Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 119(44): e2208882119, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36279431

RESUMO

Transmembrane protein 175 (TMEM175) is an evolutionarily distinct lysosomal cation channel whose mutation is associated with the development of Parkinson's disease. Here, we present a cryoelectron microscopy structure and molecular simulations of TMEM175 bound to 4-aminopyridine (4-AP), the only known small-molecule inhibitor of TMEM175 and a broad K+ channel inhibitor, as well as a drug approved by the Food and Drug Administration against multiple sclerosis. The structure shows that 4-AP, whose mode of action had not been previously visualized, binds near the center of the ion conduction pathway, in the open state of the channel. Molecular dynamics simulations reveal that this binding site is near the middle of the transmembrane potential gradient, providing a rationale for the voltage-dependent dissociation of 4-AP from TMEM175. Interestingly, bound 4-AP rapidly switches between three predominant binding poses, stabilized by alternate interaction patterns dictated by the twofold symmetry of the channel. Despite this highly dynamic binding mode, bound 4-AP prevents not only ion permeation but also water flow. Together, these studies provide a framework for the rational design of novel small-molecule inhibitors of TMEM175 that might reveal the role of this channel in human lysosomal physiology both in health and disease.


Assuntos
4-Aminopiridina , Canais de Potássio , Humanos , 4-Aminopiridina/farmacologia , Canais de Potássio/metabolismo , Microscopia Crioeletrônica , Lisossomos/metabolismo , Água/metabolismo
2.
J Am Chem Soc ; 146(33): 23230-23239, 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39116214

RESUMO

TMEM175 is a lysosomal potassium and proton channel that is associated with the development of Parkinson's disease. Advances in understanding the physiological roles of TMEM175 have been hampered by the absence of selective inhibitors, and studies involving genetic perturbations have yielded conflicting results. Here, we report the discovery and characterization of the first reported TMEM175-selective inhibitors, 2-phenylpyridin-4-ylamine (2-PPA), and AP-6. Cryo-EM structures of human TMEM175 bound by 2-PPA and AP-6 reveal that they act as pore blockers, binding at distinct sites in the pore and occluding the ion permeation pathway. Acute inhibition of TMEM175 by 2-PPA or AP-6 increases the level of lysosomal macromolecule catabolism, thereby accelerating macropinocytosis and other digestive processes. These inhibitors may serve as valuable tools to study the roles of TMEM175 in regulating lysosomal function and provide useful templates for future therapeutic development in Parkinson's disease.


Assuntos
Lisossomos , Doença de Parkinson , Humanos , Doença de Parkinson/tratamento farmacológico , Doença de Parkinson/metabolismo , Lisossomos/metabolismo , Descoberta de Drogas , Canais Iônicos/antagonistas & inibidores , Canais Iônicos/metabolismo , Canais Iônicos/química , Piridinas/química , Piridinas/farmacologia , Modelos Moleculares , Microscopia Crioeletrônica , Canais de Potássio
3.
Methods ; 76: 171-182, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25676707

RESUMO

Binding of ligands, ranging from proteins to ions, to membrane proteins is associated with absorption or release of heat that can be detected by isothermal titration calorimetry (ITC). Such measurements not only provide binding affinities but also afford direct access to thermodynamic parameters of binding--enthalpy, entropy and heat capacity. These parameters can be interpreted in a structural context, allow discrimination between different binding mechanisms and guide drug design. In this review, we introduce advantages and limitations of ITC as a methodology to study molecular interactions of membrane proteins. We further describe case studies where ITC was used to analyze thermodynamic linkage between ions and substrates in ion-coupled transporters. Similar type of linkage analysis will likely be applicable to a wide range of transporters, channels, and receptors.


