Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Environ Sci Technol ; 58(15): 6835-6842, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38570313

RESUMO

Artificial ion channel membranes hold high promise in water treatment, nanofluidics, and energy conversion, but it remains a great challenge to construct such smart membranes with both reversible ion-gating capability and desirable ion selectivity. Herein, we constructed a smart MXene-based membrane via p-phenylenediamine functionalization (MLM-PPD) with highly stable and aligned two-dimensional subnanochannels, which exhibits reversible ion-gating capability and ultrahigh metal ion selectivity similar to biological ion channels. The pH-sensitive groups within the MLM-PPD channel confers excellent reversible Mg2+-gating capability with a pH-switching ratio of up to 100. The mono/divalent metal-ion selectivity up to 1243.8 and 400.9 for K+/Mg2+ and Li+/Mg2+, respectively, outperforms other reported membranes. Theoretical calculations combined with experimental results reveal that the steric hindrance and stronger PPD-ion interactions substantially enhance the energy barrier for divalent metal ions passing through the MLM-PPD, and thus leading to ultrahigh mono/divalent metal-ion selectivity. This work provides a new strategy for developing artificial-ion channel membranes with both reversible ion-gating functionality and high-ion selectivity for various applications.


Assuntos
Canais Iônicos , Metais , Nitritos , Elementos de Transição , Íons , Cátions Bivalentes , Membranas Artificiais , Concentração de Íons de Hidrogênio
2.
FASEB J ; 36(8): e22423, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35775626

RESUMO

Orthodontic tooth movement (OTM) is induced by biomechanical stimuli and facilitated by periodontal tissue remodeling, where multiple immune cells participate in this progression. It has been demonstrated that macrophage is essential for mechanical force-induced tissue remodeling. In this study, we first found that mechanical force significantly induced macrophage proliferation in human periodontal samples and murine OTM models. Yet, how macrophages perceive mechanical stimuli and thereby modulate their biological behaviors remain elusive. To illustrate the mechanisms of mechanical force-induced macrophage proliferation, we subsequently identified Piezo1, a novel mechanosensory ion channel, to modulate macrophage response subjected to mechanical stimuli. Mechanical force upregulates Piezo1 expression in periodontal tissues and cultured bone-marrow-derived macrophages (BMDMs). Remarkably, suppressing Piezo1 with GsMTx4 retarded OTM through reduced macrophage proliferation. Moreover, knockdown of Piezo1 effectively inhibited mechanical force-induced BMDMs proliferation. RNA sequencing was further performed to dissect the underlying mechanisms of Piezo1-mediated mechanotransduction utilizing mechanical stretch system. We revealed that Piezo1-activated AKT/GSK3ß signaling was closely associated with macrophage proliferation upon mechanical stimuli. Importantly, Cyclin D1 (Ccnd1) was authenticated as a critical downstream factor of Piezo1 that facilitated proliferation by enhancing Rb phosphorylation. We generated genetically modified mice in which Ccnd1 could be deleted in macrophages in an inducible manner. Conditional ablation of Ccnd1 inhibited periodontal macrophage proliferation and therefore delayed OTM. Overall, our findings highlight that proliferation driven by mechanical force is a key process by which macrophages infiltrate in periodontal tissue during OTM, where Piezo1-AKT-Ccnd1 axis plays a pivotal role.


Assuntos
Ciclina D1 , Canais Iônicos , Macrófagos , Proteínas Proto-Oncogênicas c-akt , Animais , Proliferação de Células , Ciclina D1/genética , Ciclina D1/metabolismo , Humanos , Canais Iônicos/genética , Canais Iônicos/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Mecanotransdução Celular , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais
3.
Purinergic Signal ; 19(2): 421-439, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36279087

RESUMO

Of the extended family of ATP-gated P2X ion-channels, the P2X5 receptor has received comparatively little attention since first cloned over 25 years ago. Disinterest in studying this P2X subtype stems from two commonly held beliefs: (i) canonical human P2X5 is non-functional because the P2X5 subunit is truncated (hP2X5A, 422 aa) and missing the critical peptide sequence (22 aa) encoded by exon 10; (ii) rat and mouse P2X5 subunits are fully formed (455 aa) but the receptor is only weakly functional, and successive ATP responses rapidly run down in amplitude. However, newer studies have re-evaluated these notions. First, a low proportion (around 10%) of humans possess full-length P2X5 subunits (444 aa) and can form competent P2X5 receptors. Full-length P2X5 has been identified only in black Americans, but may occur in a wider population as more ethnicities are screened. Second, replacement of one of three amino acids in rat P2X5 subunits with corresponding residues in human P2X5 subunits (V67I, S191F, or F195H) significantly improves the responsiveness of rat P2X5 to ATP. Replaced residues exert an allosteric action on the left flipper, allowing the docking jaw for ATP to flex the lower body of the subunit and fully open the ion pore. This proposed action may drive the search for naturally occurring modulators which act allosterically on wildtype rat P2X5. This review collates the available information on the structure and function of human and rat P2X5 receptors, with the view to rehabilitating the reputation of these ATP-gated ion channels and stimulating future lines of research.


Assuntos
Receptores Purinérgicos P2 , Ratos , Humanos , Camundongos , Animais , Receptores Purinérgicos P2/metabolismo , Sequência de Aminoácidos , Trifosfato de Adenosina/química , Receptores Purinérgicos P2X5/metabolismo , Receptores Purinérgicos P2X2/metabolismo
4.
Proc Natl Acad Sci U S A ; 117(1): 405-414, 2020 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-31871207

RESUMO

Aided by efforts to improve their speed and efficiency, molecular dynamics (MD) simulations provide an increasingly powerful tool to study the structure-function relationship of pentameric ligand-gated ion channels (pLGICs). However, accurate reporting of the channel state and observation of allosteric regulation by agonist binding with MD remains difficult due to the timescales necessary to equilibrate pLGICs from their artificial and crystalized conformation to a more native, membrane-bound conformation in silico. Here, we perform multiple all-atom MD simulations of the homomeric 5-hydroxytryptamine 3A (5-HT3A) serotonin receptor for 15 to 20 µs to demonstrate that such timescales are critical to observe the equilibration of a pLGIC from its crystalized conformation to a membrane-bound conformation. These timescales, which are an order of magnitude longer than any previous simulation of 5-HT3A, allow us to observe the dynamic binding and unbinding of 5-hydroxytryptamine (5-HT) (i.e., serotonin) to the binding pocket located on the extracellular domain (ECD) and allosteric regulation of the transmembrane domain (TMD) from synergistic 5-HT binding. While these timescales are not long enough to observe complete activation of 5-HT3A, the allosteric regulation of ion gating elements by 5-HT binding is indicative of a preactive state, which provides insight into molecular mechanisms that regulate channel activation from a resting state. This mechanistic insight, enabled by microsecond-timescale MD simulations, will allow a careful examination of the regulation of pLGICs at a molecular level, expanding our understanding of their function and elucidating key structural motifs that can be targeted for therapeutic regulation.


Assuntos
Ativação do Canal Iônico , Simulação de Dinâmica Molecular , Receptores 5-HT3 de Serotonina/metabolismo , Serotonina/metabolismo , Regulação Alostérica , Membranas Artificiais , Domínios Proteicos , Fatores de Tempo
5.
Proc Natl Acad Sci U S A ; 117(30): 18079-18090, 2020 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-32647060

RESUMO

Ion channels in excitable cells function in macromolecular complexes in which auxiliary proteins modulate the biophysical properties of the pore-forming subunits. Hyperpolarization-activated, cyclic nucleotide-sensitive HCN4 channels are critical determinants of membrane excitability in cells throughout the body, including thalamocortical neurons and cardiac pacemaker cells. We previously showed that the properties of HCN4 channels differ dramatically in different cell types, possibly due to the endogenous expression of auxiliary proteins. Here, we report the discovery of a family of endoplasmic reticulum (ER) transmembrane proteins that associate with and modulate HCN4. Lymphoid-restricted membrane protein (LRMP, Jaw1) and inositol trisphosphate receptor-associated guanylate kinase substrate (IRAG, Mrvi1, and Jaw1L) are homologous proteins with small ER luminal domains and large cytoplasmic domains. Despite their homology, LRMP and IRAG have distinct effects on HCN4. LRMP is a loss-of-function modulator that inhibits the canonical depolarizing shift in the voltage dependence of HCN4 in response to the binding of cAMP. In contrast, IRAG causes a gain of HCN4 function by depolarizing the basal voltage dependence in the absence of cAMP. The mechanisms of action of LRMP and IRAG are independent of trafficking and cAMP binding, and they are specific to the HCN4 isoform. We also found that IRAG is highly expressed in the mouse sinoatrial node where computer modeling predicts that its presence increases HCN4 current. Our results suggest important roles for LRMP and IRAG in the regulation of cellular excitability, as tools for advancing mechanistic understanding of HCN4 channel function, and as possible scaffolds for coordination of signaling pathways.


Assuntos
Retículo Endoplasmático/metabolismo , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/metabolismo , Animais , Células CHO , Linhagem Celular , Cricetulus , AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Humanos , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/química , Canais Disparados por Nucleotídeos Cíclicos Ativados por Hiperpolarização/genética , Masculino , Potenciais da Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Modelos Biológicos , Família Multigênica , Miócitos Cardíacos/metabolismo , Fosfoproteínas/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Isoformas de Proteínas , Nó Sinoatrial/fisiologia , Nó Sinoatrial/fisiopatologia
6.
J Biol Chem ; 297(2): 100940, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34237302

RESUMO

The severe acute respiratory syndrome coronavirus 2 envelope protein (S2-E) is a conserved membrane protein that is important for coronavirus (CoV) assembly and budding. Here, we describe the recombinant expression and purification of S2-E in amphipol-class amphipathic polymer solutions, which solubilize and stabilize membrane proteins, but do not disrupt membranes. We found that amphipol delivery of S2-E to preformed planar bilayers results in spontaneous membrane integration and formation of viroporin cation channels. Amphipol delivery of the S2-E protein to human cells results in plasma membrane integration, followed by retrograde trafficking to the trans-Golgi network and accumulation in swollen perinuclear lysosomal-associated membrane protein 1-positive vesicles, likely lysosomes. CoV envelope proteins have previously been proposed to manipulate the luminal pH of the trans-Golgi network, which serves as an accumulation station for progeny CoV particles prior to cellular egress via lysosomes. Delivery of S2-E to cells will enable chemical biological approaches for future studies of severe acute respiratory syndrome coronavirus 2 pathogenesis and possibly even development of "Trojan horse" antiviral therapies. Finally, this work also establishes a paradigm for amphipol-mediated delivery of membrane proteins to cells.


Assuntos
Membrana Celular/efeitos dos fármacos , Proteínas do Envelope de Coronavírus/metabolismo , Polímeros/farmacologia , Propilaminas/farmacologia , Tensoativos/farmacologia , Rede trans-Golgi/metabolismo , Membrana Celular/metabolismo , Proteínas do Envelope de Coronavírus/genética , Células HeLa , Humanos , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Lisossomos/metabolismo , Polímeros/química , Propilaminas/química , Transporte Proteico , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Tensoativos/química
7.
Int J Mol Sci ; 23(19)2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36232854

RESUMO

Herein, we report that chromone-containing allylmorpholines can affect ion channels formed by pore-forming antibiotics in model lipid membranes, which correlates with their ability to influence membrane boundary potential and lipid-packing stress. At 100 µg/mL, allylmorpholines 1, 6, 7, and 8 decrease the boundary potential of the bilayers composed of palmitoyloleoylphosphocholine (POPC) by about 100 mV. At the same time, the compounds do not affect the zeta-potential of POPC liposomes, but reduce the membrane dipole potential by 80-120 mV. The allylmorpholine-induced drop in the dipole potential produce 10-30% enhancement in the conductance of gramicidin A channels. Chromone-containing allylmorpholines also affect the thermotropic behavior of dipalmytoylphosphocholine (DPPC), abolishing the pretransition, lowering melting cooperativity, and turning the main phase transition peak into a multicomponent profile. Compounds 4, 6, 7, and 8 are able to decrease DPPC's melting temperature by about 0.5-1.9 °C. Moreover, derivative 7 is shown to increase the temperature of transition of palmitoyloleoylphosphoethanolamine from lamellar to inverted hexagonal phase. The effects on lipid-phase transitions are attributed to the changes in the spontaneous curvature stress. Alterations in lipid packing induced by allylmorpholines are believed to potentiate the pore-forming ability of amphotericin B and gramicidin A by several times.


Assuntos
Gramicidina , Bicamadas Lipídicas , Anfotericina B , Antibacterianos , Cromonas/farmacologia , Gramicidina/metabolismo , Gramicidina/farmacologia , Canais Iônicos , Lipossomos
8.
Molecules ; 27(19)2022 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-36235195

RESUMO

The nanostructuration of solid matrices with lipid nanoparticles containing membrane proteins is a promising tool for the development of high-throughput screening devices. Here, sol-gel silica-derived nanocomposites loaded with liposome-reconstituted KcsA, a prokaryotic potassium channel, have been synthesized. The conformational and functional stability of these lipid nanoparticles before and after sol-gel immobilization have been characterized by using dynamic light scattering, and steady-state and time-resolved fluorescence spectroscopy methods. The lipid-reconstituted KcsA channel entrapped in the sol-gel matrix retained the conformational and stability changes induced by the presence of blocking or permeant cations in the buffer (associated with the conformation of the selectivity filter) or by a drop in the pH (associated with the opening of the activation gate of the protein). Hence, these results indicate that this novel device has the potential to be used as a screening platform to test new modulating drugs of potassium channels.


Assuntos
Lipossomos , Nanocompostos , Proteínas de Bactérias/metabolismo , Cátions , Canais Iônicos/metabolismo , Lipídeos , Nanopartículas , Canais de Potássio/química , Conformação Proteica , Dióxido de Silício/metabolismo
9.
Angew Chem Int Ed Engl ; 61(42): e202210214, 2022 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-36039469

RESUMO

A series of single-chain random heteropolymer (RHP)-derived artificial ion channels with both high K+ selectivity and controllable pH-gated behaviors were fabricated by a facile "one-pot" polymerization method. The benzo-18-crown-6 moieties appended on lateral chains of RHPs can form ion-permeable nanopores and transport K+ over Na+ through the lipid bilayers. The ion permeation selectivity was significantly enhanced by incorporating a cholesterol group to serve as a membrane anchor. Interestingly, similar to natural gated protein channels, on-off switchable characteristics were also realized by integrating an additional acid-sensitive alkylamine group into the RHP-derived channel. The unique design strategies have endowed the RHP-derived ion channels with facile synthetic procedures, desirable membrane compatibility, high K+ selectivity, and tunable pH-gated properties. This work provides an entry point for future design of novel functional nanochannels.


Assuntos
Bicamadas Lipídicas , Sódio , Concentração de Íons de Hidrogênio , Canais Iônicos , Polímeros
10.
J Periodontal Res ; 56(3): 471-481, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33381870

RESUMO

BACKGROUND AND OBJECTIVE: Activated potassium channels were found to be strongly correlated with gingival overgrowth (GO) phenotype as we reviewed syndromic hereditary gingival fibromatosis (HGF). Nevertheless, the functional roles of potassium channels in gingival fibrosis or gingival overgrowth remained uncovered. The aim of the present study was to explore the pathogenic role of aberrantly activated potassium channel in Hereditary Gingival Fibromatosis (HGF). METHODS: Gingival tissues were collected from 9 HGF patients and 15 normal controls. Expression of KCNQ1 was detected by immunohistochemistry. Gingival fibroblasts were isolated, and outward K+ currents were detected by whole-cell patch-clamp analysis, transmembrane potential was determined by flow cytometry. Normal human gingival fibroblasts (NHGFs) were transfected with KCNQ1 adenovirus or treated with KCNQ1 selective agonist ML277 and antagonist chromanol 293B. Accumulation of Extracellular Matrix (ECM) was measured by Western blotting and Sircol Soluble Collagen Assay. Content of secreted TGF-ß1 was measured by ELISA. Active RAS pull-down assay and cell immunofluorescence were utilized to verify RAS activation. RESULTS: KCNQ1 was upregulated in gingival tissues derived from HGF patients and HGF gingival fibroblasts presented increased outward K+ currents than NHGFs. Overexpression of KCNQ1, or KCNQ1 agonist ML277, promoted fibrotic responses of NHGFs. TGF-ß1 and KCNQ1 channels formed a positive feed-back loop. ML277 generated lateral clustering and activation of Ras on plasma membrane, followed by augmented MAPK/AP-1 signaling pathway output. JNK or ERK1/2 inhibitors suppressed ML277-induced AP-1 and ECM upregulation. CONCLUSION: Activation of KCNQ1 potassium channel promoted fibrogenic responses in NHGFs via Ras/MAPK/AP-1 signaling.


Assuntos
Fibromatose Gengival , Análise por Conglomerados , Fibroblastos , Fibromatose Gengival/genética , Gengiva , Humanos , Canal de Potássio KCNQ1/genética
11.
J Biol Chem ; 294(49): 18557-18570, 2019 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-31619519

RESUMO

The host-defense peptide (HDP) piscidin 1 (P1), isolated from the mast cells of striped bass, has potent activities against bacteria, viruses, fungi, and cancer cells and can also modulate the activity of membrane receptors. Given its broad pharmacological potential, here we used several approaches to better understand its interactions with multicomponent bilayers representing models of bacterial (phosphatidylethanolamine (PE)/phosphatidylglycerol) and mammalian (phosphatidylcholine/cholesterol (PC/Chol)) membranes. Using solid-state NMR, we solved the structure of P1 bound to PC/Chol and compared it with that of P3, a less potent homolog. The comparison disclosed that although both peptides are interfacially bound and α-helical, they differ in bilayer orientations and depths of insertion, and these differences depend on bilayer composition. Although Chol is thought to make mammalian membranes less susceptible to HDP-mediated destabilization, we found that Chol does not affect the permeabilization effects of P1. X-ray diffraction experiments revealed that both piscidins produce a demixing effect in PC/Chol membranes by increasing the fraction of the Chol-depleted phase. Furthermore, P1 increased the temperature required for the lamellar-to-hexagonal phase transition in PE bilayers, suggesting that it imposes positive membrane curvature. Patch-clamp measurements on the inner Escherichia coli membrane showed that P1 and P3, at concentrations sufficient for antimicrobial activity, substantially decrease the activating tension for bacterial mechanosensitive channels. This indicated that piscidins can cause lipid redistribution and restructuring in the microenvironment near proteins. We conclude that the mechanism of piscidin's antimicrobial activity extends beyond simple membrane destabilization, helping to rationalize its broader spectrum of pharmacological effects.


Assuntos
Peptídeos Catiônicos Antimicrobianos/química , Bicamadas Lipídicas/química , Antibacterianos/química , Colesterol/análogos & derivados , Colesterol/química , Escherichia coli/metabolismo , Glicerofosfolipídeos/química , Lipossomos/química , Espectroscopia de Ressonância Magnética , Técnicas de Patch-Clamp , Fosfatidilcolinas/química , Fosfatidilgliceróis/química
12.
J Biol Chem ; 293(18): 6969-6984, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29545310

RESUMO

Canonical K+ channels are tetrameric and highly K+-selective, whereas two-pore-domain K+ (K2P) channels form dimers, but with a similar pore architecture. A two-pore-domain potassium channel TWIK1 (KCNK1 or K2P1) allows permeation of Na+ and other monovalent ions, resulting mainly from the presence of Thr-118 in the P1 domain. However, the mechanistic basis for this reduced selectivity is unclear. Using ion-exchange-induced difference IR spectroscopy, we analyzed WT TWIK1 and T118I (highly K+-selective) and L228F (substitution in the P2 domain) TWIK1 variants and found that in the presence of K+ ions, WT and both variants exhibit an amide-I band at 1680 cm-1 This band corresponds to interactions of the backbone carbonyls in the selectivity filter with K+, a feature very similar to that of the canonical K+ channel KcsA. Computational analysis indicated that the relatively high frequency for the amide-I band is well explained by impairment of hydrogen bond formation with water molecules. Moreover, concentration-dependent spectral changes indicated that the K+ affinity of the WT selectivity filter was much lower than those of the variants. Furthermore, only the variants displayed a higher frequency shift of the 1680-cm-1 band upon changes from K+ to Rb+ or Cs+ conditions. High-speed atomic force microscopy disclosed that TWIK1's surface morphology largely does not change in K+ and Na+ solutions. Our results reveal the local conformational changes of the TWIK1 selectivity filter and suggest that the amide-I bands may be useful "molecular fingerprints" for assessing the properties of other K+ channels.


Assuntos
Canais de Potássio de Domínios Poros em Tandem/metabolismo , Potássio/metabolismo , Animais , Fenômenos Biofísicos , Cátions , Ligação de Hidrogênio , Lipossomos , Camundongos , Microscopia de Força Atômica , Simulação de Dinâmica Molecular , Canais de Potássio de Domínios Poros em Tandem/química , Conformação Proteica , Teoria Quântica , Sódio/metabolismo , Espectrofotometria Infravermelho , Espectroscopia de Infravermelho com Transformada de Fourier
13.
J Membr Biol ; 252(2-3): 173-182, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30863900

RESUMO

Planar lipid bilayers constitute a versatile method for measuring the activity of protein channels and pores on a single molecule level. Ongoing efforts attempt to tailor this method for detecting biomedically relevant target analytes or for high-throughput screening of drugs. To improve the mechanical stability of bilayer recordings, we use a thin-film epoxy resist ADEX as septum in free-standing vertical bilayers. Defined apertures with diameters between 30 µm and 100 µm were micro-fabricated by photolithography. The performance of these septa was tested by functional reconstitution of the K+ channel KcvNTS in lipid bilayers spanned over apertures in ADEX or Teflon films; the latter is conventionally used in bilayer recordings and serves as reference. We observe that the functional properties of the K+ channel are identical in both materials while ADEX provides no advantage in terms of capacitance and signal-to-noise ratio. In contrast to Teflon, however, ADEX enables long-term experimental recordings while the stability of the lipid bilayer is not compromised by pipetting solutions in and out of the recording chamber. Combined with the fact that the ADEX films can be cleaned with acetone, our results suggest that ADEX carries great potential for multiplexing bilayer chambers in robust and reusable sensing devices.


Assuntos
Resinas Epóxi/química , Bicamadas Lipídicas/química , Microtecnologia/métodos , Canais de Potássio/metabolismo , Imagem Individual de Molécula/métodos , Capacitância Elétrica , Ativação do Canal Iônico , Bicamadas Lipídicas/metabolismo , Processos Fotoquímicos , Politetrafluoretileno/química , Porosidade , Razão Sinal-Ruído , Imagem Individual de Molécula/instrumentação
14.
J Biol Chem ; 292(43): 17876-17884, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-28893910

RESUMO

The influenza A and B viruses are the primary cause of seasonal flu epidemics. Common to both viruses is the M2 protein, a homotetrameric transmembrane proton channel that acidifies the virion after endocytosis. Although influenza A M2 (AM2) and B M2 (BM2) are functional analogs, they have little sequence homology, except for a conserved HXXXW motif, which is responsible for proton selectivity and channel gating. Importantly, BM2 contains a second titratable histidine, His-27, in the tetrameric transmembrane domain that forms a reverse WXXXH motif with the gating tryptophan. To understand how His-27 affects the proton conduction property of BM2, we have used solid-state NMR to characterize the pH-dependent structure and dynamics of His-27. In cholesterol-containing lipid membranes mimicking the virus envelope, 15N NMR spectra show that the His-27 tetrad protonates with higher pKa values than His-19, indicating that the solvent-accessible His-27 facilitates proton conduction of the channel by increasing the proton dissociation rates of His-19. AM2 is inhibited by the amantadine class of antiviral drugs, whereas BM2 has no known inhibitors. We measured the N-terminal interhelical separation of the BM2 channel using fluorinated Phe-5. The interhelical 19F-19F distances show a bimodal distribution of a short distance of 7 Å and a long distance of 15-20 Å, indicating that the phenylene rings do not block small-molecule entry into the channel pore. These results give insights into the lack of amantadine inhibition of BM2 and reveal structural diversities in this family of viral proton channels.


Assuntos
Vírus da Influenza B/química , Canais Iônicos/química , Membranas Artificiais , Proteínas da Matriz Viral/química , Motivos de Aminoácidos , Vírus da Influenza B/genética , Vírus da Influenza B/metabolismo , Canais Iônicos/genética , Canais Iônicos/metabolismo , Ressonância Magnética Nuclear Biomolecular , Domínios Proteicos , Proteínas da Matriz Viral/genética , Proteínas da Matriz Viral/metabolismo
15.
Biol Pharm Bull ; 41(3): 294-302, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29491205

RESUMO

A multipass transmembrane (MTM) structure is prevalent in membrane proteins for a wide range of functions. Typically, the MTM structure is constructed of bundled multiple α-helices spanning the membrane which are connected by flexible domains. One characteristic feature of MTM proteins is dynamic functions such as stimuli responses and conformational changes. In this review, the development of synthetic molecules forming an MTM structure in membranes is highlighted. The MTM folded structure is developed using an amphiphilic molecular design with a multiblock strategy between rigid hydrophobic components and flexible hydrophilic units. Such synthetic amphiphiles not only form the MTM structure by folding but also self-assemble to construct supramolecular ion channels. An elaborated molecular design of the MTM structure with a ligand-binding pocket allows for ligand-gated regulation of ion transport. Light-triggered membrane deformation for vesicle budding is also demonstrated.


Assuntos
Biomimética , Proteínas de Membrana/química , Membranas Artificiais , Animais , Humanos , Interações Hidrofóbicas e Hidrofílicas , Canais Iônicos
16.
Nano Lett ; 17(2): 1204-1211, 2017 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-28103039

RESUMO

Here we report on the ion conductance through individual, small diameter single-walled carbon nanotubes. We find that they are mimics of ion channels found in natural systems. We explore the factors governing the ion selectivity and permeation through single-walled carbon nanotubes by considering an electrostatic mechanism built around a simplified version of the Gouy-Chapman theory. We find that the single-walled carbon nanotubes preferentially transported cations and that the cation permeability is size-dependent. The ionic conductance increases as the absolute hydration enthalpy decreases for monovalent cations with similar solid-state radii, hydrated radii, and bulk mobility. Charge screening experiments using either the addition of cationic or anionic polymers, divalent metal cations, or changes in pH reveal the enormous impact of the negatively charged carboxylates at the entrance of the single-walled carbon nanotubes. These observations were modeled in the low-to-medium concentration range (0.1-2.0 M) by an electrostatic mechanism that mimics the behavior observed in many biological ion channel-forming proteins. Moreover, multi-ion conduction in the high concentration range (>2.0 M) further reinforces the similarity between single-walled carbon nanotubes and protein ion channels.


Assuntos
Canais Iônicos/química , Nanotubos de Carbono/química , Cátions/química , Transporte de Íons , Tamanho da Partícula , Permeabilidade , Polímeros/química , Porosidade , Eletricidade Estática , Relação Estrutura-Atividade , Termodinâmica
17.
J Biol Chem ; 291(23): 12233-44, 2016 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-27056326

RESUMO

Cellular membrane disruption induced by ß-amyloid (Aß) peptides has been considered one of the major pathological mechanisms for Alzheimer disease. Mechanistic studies of the membrane disruption process at a high-resolution level, on the other hand, are hindered by the co-existence of multiple possible pathways, even in simplified model systems such as the phospholipid liposome. Therefore, separation of these pathways is crucial to achieve an in-depth understanding of the Aß-induced membrane disruption process. This study, which utilized a combination of multiple biophysical techniques, shows that the peptide-to-lipid (P:L) molar ratio is an important factor that regulates the selection of dominant membrane disruption pathways in the presence of 40-residue Aß peptides in liposomes. Three distinct pathways (fibrillation with membrane content leakage, vesicle fusion, and lipid uptake through a temporarily stable ionic channel) become dominant in model liposome systems under specific conditions. These individual systems are characterized by both the initial states of Aß peptides and the P:L molar ratio. Our results demonstrated the possibility to generate simplified Aß-membrane model systems with a homogeneous membrane disruption pathway, which will benefit high-resolution mechanistic studies in the future. Fundamentally, the possibility of pathway selection controlled by P:L suggests that the driving forces for Aß aggregation and Aß-membrane interactions may be similar at the molecular level.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Membrana Celular/metabolismo , Lipídeos de Membrana/metabolismo , Fragmentos de Peptídeos/metabolismo , Amiloide/química , Amiloide/metabolismo , Peptídeos beta-Amiloides/química , Membrana Celular/química , Dicroísmo Circular , Humanos , Canais Iônicos/química , Canais Iônicos/metabolismo , Bicamadas Lipídicas/química , Bicamadas Lipídicas/metabolismo , Lipossomos/química , Lipossomos/metabolismo , Espectroscopia de Ressonância Magnética , Lipídeos de Membrana/química , Microscopia Confocal , Fragmentos de Peptídeos/química , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Agregação Patológica de Proteínas , Ligação Proteica , Espectrometria de Fluorescência
18.
J Biol Chem ; 291(33): 17040-8, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27325695

RESUMO

Canonical volume-regulated anion channels (VRACs) are crucial for cell volume regulation and have many other important roles, including tumor drug resistance and release of neurotransmitters. Although VRAC-mediated swelling-activated chloride currents (ICl,vol) have been studied for decades, exploration of the structure-function relationship of VRAC has become possible only after the recent discovery that VRACs are formed by differently composed heteromers of LRRC8 proteins. Inactivation of ICl,vol at positive potentials, a typical hallmark of VRACs, strongly varies between native cell types. Exploiting the large differences in inactivation between different LRRC8 heteromers, we now used chimeras assembled from isoforms LRRC8C and LRRC8E to uncover a highly conserved extracellular region preceding the second LRRC8 transmembrane domain as a major determinant of ICl,vol inactivation. Point mutations identified two amino acids (Lys-98 and Asp-100 in LRRC8A and equivalent residues in LRRC8C and -E), which upon charge reversal strongly altered the kinetics and voltage dependence of inactivation. Importantly, charge reversal at the first position also reduced the iodide > chloride permeability of ICl,vol This change in selectivity was stronger when both the obligatory LRRC8A subunit and the other co-expressed isoform (LRR8C or -E) carried such mutations. Hence, the C-terminal part of the first extracellular loop not only determines VRAC inactivation but might also participate in forming its outer pore. Inactivation of VRACs may involve a closure of the extracellular mouth of the permeation pathway.


Assuntos
Proteínas de Membrana/biossíntese , Multimerização Proteica/fisiologia , Substituição de Aminoácidos , Linhagem Celular , Humanos , Proteínas de Membrana/genética , Mutação Puntual , Domínios Proteicos , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Estrutura Secundária de Proteína
19.
J Biol Chem ; 291(2): 640-51, 2016 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-26515068

RESUMO

Coumarin and its derivatives are fragrant natural compounds isolated from the genus Murraya that are flowering plants widely distributed in East Asia, Australia, and the Pacific Islands. Murraya plants have been widely used as medicinal herbs for relief of pain, such as headache, rheumatic pain, toothache, and snake bites. However, little is known about their analgesic components and the molecular mechanism underlying pain relief. Here, we report the bioassay-guided fractionation and identification of a novel coumarin derivative, named muralatin L, that can specifically activate the nociceptor transient receptor potential vanilloid 1 (TRPV1) channel and reverse the inflammatory pain in mice through channel desensitization. Muralatin L was identified from the active extract of Murraya alata against TRPV1 transiently expressed in HEK-293T cells in fluorescent calcium FlexStation assay. Activation of TRPV1 current by muralatin L and its selectivity were further confirmed by whole-cell patch clamp recordings of TRPV1-expressing HEK-293T cells and dorsal root ganglion neurons isolated from mice. Furthermore, muralatin L could reverse inflammatory pain induced by formalin and acetic acid in mice but not in TRPV1 knock-out mice. Taken together, our findings show that muralatin L specifically activates TRPV1 and reverses inflammatory pain, thus highlighting the potential of coumarin derivatives from Murraya plants for pharmaceutical and medicinal applications such as pain therapy.


Assuntos
Cumarínicos/uso terapêutico , Inflamação/tratamento farmacológico , Murraya/química , Nociceptores/metabolismo , Dor/tratamento farmacológico , Canais de Cátion TRPV/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Cálcio/metabolismo , Capsaicina/farmacologia , Capsaicina/uso terapêutico , Cumarínicos/química , Cumarínicos/farmacologia , Gânglios Espinais/patologia , Células HEK293 , Humanos , Inflamação/complicações , Ativação do Canal Iônico/efeitos dos fármacos , Espectroscopia de Ressonância Magnética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Dor/complicações , Ratos , Canais de Cátion TRPV/agonistas , Canais de Cátion TRPV/química
20.
Biochim Biophys Acta ; 1858(4): 733-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26724207

RESUMO

Viral ion channels or viroporins are short membrane proteins that participate in wide-ranging functions including virus replication and entry, assembly, and virus release. One such viroporin is the 81 amino acid residue Vpu protein derived from HIV-1. This protein consists of one transmembrane (TM) and two cytoplasmic helical domains, the former of which oligomerises to form cation-selective ion channels. In this study, we investigate the binding properties of amiloride compounds to Vpu embedded into liposomes using surface plasmon resonance (SPR). We explore the Vpu ion channel inhibitor, hexamethylene amiloride (HMA), as a molecular tool to examine the potential interactive role of key TM residues, Trp23, Ser24, and Glu29, in terms of positioning of these residues on the channel pore and the orientation of its constituent helices. The study provides experimental support that a direct interaction between Ser24 and HMA occurs and that this residue is most likely located in the channel pore. Mutation of Trp23 does not impact HMA affinity suggesting no direct involvement in binding and that this residue is lipid facing. These findings indicate that small molecules such as amilorides are capable of specifically interacting with Vpu ion channels. Although a correlation between ion channel and functional activity cannot be dismissed, alternative mechanisms involving protein-protein interactions may play an important role in the efficacy of these compounds.


Assuntos
HIV-1/química , Proteínas do Vírus da Imunodeficiência Humana/química , Lipossomos/química , Proteínas Virais Reguladoras e Acessórias/química , Amilorida/farmacologia , Sequência de Aminoácidos/efeitos dos fármacos , HIV-1/patogenicidade , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Canais Iônicos/química , Canais Iônicos/metabolismo , Mutação , Ressonância de Plasmônio de Superfície , Proteínas Virais Reguladoras e Acessórias/metabolismo , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA