Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 82(5): 920-932.e7, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35245456

RESUMO

IDO1 oxidizes tryptophan (TRP) to generate kynurenine (KYN), the substrate for 1-carbon and NAD metabolism, and is implicated in pro-cancer pathophysiology and infection biology. However, the mechanistic relationships between IDO1 in amino acid depletion versus product generation have remained a longstanding mystery. We found an unrecognized link between IDO1 and cell survival mediated by KYN that serves as the source for molecules that inhibit ferroptotic cell death. We show that this effect requires KYN export from IDO1-expressing cells, which is then available for non-IDO1-expressing cells via SLC7A11, the central transporter involved in ferroptosis suppression. Whether inside the "producer" IDO1+ cell or the "receiver" cell, KYN is converted into downstream metabolites, suppressing ferroptosis by ROS scavenging and activating an NRF2-dependent, AHR-independent cell-protective pathway, including SLC7A11, propagating anti-ferroptotic signaling. IDO1, therefore, controls a multi-pronged protection pathway from ferroptotic cell death, underscoring the need to re-evaluate the use of IDO1 inhibitors in cancer treatment.


Assuntos
Sistema y+ de Transporte de Aminoácidos , Ferroptose , Cinurenina , Neoplasias , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Cinurenina/metabolismo , Cinurenina/farmacologia , Neoplasias/metabolismo , Transdução de Sinais , Triptofano/metabolismo
2.
Nature ; 595(7865): 130-134, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34040256

RESUMO

Folates (also known as vitamin B9) have a critical role in cellular metabolism as the starting point in the synthesis of nucleic acids, amino acids and the universal methylating agent S-adenylsmethionine1,2. Folate deficiency is associated with a number of developmental, immune and neurological disorders3-5. Mammals cannot synthesize folates de novo; several systems have therefore evolved to take up folates from the diet and distribute them within the body3,6. The proton-coupled folate transporter (PCFT) (also known as SLC46A1) mediates folate uptake across the intestinal brush border membrane and the choroid plexus4,7, and is an important route for the delivery of antifolate drugs in cancer chemotherapy8-10. How PCFT recognizes folates or antifolate agents is currently unclear. Here we present cryo-electron microscopy structures of PCFT in a substrate-free state and in complex with a new-generation antifolate drug (pemetrexed). Our results provide a structural basis for understanding antifolate recognition and provide insights into the pH-regulated mechanism of folate transport mediated by PCFT.


Assuntos
Microscopia Crioeletrônica , Antagonistas do Ácido Fólico/química , Antagonistas do Ácido Fólico/metabolismo , Pemetrexede/química , Pemetrexede/metabolismo , Transportador de Folato Acoplado a Próton/química , Transportador de Folato Acoplado a Próton/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestrutura , Transporte Biológico , Humanos , Modelos Moleculares , Transportador de Folato Acoplado a Próton/ultraestrutura , Prótons
3.
Am J Hum Genet ; 107(5): 989-999, 2020 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-33053334

RESUMO

Osteogenesis imperfecta (OI) is characterized primarily by susceptibility to fractures with or without bone deformation. OI is genetically heterogeneous: over 20 genetic causes are recognized. We identified bi-allelic pathogenic KDELR2 variants as a cause of OI in four families. KDELR2 encodes KDEL endoplasmic reticulum protein retention receptor 2, which recycles ER-resident proteins with a KDEL-like peptide from the cis-Golgi to the ER through COPI retrograde transport. Analysis of patient primary fibroblasts showed intracellular decrease of HSP47 and FKBP65 along with reduced procollagen type I in culture media. Electron microscopy identified an abnormal quality of secreted collagen fibrils with increased amount of HSP47 bound to monomeric and multimeric collagen molecules. Mapping the identified KDELR2 variants onto the crystal structure of G. gallus KDELR2 indicated that these lead to an inactive receptor resulting in impaired KDELR2-mediated Golgi-ER transport. Therefore, in KDELR2-deficient individuals, OI most likely occurs because of the inability of HSP47 to bind KDELR2 and dissociate from collagen type I. Instead, HSP47 remains bound to collagen molecules extracellularly, disrupting fiber formation. This highlights the importance of intracellular recycling of ER-resident molecular chaperones for collagen type I and bone metabolism and a crucial role of HSP47 in the KDELR2-associated pathogenic mechanism leading to OI.


Assuntos
Osso e Ossos/metabolismo , Colágeno Tipo I/metabolismo , Proteínas de Choque Térmico HSP47/metabolismo , Osteogênese Imperfeita/genética , Proteínas de Transporte Vesicular/metabolismo , Adulto , Alelos , Sequência de Aminoácidos , Animais , Sítios de Ligação , Osso e Ossos/patologia , Galinhas , Pré-Escolar , Colágeno Tipo I/química , Colágeno Tipo I/genética , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia , Proteínas de Choque Térmico HSP47/química , Proteínas de Choque Térmico HSP47/genética , Humanos , Lactente , Masculino , Osteogênese Imperfeita/diagnóstico , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Linhagem , Cultura Primária de Células , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Transporte Proteico , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/genética
4.
Nature ; 551(7681): 521-524, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-29143814

RESUMO

Glycosylation is a fundamental cellular process that, in eukaryotes, occurs in the lumen of both the Golgi apparatus and the endoplasmic reticulum. Nucleotide sugar transporters (NSTs) are an essential component of the glycosylation pathway, providing the diverse range of substrates required for the glycosyltransferases. NSTs are linked to several developmental and immune disorders in humans, and in pathogenic microbes they have an important role in virulence. How NSTs recognize and transport activated monosaccharides, however, is currently unclear. Here we present the crystal structure of an NST, the GDP-mannose transporter Vrg4, in both the substrate-free and the bound states. A hitherto unobserved requirement of short-chain lipids in activating the transporter supports a model for regulation within the highly dynamic membranes of the Golgi apparatus. Our results provide a structural basis for understanding nucleotide sugar recognition, and provide insights into the transport and regulatory mechanism of this family of intracellular transporters.


Assuntos
Complexo de Golgi/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Monossacarídeos/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/citologia , Transporte Biológico , Cristalografia por Raios X , Glicosilação , Relação Estrutura-Atividade , Especificidade por Substrato
5.
Biophys J ; 121(12): 2266-2278, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35614850

RESUMO

Proton-coupled peptide transporters (POTs) are crucial for the uptake of di- and tripeptides as well as drug and prodrug molecules in prokaryotes and eukaryotic cells. We illustrate from multiscale modeling how transmembrane proton flux couples within a POT protein to drive essential steps of the full functional cycle: 1) protonation of a glutamate on transmembrane helix 7 (TM7) opens the extracellular gate, allowing ligand entry; 2) inward proton flow induces the cytosolic release of ligand by varying the protonation state of a second conserved glutamate on TM10; 3) proton movement between TM7 and TM10 is thermodynamically driven and kinetically permissible via water proton shuttling without the participation of ligand. Our results, for the first time, give direct computational confirmation for the alternating access model of POTs, and point to a quantitative multiscale kinetic picture of the functioning protein mechanism.


Assuntos
Proteínas de Membrana Transportadoras , Prótons , Ácido Glutâmico , Ligantes , Proteínas de Membrana Transportadoras/metabolismo , Peptídeos/metabolismo
6.
J Cell Sci ; 133(19)2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-33037041

RESUMO

Protein localisation in the cell is controlled through the function of trafficking receptors, which recognise specific signal sequences and direct cargo proteins to different locations. The KDEL receptor (KDELR) was one of the first intracellular trafficking receptors identified and plays an essential role in maintaining the integrity of the early secretory pathway. The receptor recognises variants of a canonical C-terminal Lys-Asp-Glu-Leu (KDEL) signal sequence on ER-resident proteins when these escape to the Golgi, and targets these proteins to COPI- coated vesicles for retrograde transport back to the ER. The empty receptor is then recycled from the ER back to the Golgi by COPII-coated vesicles. Crystal structures of the KDELR show that it is structurally related to the PQ-loop family of transporters that are found in both pro- and eukaryotes, and shuttle sugars, amino acids and vitamins across cellular membranes. Furthermore, analogous to PQ-loop transporters, the KDELR undergoes a pH-dependent and ligand-regulated conformational cycle. Here, we propose that the striking structural similarity between the KDELR and PQ-loop transporters reveals a connection between transport and trafficking in the cell, with important implications for understanding trafficking receptor evolution and function.


Assuntos
Retículo Endoplasmático , Complexo de Golgi , Vesículas Revestidas pelo Complexo de Proteína do Envoltório , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Transporte Proteico , Proteínas/metabolismo
7.
Int J Obes (Lond) ; 46(4): 859-865, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35017713

RESUMO

BACKGROUND/OBJECTIVES: Obesity affects more than forty percent of adults over the age of sixty. Aberrant eating styles such as disinhibition have been associated with the engagement of brain networks underlying executive functioning, attentional control, and interoception. However, these effects have been exclusively studied in young samples overlooking those most at risk of obesity related harm. METHODS: Here we assessed associations between resting-state functional connectivity and disinhibited eating (using the Three Factor Eating Questionnaire) in twenty-one younger (aged 19-34 years, BMI range: 18-31) and twenty older (aged 60-73 years, BMI range: 19-32) adults matched for BMI. The Alternative Healthy Eating Index was used to quantify diet quality. RESULTS: Older, compared to younger, individuals reported lower levels of disinhibited eating, consumed a healthier diet, and had weaker connectivity in the frontoparietal (FPN) and default mode (DMN) networks. In addition, associations between functional connectivity and eating behaviour differed between the two age groups. In older adults, disinhibited eating was associated with weaker connectivity in the FPN and DMN--effects that were absent in the younger sample. Importantly, these effects could not be explained by differences in habitual diet. CONCLUSIONS: These findings point to a change in interoceptive signalling as part of the ageing process, which may contribute to behavioural changes in energy intake, and highlight the importance of studying this under researched population.


Assuntos
Encéfalo , Imageamento por Ressonância Magnética , Idoso , Encéfalo/fisiologia , Mapeamento Encefálico , Função Executiva , Comportamento Alimentar , Humanos , Obesidade
8.
Nature ; 535(7613): 517-522, 2016 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-27437577

RESUMO

Developmental signals of the Hedgehog (Hh) and Wnt families are transduced across the membrane by Frizzledclass G-protein-coupled receptors (GPCRs) composed of both a heptahelical transmembrane domain (TMD) and an extracellular cysteine-rich domain (CRD). How the large extracellular domains of GPCRs regulate signalling by the TMD is unknown. We present crystal structures of the Hh signal transducer and oncoprotein Smoothened, a GPCR that contains two distinct ligand-binding sites: one in its TMD and one in the CRD. The CRD is stacked a top the TMD, separated by an intervening wedge-like linker domain. Structure-guided mutations show that the interface between the CRD, linker domain and TMD stabilizes the inactive state of Smoothened. Unexpectedly, we find a cholesterol molecule bound to Smoothened in the CRD binding site. Mutations predicted to prevent cholesterol binding impair the ability of Smoothened to transmit native Hh signals. Binding of a clinically used antagonist, vismodegib, to the TMD induces a conformational change that is propagated to the CRD, resulting in loss of cholesterol from the CRD-linker domain-TMD interface. Our results clarify the structural mechanism by which the activity of a GPCR is controlled by ligand-regulated interactions between its extracellular and transmembrane domains.


Assuntos
Espaço Extracelular/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Anilidas/química , Anilidas/metabolismo , Anilidas/farmacologia , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Sítios de Ligação/genética , Colesterol/metabolismo , Colesterol/farmacologia , Cristalografia por Raios X , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Ligantes , Modelos Moleculares , Ligação Proteica/genética , Estabilidade Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína/efeitos dos fármacos , Estrutura Terciária de Proteína/genética , Piridinas/química , Piridinas/metabolismo , Piridinas/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened
9.
Proc Natl Acad Sci U S A ; 116(3): 804-809, 2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30602453

RESUMO

A major challenge in drug development is the optimization of intestinal absorption and cellular uptake. A successful strategy has been to develop prodrug molecules, which hijack solute carrier (SLC) transporters for active transport into the body. The proton-coupled oligopeptide transporters, PepT1 and PepT2, have been successfully targeted using this approach. Peptide transporters display a remarkable capacity to recognize a diverse library of di- and tripeptides, making them extremely promiscuous and major contributors to the pharmacokinetic profile of several important drug classes, including beta-lactam antibiotics and antiviral and antineoplastic agents. Of particular interest has been their ability to recognize amino acid and peptide-based prodrug molecules, thereby providing a rational approach to improving drug transport into the body. However, the structural basis for prodrug recognition has remained elusive. Here we present crystal structures of a prokaryotic homolog of the mammalian transporters in complex with the antiviral prodrug valacyclovir and the peptide-based photodynamic therapy agent, 5-aminolevulinic acid. The valacyclovir structure reveals that prodrug recognition is mediated through both the amino acid scaffold and the ester bond, which is commonly used to link drug molecules to the carrier's physiological ligand, whereas 5-aminolevulinic acid makes far fewer interactions compared with physiological peptides. These structures provide a unique insight into how peptide transporters interact with xenobiotic molecules and provide a template for further prodrug development.


Assuntos
Transportador 1 de Peptídeos/química , Pró-Fármacos/química , Staphylococcus hominis/química , Ácido Aminolevulínico/administração & dosagem , Antivirais/administração & dosagem , Fármacos Fotossensibilizantes/administração & dosagem , Valaciclovir/administração & dosagem
10.
Nature ; 525(7570): 548-51, 2015 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-26344196

RESUMO

To contend with hazards posed by environmental fluoride, microorganisms export this anion through F(-)-specific ion channels of the Fluc family. Since the recent discovery of Fluc channels, numerous idiosyncratic features of these proteins have been unearthed, including strong selectivity for F(-) over Cl(-) and dual-topology dimeric assembly. To understand the chemical basis for F(-) permeation and how the antiparallel subunits convene to form a F(-)-selective pore, here we solve the crystal structures of two bacterial Fluc homologues in complex with three different monobody inhibitors, with and without F(-) present, to a maximum resolution of 2.1 Å. The structures reveal a surprising 'double-barrelled' channel architecture in which two F(-) ion pathways span the membrane, and the dual-topology arrangement includes a centrally coordinated cation, most likely Na(+). F(-) selectivity is proposed to arise from the very narrow pores and an unusual anion coordination that exploits the quadrupolar edges of conserved phenylalanine rings.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Fluoretos/metabolismo , Fluoretos/farmacologia , Canais Iônicos/química , Canais Iônicos/metabolismo , Ânions/química , Ânions/metabolismo , Ânions/farmacologia , Membrana Celular/metabolismo , Cristalografia por Raios X , Fluoretos/química , Modelos Biológicos , Modelos Moleculares , Fenilalanina/metabolismo , Conformação Proteica
11.
Alcohol Alcohol ; 56(6): 737-745, 2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33754640

RESUMO

AIMS: Primary aim: to determine the efficacy of FAST (the Fast Alcohol Screening Test) for detecting harmful and dependent levels of alcohol use. Secondary aim: to compare the performance of the FAST to two short forms of the Alcohol Use Disorder Identification Test (AUDIT): the AUDIT-C and AUDIT-3. METHODS: Data from 3336 individuals in South Wales, compiled from full AUDIT datasets, were examined. AUROC analysis, alongside measures of sensitivity and specificity of the FAST, AUDIT-C and AUDIT-3 were utilized for the identification of harmful and dependent alcohol use. RESULTS: The FAST demonstrated efficacy in the identification of harmful and dependent levels of alcohol use, with superior performance to both the AUDIT-C and AUDIT-3. CONCLUSION: The present paper demonstrates the potential of the FAST as a cost- and time-effective method for appropriate screening and signposting in the stepped care model utilized by many health care and treatment services. Further studies are needed to ensure validity, both within the general population and for specific services and populations.


Assuntos
Consumo de Bebidas Alcoólicas , Alcoolismo/classificação , Alcoolismo/diagnóstico , Programas de Rastreamento/instrumentação , Gravidade do Paciente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Curva ROC , Sensibilidade e Especificidade , Inquéritos e Questionários/normas , País de Gales/epidemiologia
12.
Biochem Soc Trans ; 48(2): 337-346, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32219385

RESUMO

Solute carrier (SLC) transporters play important roles in regulating the movement of small molecules and ions across cellular membranes. In mammals, they play an important role in regulating the uptake of nutrients and vitamins from the diet, and in controlling the distribution of their metabolic intermediates within the cell. Several SLC families also play an important role in drug transport and strategies are being developed to hijack SLC transporters to control and regulate drug transport within the body. Through the addition of amino acid and peptide moieties several novel antiviral and anticancer agents have been developed that hijack the proton-coupled oligopeptide transporters, PepT1 (SCL15A1) and PepT2 (SLC15A2), for improved intestinal absorption and renal retention in the body. A major goal is to understand the rationale behind these successes and expand the library of prodrug molecules that utilise SLC transporters. Recent co-crystal structures of prokaryotic homologues of the human PepT1 and PepT2 transporters have shed important new insights into the mechanism of prodrug recognition. Here, I will review recent developments in our understanding of ligand recognition and binding promiscuity within the SLC15 family, and discuss current models for prodrug recognition.


Assuntos
Transportador 1 de Peptídeos/fisiologia , Pró-Fármacos/farmacologia , Simportadores/fisiologia , Animais , Transporte Biológico , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Oligopeptídeos/química , Transportador 1 de Peptídeos/química , Simportadores/química , Valaciclovir/farmacologia , Valganciclovir/farmacologia
13.
Nature ; 507(7490): 68-72, 2014 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-24572366

RESUMO

The NRT1/PTR family of proton-coupled transporters are responsible for nitrogen assimilation in eukaryotes and bacteria through the uptake of peptides. However, in most plant species members of this family have evolved to transport nitrate as well as additional secondary metabolites and hormones. In response to falling nitrate levels, NRT1.1 is phosphorylated on an intracellular threonine that switches the transporter from a low-affinity to high-affinity state. Here we present both the apo and nitrate-bound crystal structures of Arabidopsis thaliana NRT1.1, which together with in vitro binding and transport data identify a key role for His 356 in nitrate binding. Our data support a model whereby phosphorylation increases structural flexibility and in turn the rate of transport. Comparison with peptide transporters further reveals how the NRT1/PTR family has evolved to recognize diverse nitrogenous ligands, while maintaining elements of a conserved coupling mechanism within this superfamily of nutrient transporters.


Assuntos
Proteínas de Transporte de Ânions/química , Proteínas de Transporte de Ânions/metabolismo , Arabidopsis/química , Nitratos/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Arabidopsis/metabolismo , Cristalografia por Raios X , Histidina/química , Histidina/metabolismo , Transporte de Íons , Modelos Moleculares , Transportadores de Nitrato , Nitratos/química , Fosforilação , Fosfotreonina/metabolismo , Conformação Proteica , Prótons , Relação Estrutura-Atividade , Especificidade por Substrato
14.
Proc Natl Acad Sci U S A ; 114(50): 13182-13187, 2017 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-29180426

RESUMO

POT transporters represent an evolutionarily well-conserved family of proton-coupled transport systems in biology. An unusual feature of the family is their ability to couple the transport of chemically diverse ligands to an inwardly directed proton electrochemical gradient. For example, in mammals, fungi, and bacteria they are predominantly peptide transporters, whereas in plants the family has diverged to recognize nitrate, plant defense compounds, and hormones. Although recent structural and biochemical studies have identified conserved sites of proton binding, the mechanism through which transport is coupled to proton movement remains enigmatic. Here we show that different POT transporters operate through distinct proton-coupled mechanisms through changes in the extracellular gate. A high-resolution crystal structure reveals the presence of ordered water molecules within the peptide binding site. Multiscale molecular dynamics simulations confirm proton transport occurs through these waters via Grotthuss shuttling and reveal that proton binding to the extracellular side of the transporter facilitates a reorientation from an inward- to outward-facing state. Together these results demonstrate that within the POT family multiple mechanisms of proton coupling have likely evolved in conjunction with variation of the extracellular gate.


Assuntos
Proteínas de Bactérias/química , Proteínas de Membrana Transportadoras/química , Peptídeos/metabolismo , Prótons , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Proteínas de Membrana Transportadoras/metabolismo , Simulação de Dinâmica Molecular , Ligação Proteica , Xanthomonas/química , Xanthomonas/metabolismo
15.
Biophys J ; 117(7): 1342-1351, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31500802

RESUMO

Amino acid transport into the cell is often coupled to the proton electrochemical gradient, as found in the solute carrier 36 family of proton-coupled amino acid transporters. Although no structure of a human proton-coupled amino acid transporter exists, the crystal structure of a related homolog from bacteria, GkApcT, has recently been solved in an inward-occluded state and allows an opportunity to examine how protons are coupled to amino acid transport. Our working hypothesis is that release of the amino acid substrate is facilitated by the deprotonation of a key glutamate residue (E115) located at the bottom of the binding pocket, which forms part of the intracellular gate, allowing the protein to transition from an inward-occluded to an inward-open conformation. During unbiased molecular dynamics simulations, we observed a transition from the inward-occluded state captured in the crystal structure to a much more open state, which we consider likely to be representative of the inward-open state associated with substrate release. To explore this and the role of protons in these transitions, we have used umbrella sampling to demonstrate that the transition from inward occluded to inward open is more energetically favorable when E115 is deprotonated. That E115 is likely to be protonated in the inward-occluded state and deprotonated in the inward-open state is further confirmed via the use of absolute binding free energies. Finally, we also show, via the use of absolute binding free energy calculations, that the affinity of the protein for alanine is very similar regardless of either the conformational state or the protonation of E115, presumably reflecting the fact that all the key interactions are deep within the binding cavity. Together, our results give a detailed picture of the role of protons in driving one of the major transitions in this transporter.


Assuntos
Sistemas de Transporte de Aminoácidos/metabolismo , Prótons , Bicamadas Lipídicas/química , Modelos Moleculares , Simulação de Dinâmica Molecular , Termodinâmica
16.
Nature ; 501(7468): 573-7, 2013 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-23995679

RESUMO

Sodium/proton (Na(+)/H(+)) antiporters, located at the plasma membrane in every cell, are vital for cell homeostasis. In humans, their dysfunction has been linked to diseases, such as hypertension, heart failure and epilepsy, and they are well-established drug targets. The best understood model system for Na(+)/H(+) antiport is NhaA from Escherichia coli, for which both electron microscopy and crystal structures are available. NhaA is made up of two distinct domains: a core domain and a dimerization domain. In the NhaA crystal structure a cavity is located between the two domains, providing access to the ion-binding site from the inward-facing surface of the protein. Like many Na(+)/H(+) antiporters, the activity of NhaA is regulated by pH, only becoming active above pH 6.5, at which point a conformational change is thought to occur. The only reported NhaA crystal structure so far is of the low pH inactivated form. Here we describe the active-state structure of a Na(+)/H(+) antiporter, NapA from Thermus thermophilus, at 3 Å resolution, solved from crystals grown at pH 7.8. In the NapA structure, the core and dimerization domains are in different positions to those seen in NhaA, and a negatively charged cavity has now opened to the outside. The extracellular cavity allows access to a strictly conserved aspartate residue thought to coordinate ion binding directly, a role supported here by molecular dynamics simulations. To alternate access to this ion-binding site, however, requires a surprisingly large rotation of the core domain, some 20° against the dimerization interface. We conclude that despite their fast transport rates of up to 1,500 ions per second, Na(+)/H(+) antiporters operate by a two-domain rocking bundle model, revealing themes relevant to secondary-active transporters in general.


Assuntos
Trocadores de Sódio-Hidrogênio/química , Thermus thermophilus/química , Ácido Aspártico/química , Ácido Aspártico/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Proteínas de Escherichia coli/química , Concentração de Íons de Hidrogênio , Modelos Moleculares , Simulação de Dinâmica Molecular , Multimerização Proteica , Estrutura Terciária de Proteína , Prótons , Sódio/metabolismo , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , Eletricidade Estática , Thermus thermophilus/genética
17.
Mol Microbiol ; 103(1): 117-133, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27696579

RESUMO

Nitrate and nitrite transport across biological membranes is often facilitated by protein transporters that are members of the major facilitator superfamily. Paracoccus denitrificans contains an unusual arrangement whereby two of these transporters, NarK1 and NarK2, are fused into a single protein, NarK, which delivers nitrate to the respiratory nitrate reductase and transfers the product, nitrite, to the periplasm. Our complementation studies, using a mutant lacking the nitrate/proton symporter NasA from the assimilatory nitrate reductase pathway, support that NarK1 functions as a nitrate/proton symporter while NarK2 is a nitrate/nitrite antiporter. Through the same experimental system, we find that Escherichia coli NarK and NarU can complement deletions in both narK and nasA in P. denitrificans, suggesting that, while these proteins are most likely nitrate/nitrite antiporters, they can also act in the net uptake of nitrate. Finally, we argue that primary sequence analysis and structural modelling do not readily explain why NasA, NarK1 and NarK2, as well as other transporters from this protein family, have such different functions, ranging from net nitrate uptake to nitrate/nitrite exchange.


Assuntos
Proteínas de Transporte de Ânions/metabolismo , Paracoccus denitrificans/metabolismo , Proteínas de Bactérias/metabolismo , Transporte Biológico , Escherichia coli/metabolismo , Teste de Complementação Genética , Transporte de Íons , Nitrato Redutase/metabolismo , Transportadores de Nitrato , Nitratos/metabolismo , Nitrito Redutases/metabolismo , Nitritos/metabolismo
18.
Exp Brain Res ; 236(1): 83-97, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29098314

RESUMO

Transcranial direct current stimulation (tDCS) is a non-invasive form of brain stimulation, which allows for selective inhibition or excitation of neural structures. It has demonstrated some efficacy in the treatment of mood disorders. However, these studies have predominately focused on stimulation of the prefrontal cortex (PFC). The cerebellum has an increasingly recognized role in emotional control, affective state, and some psychopathologies. As such, tDCS research into mood modulation needs to expand beyond conventional PFC-focused paradigms. Using a contralateral stimulation electrode placement [anodal left dorsolateral(dl)PFC, cathodal right cerebellum], and a single-blind, repeated-measures design, we initially assessed changes in the mood of healthy participants in response to acute stimulation (n = 44) and three repeated stimulations delivered second-daily (n = 21). In a second experiment, we separately investigated the influence of reversed polarity upon these same measures, in response to acute stimulation (n = 23) and repeated stimulation (n = 11). We observed a systematic elevation of mood in both active conditions following single and repeated tDCS, the latter of which displayed a progressive elevation of mood from baseline. No mood change was noted in response to either single or repeated stimulation in the sham condition. Frontocerebellar tDCS stimulation advantageously influences mood in healthy participants, with an accumulative and potentiated effect following successive stimulations. The possibility that frontocerebellar stimulation may provide a novel therapeutic adjunctive or pre-emptive intervention in stress-related disorders and mood-related psychopathologies should be considered.


Assuntos
Afeto/fisiologia , Cerebelo/fisiologia , Córtex Pré-Frontal/fisiologia , Estimulação Transcraniana por Corrente Contínua/métodos , Adulto , Feminino , Voluntários Saudáveis , Humanos , Masculino , Método Simples-Cego , Adulto Jovem
19.
Biochim Biophys Acta ; 1850(3): 488-99, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24859687

RESUMO

BACKGROUND: Cellular uptake of small peptides is an important physiological process mediated by the PTR family of proton-coupled peptide transporters. In bacteria peptides can be used as a source of amino acids and nitrogen. Similarly in humans peptide transport is the principle route for the uptake and retention of dietary protein in the form of short di- and tri-peptides for cellular metabolism. SCOPE OF THE REVIEW: Recent crystal structures of bacterial PTR family transporters, combined with biochemical studies of transport have revealed key molecular details underpinning ligand promiscuity and the mechanism of proton-coupled transport within the family. MAJOR CONCLUSIONS: Pairs of salt bridge interactions between transmembrane helices work in tandem to orchestrate alternating access transport within the PTR family. Key roles for residues conserved between bacterial and eukaryotic homologues suggest a conserved mechanism of peptide recognition and transport that in some cases has been subtly modified in individual species. GENERAL SIGNIFICANCE: Physiological studies on PepT1 and PepT2, the mammalian members of this family, have identified these transporters as being responsible for the uptake of many pharmaceutically important drug molecules, including antibiotics and antiviral medications and demonstrated their promiscuity can be used for improving the oral bioavailability of poorly absorbed compounds. The insights gained from recent structural studies combined with previous physiological and biochemical analyses are rapidly advancing our understanding of this medically important transporter superfamily. This article is part of a Special Issue entitled Structural biochemistry and biophysics of membrane proteins.


Assuntos
Proteínas de Bactérias/química , Proteínas de Membrana Transportadoras/química , Oligopeptídeos/metabolismo , Oligopeptídeos/farmacocinética , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Transporte Biológico , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
20.
EMBO J ; 31(16): 3411-21, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22659829

RESUMO

Short chain peptides are actively transported across membranes as an efficient route for dietary protein absorption and for maintaining cellular homeostasis. In mammals, peptide transport occurs via PepT1 and PepT2, which belong to the proton-dependent oligopeptide transporter, or POT family. The recent crystal structure of a bacterial POT transporter confirmed that they belong to the major facilitator superfamily of secondary active transporters. Despite the functional characterization of POT family members in bacteria, fungi and mammals, a detailed model for peptide recognition and transport remains unavailable. In this study, we report the 3.3-Å resolution crystal structure and functional characterization of a POT family transporter from the bacterium Streptococcus thermophilus. Crystallized in an inward open conformation the structure identifies a hinge-like movement within the C-terminal half of the transporter that facilitates opening of an intracellular gate controlling access to a central peptide-binding site. Our associated functional data support a model for peptide transport that highlights the importance of salt bridge interactions in orchestrating alternating access within the POT family.


Assuntos
Proteínas de Membrana Transportadoras/química , Streptococcus thermophilus/enzimologia , Cristalografia por Raios X , Modelos Biológicos , Modelos Químicos , Modelos Moleculares , Conformação Proteica , Streptococcus thermophilus/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA