate mutation-specific, context-agnostic PTC correction and elevate the therapeutic potential of this approach for rare genetic diseases caused by stop codons.

3.
Nat Commun ; 13(1): 7556, 2022 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-36494348

RESUMO

Ca2+ influx through high-voltage-activated calcium channels (HVACCs) controls diverse cellular functions. A critical feature enabling a singular signal, Ca2+ influx, to mediate disparate functions is diversity of HVACC pore-forming α1 and auxiliary CaVß1-CaVß4 subunits. Selective CaVα1 blockers have enabled deciphering their unique physiological roles. By contrast, the capacity to post-translationally inhibit HVACCs based on CaVß isoform is non-existent. Conventional gene knockout/shRNA approaches do not adequately address this deficit owing to subunit reshuffling and partially overlapping functions of CaVß isoforms. Here, we identify a nanobody (nb.E8) that selectively binds CaVß1 SH3 domain and inhibits CaVß1-associated HVACCs by reducing channel surface density, decreasing open probability, and speeding inactivation. Functionalizing nb.E8 with Nedd4L HECT domain yielded Chisel-1 which eliminated current through CaVß1-reconstituted CaV1/CaV2 and native CaV1.1 channels in skeletal muscle, strongly suppressed depolarization-evoked Ca2+ influx and excitation-transcription coupling in hippocampal neurons, but was inert against CaVß2-associated CaV1.2 in cardiomyocytes. The results introduce an original method for probing distinctive functions of ion channel auxiliary subunit isoforms, reveal additional dimensions of CaVß1 signaling in neurons, and describe a genetically-encoded HVACC inhibitor with unique properties.


Assuntos
Canais de Cálcio , Miócitos Cardíacos , Canais de Cálcio/metabolismo , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Domínios de Homologia de src , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Canais de Cálcio Tipo L/genética , Canais de Cálcio Tipo L/metabolismo , Cálcio/metabolismo
4.
Nat Methods ; 17(12): 1245-1253, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33169015

RESUMO

Impaired protein stability or trafficking underlies diverse ion channelopathies and represents an unexploited unifying principle for developing common treatments for otherwise dissimilar diseases. Ubiquitination limits ion channel surface density, but targeting this pathway for the purposes of basic study or therapy is challenging because of its prevalent role in proteostasis. We developed engineered deubiquitinases (enDUBs) that enable selective ubiquitin chain removal from target proteins to rescue the functional expression of disparate mutant ion channels that underlie long QT syndrome (LQT) and cystic fibrosis (CF). In an LQT type 1 (LQT1) cardiomyocyte model, enDUB treatment restored delayed rectifier potassium currents and normalized action potential duration. CF-targeted enDUBs synergistically rescued common (ΔF508) and pharmacotherapy-resistant (N1303K) CF mutations when combined with the US Food and Drug Administation (FDA)-approved drugs Orkambi (lumacaftor/ivacaftor) and Trikafta (elexacaftor/tezacaftor/ivacaftor and ivacaftor). Altogether, targeted deubiquitination via enDUBs provides a powerful protein stabilization method that not only corrects diverse diseases caused by impaired ion channel trafficking, but also introduces a new tool for deconstructing the ubiquitin code in situ.


Assuntos
Canalopatias/patologia , Fibrose Cística/patologia , Enzimas Desubiquitinantes/metabolismo , Transporte de Íons/fisiologia , Síndrome do QT Longo/patologia , Canais de Potássio/fisiologia , Aminofenóis/farmacologia , Aminopiridinas/farmacologia , Benzodioxóis/farmacologia , Canalopatias/genética , Fibrose Cística/genética , Enzimas Desubiquitinantes/genética , Combinação de Medicamentos , Humanos , Indóis/farmacologia , Transporte de Íons/genética , Síndrome do QT Longo/genética , Miócitos Cardíacos/fisiologia , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Canais de Potássio/genética , Pirazóis/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia , Quinolonas/farmacologia
5.
Proc Natl Acad Sci U S A ; 115(47): 12051-12056, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30397133

RESUMO

Genetically encoded inhibitors for voltage-dependent Ca2+ (CaV) channels (GECCIs) are useful research tools and potential therapeutics. Rad/Rem/Rem2/Gem (RGK) proteins are Ras-like G proteins that potently inhibit high voltage-activated (HVA) Ca2+ (CaV1/CaV2 family) channels, but their nonselectivity limits their potential applications. We hypothesized that nonselectivity of RGK inhibition derives from their binding to auxiliary CaVß-subunits. To investigate latent CaVß-independent components of inhibition, we coexpressed each RGK individually with CaV1 (CaV1.2/CaV1.3) or CaV2 (CaV2.1/CaV2.2) channels reconstituted in HEK293 cells with either wild-type (WT) ß2a or a mutant version (ß2a,TM) that does not bind RGKs. All four RGKs strongly inhibited CaV1/CaV2 channels reconstituted with WT ß2a By contrast, when channels were reconstituted with ß2a,TM, Rem inhibited only CaV1.2, Rad selectively inhibited CaV1.2 and CaV2.2, while Gem and Rem2 were ineffective. We generated mutant RGKs (Rem[R200A/L227A] and Rad[R208A/L235A]) unable to bind WT CaVß, as confirmed by fluorescence resonance energy transfer. Rem[R200A/L227A] selectively blocked reconstituted CaV1.2 while Rad[R208A/L235A] inhibited CaV1.2/CaV2.2 but not CaV1.3/CaV2.1. Rem[R200A/L227A] and Rad[R208A/L235A] both suppressed endogenous CaV1.2 channels in ventricular cardiomyocytes and selectively blocked 25 and 62%, respectively, of HVA currents in somatosensory neurons of the dorsal root ganglion, corresponding to their distinctive selectivity for CaV1.2 and CaV1.2/CaV2.2 channels. Thus, we have exploited latent ß-binding-independent Rem and Rad inhibition of specific CaV1/CaV2 channels to develop selective GECCIs with properties unmatched by current small-molecule CaV channel blockers.


Assuntos
Bloqueadores dos Canais de Cálcio/metabolismo , Canais de Cálcio/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Fenômenos Biofísicos , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Canais de Cálcio Tipo L/metabolismo , Canais de Cálcio Tipo N/metabolismo , Células HEK293 , Humanos , Ativação do Canal Iônico/fisiologia , Miócitos Cardíacos/metabolismo , Neurônios/metabolismo , Engenharia de Proteínas/métodos , Especificidade por Substrato/genética , Proteínas ras/metabolismo
6.
J Neurosci ; 37(9): 2336-2348, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28130358

RESUMO

The superficial dorsal horn is the synaptic termination site for many peripheral sensory fibers of the somatosensory system. A wide range of sensory modalities are represented by these fibers, including pain, itch, and temperature. Because the involvement of local inhibition in the dorsal horn, specifically that mediated by the inhibitory amino acids GABA and glycine, is so important in signal processing, we investigated regional inhibitory control of excitatory interneurons under control conditions and peripheral inflammation-induced mechanical allodynia. We found that excitatory interneurons and projection neurons in lamina I and IIo are dominantly inhibited by GABA while those in lamina IIi and III are dominantly inhibited by glycine. This was true of identified neuronal subpopulations: neurokinin 1 receptor-expressing (NK1R+) neurons in lamina I were GABA-dominant while protein kinase C gamma-expressing (PKCγ+) neurons at the lamina IIi-III border were glycine-dominant. We found this pattern of synaptic inhibition to be consistent with the distribution of GABAergic and glycinergic neurons identified by immunohistochemistry. Following complete Freund's adjuvant injection into mouse hindpaw, the frequency of spontaneous excitatory synaptic activity increased and inhibitory synaptic activity decreased. Surprisingly, these changes were accompanied by an increase in GABA dominance in lamina IIi. Because this shift in inhibitory dominance was not accompanied by a change in the number of inhibitory synapses or the overall postsynaptic expression of glycine receptor α1 subunits, we propose that the dominance shift is due to glycine receptor modulation and the depressed function of glycine receptors is partially compensated by GABAergic inhibition.SIGNIFICANCE STATEMENT Pain associated with inflammation is a sensation we would all like to minimize. Persistent inflammation leads to cellular and molecular changes in the spinal cord dorsal horn, including diminished inhibition, which may be responsible for enhance excitability. Investigating inhibition in the dorsal horn following peripheral inflammation is essential for development of improved ways to control the associated pain. In this study, we have elucidated regional differences in inhibition of excitatory interneurons in mouse dorsal horn. We have also discovered that the dominating inhibitory neurotransmission within specific regions of dorsal horn switches following peripheral inflammation and the accompanying hypersensitivity to thermal and mechanical stimuli. Our novel findings contribute to a more complete understanding of inflammatory pain.


Assuntos
Inflamação/patologia , Inibição Neural/fisiologia , Células do Corno Posterior/fisiologia , Receptores de GABA/metabolismo , Receptores de Glicina/metabolismo , Medula Espinal/citologia , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Adjuvante de Freund/toxicidade , Glicina/farmacologia , Hiperalgesia/fisiopatologia , Técnicas In Vitro , Inflamação/induzido quimicamente , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Masculino , Camundongos , Inibição Neural/efeitos dos fármacos , Medição da Dor/efeitos dos fármacos , Células do Corno Posterior/efeitos dos fármacos , Proteína Quinase C/metabolismo , Receptores da Neurocinina-1/metabolismo , Potenciais Sinápticos/efeitos dos fármacos , Ácido gama-Aminobutírico/farmacologia
7.
Neuron ; 81(6): 1312-1327, 2014 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-24583022

RESUMO

Cutaneous mechanosensory neurons detect mechanical stimuli that generate touch and pain sensation. Although opioids are generally associated only with the control of pain, here we report that the opioid system in fact broadly regulates cutaneous mechanosensation, including touch. This function is predominantly subserved by the delta opioid receptor (DOR), which is expressed by myelinated mechanoreceptors that form Meissner corpuscles, Merkel cell-neurite complexes, and circumferential hair follicle endings. These afferents also include a small population of CGRP-expressing myelinated nociceptors that we now identify as the somatosensory neurons that coexpress mu and delta opioid receptors. We further demonstrate that DOR activation at the central terminals of myelinated mechanoreceptors depresses synaptic input to the spinal dorsal horn, via the inhibition of voltage-gated calcium channels. Collectively our results uncover a molecular mechanism by which opioids modulate cutaneous mechanosensation and provide a rationale for targeting DOR to alleviate injury-induced mechanical hypersensitivity.


Assuntos
Mecanorreceptores/fisiologia , Neurônios/fisiologia , Nociceptores/fisiologia , Receptores Opioides delta/metabolismo , Medula Espinal/metabolismo , Analgésicos Opioides/farmacologia , Animais , Canais de Cálcio/metabolismo , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/fisiologia , Mecanorreceptores/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Nociceptores/efeitos dos fármacos , Dor/fisiopatologia , Medula Espinal/efeitos dos fármacos
9.
Ann N Y Acad Sci ; 1279: 90-6, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23531006

RESUMO

Sensory information transmitted to the spinal cord dorsal horn is modulated by a complex network of excitatory and inhibitory interneurons. The two main inhibitory transmitters, GABA and glycine, control the flow of sensory information mainly by regulating the excitability of dorsal horn neurons. A presynaptic action of GABA has also been proposed as an important modulatory mechanism of transmitter release from sensory primary afferent terminals. By inhibiting the release of glutamate from primary afferent terminals, activation of presynaptic GABA receptors could play an important role in nociceptive and tactile sensory coding, while changes in their expression or function could be involved in pathological pain conditions, such as allodynia.


Assuntos
Potenciais Pós-Sinápticos Inibidores/fisiologia , Inibição Neural/fisiologia , Células do Corno Posterior/fisiologia , Terminações Pré-Sinápticas/fisiologia , Animais , Glicina/metabolismo , Humanos , Modelos Biológicos , Células do Corno Posterior/citologia , Terminações Pré-Sinápticas/metabolismo , Receptores Pré-Sinápticos/metabolismo , Receptores Pré-Sinápticos/fisiologia , Medula Espinal/citologia , Medula Espinal/fisiologia , Transmissão Sináptica/fisiologia , Ácido gama-Aminobutírico/metabolismo
10.
J Neurosci Methods ; 189(2): 197-204, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20385165

RESUMO

Synapses between nociceptive dorsal root ganglion (DRG) neurons and spinal cord dorsal horn neurons represent the first loci for transmission of painful stimuli. Our knowledge of the molecular organization and development of these synapses is sparse due, partly, to a lack of a reliable model system that reconstitutes synaptogenesis between these two neuronal populations. To address this issue, we have established an in vitro assay system consisting of separately purified DRG neurons and dorsal horn neurons on astrocyte microislands. Using immunocytochemistry, we have found that 97%, 93%, 98%, 96%, and 94% of DRG neurons on these microislands express markers often associated with nociceptive neurons including Substance P, TRPV1, calcitonin-gene related peptide (CGRP), TrKA, and peripherin, respectively. Triple labeling with these nociceptive-like markers, synaptic vesicle marker Vglut2 and using MAP2 as a dendritic marker revealed the presence of nociceptive-like markers at synaptic terminals. Using this immunocytochemical approach, we counted contact points as overlapping MAP2/Vglut2 puncta and showed that they increased with time in culture. Single and dual patch-clamp recordings showed that overlapping Vglut2/MAP2 puncta observed after a few days in culture are likely to be functional synapses between DRG and dorsal horn neurons in our in vitro assay system. Taken together, these data suggest our co-culture microisland model system consists of mostly nociceptive-like DRG neurons that express presynaptic markers and form functional synapses with their dorsal horn partners. Thus, this model system may have direct application for studies on factors regulating development of nociceptive DRG/dorsal horn synapses.


Assuntos
Técnicas de Cocultura/métodos , Gânglios Espinais/fisiologia , Neurônios/fisiologia , Células do Corno Posterior/fisiologia , Sinapses/fisiologia , Animais , Astrócitos , Células Cultivadas , Colágeno , Gânglios Espinais/citologia , Imuno-Histoquímica , Potenciais da Membrana , Neurônios/citologia , Dor , Técnicas de Patch-Clamp , Células do Corno Posterior/citologia , Terminações Pré-Sinápticas/fisiologia , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
15.
Mol Cell ; 24(5): 665-675, 2006 Dec 08.
Artigo em Inglês | MEDLINE | ID: mdl-17157250

RESUMO

The Ca2+ -activated K+ channel KCa3.1 is required for Ca2+ influx and the subsequent activation of B and T cells. Inhibitors of KCa3.1 are in development to treat autoimmune diseases and transplant rejection, underscoring the importance in understanding how these channels are regulated. We show that nucleoside diphosphate kinase B (NDPK-B), a mammalian histidine kinase, functions downstream of PI(3)P to activate KCa3.1. NDPK-B directly binds and activates KCa3.1 by phosphorylating histidine 358 in the carboxyl terminus of KCa3.1. Endogenous NDPK-B is also critical for KCa3.1 channel activity and the subsequent activation of CD4 T cells. These findings provide one of the best examples whereby histidine phosphorylation regulates a biological process in mammals, and provide an example whereby a channel is regulated by histidine phosphorylation. The critical role for NDPK-B in the reactivation of CD4 T cells indicates that understanding NDPK-B regulation should uncover novel pathways required for T cell activation.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Histidina/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Núcleosídeo-Difosfato Quinase/metabolismo , Sequência de Aminoácidos , Androstadienos/farmacologia , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Células CHO , Células Cultivadas , Cricetinae , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/efeitos dos fármacos , Ativação Linfocitária/fisiologia , Dados de Sequência Molecular , Nucleosídeo NM23 Difosfato Quinases , Núcleosídeo-Difosfato Quinase/biossíntese , Fosforilação , Ligação Proteica , Fatores de Tempo , Wortmanina
16.
J Biol Chem ; 281(42): 31762-9, 2006 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16914545

RESUMO

Myotubularins (MTM) are a large subfamily of lipid phosphatases that specifically dephosphorylate at the D3 position of phosphatidylinositol 3-phosphate (PI(3)P) in PI(3)P and PI(3,5)P2. We recently found that MTMR6 specifically inhibits the Ca2+-activated K+ channel, KCa3.1, by dephosphorylating PI(3)P. We now show that inhibition is specific for MTMR6 and other MTMs do not inhibit KCa3.1. By replacing either or both of the coiled-coil (CC) and pleckstrin homology/GRAM (PH/G) domains of MTMs that failed to inhibit KCa3.1 with the CC and PH/G domains of MTMR6, we found that chimeric MTMs containing both the MTMR6 CC and PH/G domains functioned like MTMR6 to inhibit KCa3.1 channel activity, whereas chimeric MTMs containing either domain alone did not. Immunofluorescent microscopy demonstrated that both the MTMR6 CC and PH/G domains are required to co-localize MTMR6 to the plasma membrane with KCa3.1. These findings support a model in which two specific low affinity interactions are required to co-localize MTMR6 with KCa3.1: 1) between the CC domains on MTMR6 and KCa3.1 and (2) between the PH/G domain and a component of the plasma membrane. Our inability to detect significant interaction of the MTMR6 G/PH domain with phosphoinositides suggests that this domain may bind a protein. Identifying the specific binding partners of the CC and PH/G domains on other MTMs will provide important clues to the specific functions regulated by other MTMs as well as the mechanism(s) whereby loss of some MTMs lead to disease.


Assuntos
Regulação Enzimológica da Expressão Gênica , Proteínas Tirosina Fosfatases/química , Animais , Células CHO , Membrana Celular/metabolismo , Cricetinae , Cães , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/química , Técnicas de Patch-Clamp , Fosfatidilinositóis/química , Monoéster Fosfórico Hidrolases/química , Fosforilação , Estrutura Terciária de Proteína , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Fosfatases não Receptoras , Proteínas Recombinantes de Fusão/química , Especificidade por Substrato
17.
Mol Cell Biol ; 26(15): 5595-602, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847315

RESUMO

Intracellular Ca2+ levels rapidly rise following cross-linking of the T-cell receptor (TCR) and function as a critical intracellular second messenger in T-cell activation. It has been relatively under appreciated that K+ channels play an important role in Ca2+ influx into T lymphocytes by helping to maintain a negative membrane potential which provides an electrochemical gradient to drive Ca2+ influx. Here we show that the Ca2+-activated K+ channel, KCa3.1, which is critical for Ca2+ influx in reactivated naive T cells and central memory T cells, requires phosphatidylinositol-3 phosphatase [PI(3)P] for activation and is inhibited by the PI(3)P phosphatase myotubularin-related protein 6 (MTMR6). Moreover, by inhibiting KCa3.1, MTMR6 functions as a negative regulator of Ca2+ influx and proliferation of reactivated human CD4 T cells. These findings point to a new and unexpected role for PI(3)P and the PI(3)P phosphatase MTMR6 in the regulation of Ca2+ influx in activated CD4 T cells and suggest that MTMR6 plays a critical role in setting a minimum threshold for a stimulus to activate a T cell.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Linfócitos T CD4-Positivos/citologia , Cálcio/metabolismo , Proliferação de Células , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Ativação Linfocitária , Técnicas de Patch-Clamp , Monoéster Fosfórico Hidrolases/genética , Proteínas Tirosina Fosfatases não Receptoras , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo
18.
J Neurochem ; 96(5): 1227-41, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16464240

RESUMO

Rats learning the Morris water maze exhibit hippocampal changes in synaptic morphology and physiology that manifest as altered synaptic efficacy. Learning requires structural changes in the synapse, and multiple cell adhesion molecules appear to participate. The activity of these cell adhesion molecules is, in large part, dependent on their interaction with the extracellular matrix (ECM). Given that matrix metalloproteinases (MMPs) are responsible for transient alterations in the ECM, we predicted that MMP function is critical for hippocampal-dependent learning. In support of this, it was observed that hippocampal MMP-3 and -9 increased transiently during water maze acquisition as assessed by western blotting and mRNA analysis. The ability of the NMDA receptor channel blocker MK801 to attenuate these changes indicated that the transient MMP changes were in large part dependent upon NMDA receptor activation. Furthermore, inhibition of MMP activity with MMP-3 and -9 antisense oligonucleotides and/or MMP inhibitor FN-439 altered long-term potentiation and prevented acquisition in the Morris water maze. The learning-dependent MMP alterations were shown to modify the stability of the actin-binding protein cortactin, which plays an essential role in regulating the dendritic cytoskeleton and synaptic efficiency. Together these results indicate that changes in MMP function are critical to synaptic plasticity and hippocampal-dependent learning.


Assuntos
Aprendizagem/fisiologia , Metaloproteinase 3 da Matriz/fisiologia , Metaloproteinase 9 da Matriz/fisiologia , Plasticidade Neuronal/fisiologia , Comportamento Espacial/fisiologia , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Northern Blotting , Western Blotting/métodos , Cortactina/metabolismo , Maleato de Dizocilpina/farmacologia , Estimulação Elétrica/métodos , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiologia , Ácidos Hidroxâmicos/farmacologia , Imuno-Histoquímica/métodos , Aprendizagem/efeitos dos fármacos , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/efeitos da radiação , Masculino , Metaloproteinase 3 da Matriz/química , Metaloproteinase 9 da Matriz/química , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Oligopeptídeos/farmacologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Comportamento Espacial/efeitos dos fármacos , Fatores de Tempo
19.
Mol Biol Cell ; 17(1): 146-54, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16251351

RESUMO

KCa3.1 is an intermediate conductance Ca2+-activated K+ channel that is expressed predominantly in hematopoietic cells, smooth muscle cells, and epithelia where it functions to regulate membrane potential, Ca2+ influx, cell volume, and chloride secretion. We recently found that the KCa3.1 channel also specifically requires phosphatidylinositol-3 phosphate [PI(3)P] for channel activity and is inhibited by myotubularin-related protein 6 (MTMR6), a PI(3)P phosphatase. We now show that PI(3)P indirectly activates KCa3.1. Unlike KCa3.1 channels, the related KCa2.1, KCa2.2, or KCa2.3 channels do not require PI(3)P for activity, suggesting that the KCa3.1 channel has evolved a unique means of regulation that is critical for its biological function. By making chimeric channels between KCa3.1 and KCa2.3, we identified a stretch of 14 amino acids in the carboxy-terminal calmodulin binding domain of KCa3.1 that is sufficient to confer regulation of KCa2.3 by PI(3)P. However, mutation of a single potential phosphorylation site in these 14 amino acids did not affect channel activity. These data together suggest that PI(3)P and these 14 amino acids regulate KCa3.1 channel activity by recruiting an as yet to be defined regulatory subunit that is required for Ca2+ gating of KCa3.1.


Assuntos
Canais de Potássio Ativados por Cálcio de Condutância Intermediária/química , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Sequência de Aminoácidos , Animais , Células CHO , Membrana Celular/metabolismo , Sequência Conservada , Cricetinae , Citosol , Eletrofisiologia , Ativação Enzimática , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Dados de Sequência Molecular , Mutação/genética , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Monoéster Fosfórico Hidrolases , Fosforilação , Proteínas Tirosina Fosfatases não Receptoras , Ratos , Alinhamento de Sequência
20.
Biochemistry ; 42(18): 5453-60, 2003 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-12731887

RESUMO

Eukaryotic initiation factor 2- (eIF2-) associated glycoprotein p67 blocks eIF2alpha phosphorylation by kinases, and its N-terminal 1-97 amino acid segment can induce efficient translation. To investigate whether glycosylation at the serine/threonine clusters at this region is important in protein synthesis, we selected (27)TSST(30) and (60)SGTS(63) clusters for further analysis. By site-directed mutagenesis, (27)TSST(30) and (60)SGTS(63) clusters were substituted with (27)AAGA(30) and (60)AGAA(63) amino acid residues in full-length p67, and their EGFP fusions were constitutively expressed in rat tumor hepatoma cells (KRC-7). The (60)AGAA(63) mutant blocked eIF2alpha phosphorylation less than either wild-type p67 or the (27)AAGA(30) mutant. The (60)AGAA(63) mutant also showed a low level of protein synthesis rate, a lower level of glycosylation, increased turnover rate, and weaker binding to eIF2alpha. These results suggest that glycosylation within the (60)SGTS(63) sequence of p67 plays an important role in its stability and thus its regulation of protein synthesis by modulating the phosphorylation of the alpha-subunit of eIF2.


Assuntos
Acetilglucosaminidase/metabolismo , Aminopeptidases , Fator de Iniciação 2 em Eucariotos/metabolismo , Glutationa Transferase/metabolismo , Glicoproteínas/metabolismo , Proteínas Luminescentes/metabolismo , Acetilglucosaminidase/genética , Substituição de Aminoácidos , Animais , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Primers do DNA/química , Regulação da Expressão Gênica , Glicosilação , Proteínas de Fluorescência Verde , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas Luminescentes/genética , Metionina/química , Mutagênese Sítio-Dirigida , Mutação/genética , Fosforilação , Reação em Cadeia da Polimerase , Testes de Precipitina , Conformação Proteica , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Serina/química , Treonina/química , Células Tumorais Cultivadas , eIF-2 Quinase/química , eIF-2 Quinase/genética , eIF-2 Quinase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...