Assuntos
Calorimetria/métodos , Proteínas de Membrana/química , Sítios de Ligação , Proteínas de Membrana/metabolismo , Modelos Moleculares , Ligação Proteica , Termodinâmica
4.
Sci Adv ; 8(9): eabl5508, 2022 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-35245129

RESUMO

ATP7A and ATP7B, two homologous copper-transporting P1B-type ATPases, play crucial roles in cellular copper homeostasis, and mutations cause Menkes and Wilson diseases, respectively. ATP7A/B contains a P-type ATPase core consisting of a membrane transport domain and three cytoplasmic domains, the A, P, and N domains, and a unique amino terminus comprising six consecutive metal-binding domains. Here, we present a cryo-electron microscopy structure of frog ATP7B in a copper-free state. Interacting with both the A and P domains, the metal-binding domains are poised to exert copper-dependent regulation of ATP hydrolysis coupled to transmembrane copper transport. A ring of negatively charged residues lines the cytoplasmic copper entrance that is presumably gated by a conserved basic residue sitting at the center. Within the membrane, a network of copper-coordinating ligands delineates a stepwise copper transport pathway. This work provides the first glimpse into the structure and function of ATP7 proteins and facilitates understanding of disease mechanisms and development of rational therapies.

5.
Elife ; 112022 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-35608336

RESUMO

Structures of the human lysosomal K+ channel transmembrane protein 175 (TMEM175) in open and closed states revealed a novel architecture lacking the canonical K+ selectivity filter motif present in previously known K+ channel structures. A hydrophobic constriction composed of four isoleucine residues was resolved in the pore and proposed to serve as the gate in the closed state, and to confer ion selectivity in the open state. Here, we achieve higher-resolution structures of the open and closed states and employ molecular dynamics simulations to analyze the conducting properties of the putative open state, demonstrating that it is permeable to K+ and, to a lesser degree, also Na+. Both cations must dehydrate significantly to penetrate the narrow hydrophobic constriction, but ion flow is assisted by a favorable electrostatic field generated by the protein that spans the length of the pore. The balance of these opposing energetic factors explains why permeation is feasible, and why TMEM175 is selective for K+ over Na+, despite the absence of the canonical selectivity filter. Accordingly, mutagenesis experiments reveal an exquisite sensitivity of the channel to perturbations that mitigate the constriction. Together, these data reveal a novel mechanism for selective permeation of ions by TMEM175 that is unlike that of other K+ channels.


Assuntos
Desidratação , Canais de Potássio , Humanos , Íons/metabolismo , Lisossomos/metabolismo , Simulação de Dinâmica Molecular , Potássio/metabolismo , Canais de Potássio/metabolismo , Conformação Proteica , Sódio/metabolismo
6.
Elife ; 92020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32228865

RESUMO

Transmembrane protein 175 (TMEM175) is a K+-selective ion channel expressed in lysosomal membranes, where it establishes a membrane potential essential for lysosomal function and its dysregulation is associated with the development of Parkinson's Disease. TMEM175 is evolutionarily distinct from all known channels, predicting novel ion-selectivity and gating mechanisms. Here we present cryo-EM structures of human TMEM175 in open and closed conformations, enabled by resolutions up to 2.6 Å. Human TMEM175 adopts a homodimeric architecture with a central ion-conduction pore lined by the side chains of the pore-lining helices. Conserved isoleucine residues in the center of the pore serve as the gate in the closed conformation. In the widened channel in the open conformation, these same residues establish a constriction essential for K+ selectivity. These studies reveal the mechanisms of permeation, selectivity and gating and lay the groundwork for understanding the role of TMEM175 in lysosomal function.


Assuntos
Ativação do Canal Iônico , Lisossomos/metabolismo , Canais de Potássio/metabolismo , Microscopia Crioeletrônica , Células HEK293 , Humanos , Potenciais da Membrana , Canais de Potássio/ultraestrutura , Conformação Proteica
7.
Elife ; 82019 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-30775971

RESUMO

Hypoosmotic conditions activate volume-regulated anion channels in vertebrate cells. These channels are formed by leucine-rich repeat-containing protein 8 (LRRC8) family members and contain LRRC8A in homo- or hetero-hexameric assemblies. Here, we present single-particle cryo-electron microscopy structures of Mus musculus LRRC8A in complex with the inhibitor DCPIB reconstituted in lipid nanodiscs. DCPIB plugs the channel like a cork in a bottle - binding in the extracellular selectivity filter and sterically occluding ion conduction. Constricted and expanded structures reveal coupled dilation of cytoplasmic LRRs and the channel pore, suggesting a mechanism for channel gating by internal stimuli. Conformational and symmetry differences between LRRC8A structures determined in detergent micelles and lipid bilayers related to reorganization of intersubunit lipid binding sites demonstrate a critical role for the membrane in determining channel structure. These results provide insight into LRRC8 gating and inhibition and the role of lipids in the structure of an ionic-strength sensing ion channel.


Assuntos
Lipídeos/química , Proteínas de Membrana/ultraestrutura , Conformação Proteica , Animais , Ânions/química , Tamanho Celular , Microscopia Crioeletrônica , Ciclopentanos/química , Citoplasma/química , Humanos , Indanos/química , Bicamadas Lipídicas/química , Proteínas de Membrana/química , Camundongos , Nanoestruturas/química , Concentração Osmolar
8.
Elife ; 72018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30255846

RESUMO

Many secondary active membrane transporters pump substrates against concentration gradients by coupling their uptake to symport of sodium ions. Symport requires the substrate and ions to be always transported together. Cooperative binding of the solutes is a key mechanism contributing to coupled transport in the sodium and aspartate symporter from Pyrococcus horikoshii GltPh. Here, we describe the kinetic mechanism of coupled binding for GltPh in the inward facing state. The first of the three coupled sodium ions, binds weakly and slowly, enabling the protein to accept the rest of the ions and the substrate. The last ion binds tightly, but is in rapid equilibrium with solution. Its release is required for the complex disassembly. Thus, the first ion serves to 'open the door' for the substrate, the last ion 'locks the door' once the substrate is in, and one ion contributes to both events.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/química , Pyrococcus horikoshii/química , Simportadores/química , Sistema X-AG de Transporte de Aminoácidos/genética , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Sítios de Ligação , Transporte Biológico , Íons/química , Cinética , Conformação Proteica , Pyrococcus horikoshii/genética , Sódio/química , Sódio/metabolismo , Especificidade por Substrato , Simportadores/genética
9.
Elife ; 3: e02283, 2014 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-24842876

RESUMO

Membrane transporters that clear the neurotransmitter glutamate from synapses are driven by symport of sodium ions and counter-transport of a potassium ion. Previous crystal structures of a homologous archaeal sodium and aspartate symporter showed that a dedicated transport domain carries the substrate and ions across the membrane. Here, we report new crystal structures of this homologue in ligand-free and ions-only bound outward- and inward-facing conformations. We show that after ligand release, the apo transport domain adopts a compact and occluded conformation that can traverse the membrane, completing the transport cycle. Sodium binding primes the transport domain to accept its substrate and triggers extracellular gate opening, which prevents inward domain translocation until substrate binding takes place. Furthermore, we describe a new cation-binding site ideally suited to bind a counter-transported ion. We suggest that potassium binding at this site stabilizes the translocation-competent conformation of the unloaded transport domain in mammalian homologues.DOI: http://dx.doi.org/10.7554/eLife.02283.001.


Assuntos
Sistema X-AG de Transporte de Aminoácidos/metabolismo , Sistema X-AG de Transporte de Aminoácidos/química , Sítios de Ligação , Cristalografia por Raios X , Ativação do Canal Iônico , Íons , Modelos Moleculares , Conformação Proteica , Canais de Sódio/metabolismo
10.
Nat Struct Mol Biol ; 20(5): 634-40, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23563139

RESUMO

Glutamate transporters catalyze concentrative uptake of the neurotransmitter into glial cells and neurons. Their transport cycle involves binding and release of the substrate on the extra- and intracellular sides of the plasma membranes and translocation of the substrate-binding site across the lipid bilayers. The energy of the ionic gradients, mainly sodium, fuels the cycle. Here, we used a cross-linking approach to trap a glutamate transporter homolog from Pyrococcus horikoshii in key conformational states with the substrate-binding site facing either the extracellular or the intracellular side of the membrane to study binding thermodynamics. We show that the chemical potential of sodium ions in solution is exclusively coupled to substrate binding and release, not to substrate translocation. Despite the transporter's structural symmetry, the binding mechanisms are distinct on the opposite sides of the membrane and more complex than the current models suggest.


Assuntos
Proteínas de Transporte de Glutamato da Membrana Plasmática/química , Proteínas de Transporte de Glutamato da Membrana Plasmática/metabolismo , Neurotransmissores/química , Neurotransmissores/metabolismo , Pyrococcus horikoshii/química , Pyrococcus horikoshii/metabolismo , Termodinâmica , Sítios de Ligação , Íons/química , Íons/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Sódio/química , Sódio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA