Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
1.
Elife ; 132024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39042711

RESUMEN

Proton-coupled oligopeptide transporters (POTs) are of great pharmaceutical interest owing to their promiscuous substrate binding site that has been linked to improved oral bioavailability of several classes of drugs. Members of the POT family are conserved across all phylogenetic kingdoms and function by coupling peptide uptake to the proton electrochemical gradient. Cryo-EM structures and alphafold models have recently provided new insights into different conformational states of two mammalian POTs, SLC15A1, and SLC15A2. Nevertheless, these studies leave open important questions regarding the mechanism of proton and substrate coupling, while simultaneously providing a unique opportunity to investigate these processes using molecular dynamics (MD) simulations. Here, we employ extensive unbiased and enhanced-sampling MD to map out the full SLC15A2 conformational cycle and its thermodynamic driving forces. By computing conformational free energy landscapes in different protonation states and in the absence or presence of peptide substrate, we identify a likely sequence of intermediate protonation steps that drive inward-directed alternating access. These simulations identify key differences in the extracellular gate between mammalian and bacterial POTs, which we validate experimentally in cell-based transport assays. Our results from constant-PH MD and absolute binding free energy (ABFE) calculations also establish a mechanistic link between proton binding and peptide recognition, revealing key details underpining secondary active transport in POTs. This study provides a vital step forward in understanding proton-coupled peptide and drug transport in mammals and pave the way to integrate knowledge of solute carrier structural biology with enhanced drug design to target tissue and organ bioavailability.


The cells in our body are sealed by a surrounding membrane that allows them to control which molecules can enter or leave. Desired molecules are often imported via transport proteins that require a source of energy. One way that transporter proteins achieve this is by simultaneously moving positively charged particles called protons across the membrane. Proteins called POTs (short for proton-coupled oligopeptide transporters) use this mechanism to import small peptides and drugsin to the cells of the kidney and small intestine. Sitting in the centre of these transporters is a pocket that binds to the imported peptide which has a gate on either side: an outer gate that opens towards the outside of the cell, and an inner gate that opens towards the cell's interior. The movement of protons from the outer to the inner gate is thought to shift the shape of the transporter from an outwards to an inwards-facing state. However, the molecular details of this energetic coupling are not well understood. To explore this, Lichtinger et al. used computer simulations to pinpoint where protons bind on POTs to trigger the gates to open. The simulations proposed that two sites together make up the outward-facing gate, which opens upon proton binding. Lichtinger et al. then validated these sites experimentally in cultured human cells that produce mutant POTs. After the desired peptide/drug has attached to the binding pocket, the protons then move to two more sites further down the transporter. This triggers the inner gate to open, which ultimately allows the small molecule to move into the cell. These findings represent a significant step towards understanding how POTs transport their cargo. Since POTs can transport a range of drugs from the digestive tract into the body, these results could help researchers design molecules that are better absorbed. This could lead to more orally available medications, making it easier for patients to adhere to their treatment regimen.


Asunto(s)
Simulación de Dinámica Molecular , Protones , Animales , Conformación Proteica , Humanos , Proteínas de Transporte de Membrana/metabolismo , Proteínas de Transporte de Membrana/química , Mamíferos/metabolismo , Transporte Biológico , Termodinámica
2.
Res Sq ; 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38903084

RESUMEN

The uptake and elimination of beta-lactam antibiotics in the human body are facilitated by the proton-coupled peptide transporters PepT1 (SLC15A1) and PepT2 (SLC15A2). The mechanism by which SLC15 family transporters recognize and discriminate between different drug classes and dietary peptides remains unclear, hampering efforts to improve antibiotic pharmacokinetics through targeted drug design and delivery. Here, we present cryo-EM structures of the mammalian proton-coupled peptide transporter, PepT2, in complex with the widely used beta-lactam antibiotics cefadroxil, amoxicillin and cloxacillin. Our structures, combined with pharmacophore mapping, molecular dynamics simulations and biochemical assays, establish the mechanism of antibiotic recognition and the important role of protonation in drug binding and transport.

3.
Structure ; 32(7): 866-877.e4, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38626766

RESUMEN

Trafficking receptors control protein localization through the recognition of specific signal sequences that specify unique cellular locations. Differences in luminal pH are important for the vectorial trafficking of cargo receptors. The KDEL receptor is responsible for maintaining the integrity of the ER by retrieving luminally localized folding chaperones in a pH-dependent mechanism. Structural studies have revealed the end states of KDEL receptor activation and the mechanism of selective cargo binding. However, precisely how the KDEL receptor responds to changes in luminal pH remains unclear. To explain the mechanism of pH sensing, we combine analysis of X-ray crystal structures of the KDEL receptor at neutral and acidic pH with advanced computational methods and cell-based assays. We show a critical role for ordered water molecules that allows us to infer a direct connection between protonation in different cellular compartments and the consequent changes in the affinity of the receptor for cargo.


Asunto(s)
Receptores de Péptidos , Concentración de Iones de Hidrógeno , Humanos , Receptores de Péptidos/metabolismo , Receptores de Péptidos/química , Receptores de Péptidos/genética , Cristalografía por Rayos X , Transporte de Proteínas , Unión Proteica , Modelos Moleculares , Retículo Endoplásmico/metabolismo , Sitios de Unión
4.
Nat Struct Mol Biol ; 30(11): 1786-1793, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37482561

RESUMEN

In mammals, the kidney plays an essential role in maintaining blood homeostasis through the selective uptake, retention or elimination of toxins, drugs and metabolites. Organic anion transporters (OATs) are responsible for the recognition of metabolites and toxins in the nephron and their eventual urinary excretion. Inhibition of OATs is used therapeutically to improve drug efficacy and reduce nephrotoxicity. The founding member of the renal organic anion transporter family, OAT1 (also known as SLC22A6), uses the export of α-ketoglutarate (α-KG), a key intermediate in the Krebs cycle, to drive selective transport and is allosterically regulated by intracellular chloride. However, the mechanisms linking metabolite cycling, drug transport and intracellular chloride remain obscure. Here, we present cryogenic-electron microscopy structures of OAT1 bound to α-KG, the antiviral tenofovir and clinical inhibitor probenecid, used in the treatment of Gout. Complementary in vivo cellular assays explain the molecular basis for α-KG driven drug elimination and the allosteric regulation of organic anion transport in the kidney by chloride.


Asunto(s)
Cloruros , Proteína 1 de Transporte de Anión Orgánico , Animales , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Cloruros/metabolismo , Riñón/metabolismo , Transporte Biológico , Aniones/metabolismo , Ácidos Cetoglutáricos/metabolismo , Mamíferos/metabolismo
5.
Nat Commun ; 13(1): 4845, 2022 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-35977944

RESUMEN

Amino acid transporters play a key role controlling the flow of nutrients across the lysosomal membrane and regulating metabolism in the cell. Mutations in the gene encoding the transporter cystinosin result in cystinosis, an autosomal recessive metabolic disorder characterised by the accumulation of cystine crystals in the lysosome. Cystinosin is a member of the PQ-loop family of solute carrier (SLC) transporters and uses the proton gradient to drive cystine export into the cytoplasm. However, the molecular basis for cystinosin function remains elusive, hampering efforts to develop novel treatments for cystinosis and understand the mechanisms of ion driven transport in the PQ-loop family. To address these questions, we present the crystal structures of cystinosin from Arabidopsis thaliana in both apo and cystine bound states. Using a combination of in vitro and in vivo based assays, we establish a mechanism for cystine recognition and proton coupled transport. Mutational mapping and functional characterisation of human cystinosin further provide a framework for understanding the molecular impact of disease-causing mutations.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Cistinosis , Sistemas de Transporte de Aminoácidos Neutros/genética , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Transporte Biológico , Cistina/metabolismo , Cistinosis/genética , Humanos , Lisosomas/metabolismo , Protones
6.
Mol Cell ; 82(5): 920-932.e7, 2022 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-35245456

RESUMEN

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.


Asunto(s)
Sistema de Transporte de Aminoácidos y+ , Ferroptosis , Quinurenina , Neoplasias , Sistema de Transporte de Aminoácidos y+/genética , Sistema de Transporte de Aminoácidos y+/metabolismo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Quinurenina/metabolismo , Quinurenina/farmacología , Neoplasias/metabolismo , Transducción de Señal , Triptófano/metabolismo
7.
Nat Commun ; 12(1): 7147, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34880232

RESUMEN

Cysteine plays an essential role in cellular redox homoeostasis as a key constituent of the tripeptide glutathione (GSH). A rate limiting step in cellular GSH synthesis is the availability of cysteine. However, circulating cysteine exists in the blood as the oxidised di-peptide cystine, requiring specialised transport systems for its import into the cell. System xc- is a dedicated cystine transporter, importing cystine in exchange for intracellular glutamate. To counteract elevated levels of reactive oxygen species in cancerous cells system xc- is frequently upregulated, making it an attractive target for anticancer therapies. However, the molecular basis for ligand recognition remains elusive, hampering efforts to specifically target this transport system. Here we present the cryo-EM structure of system xc- in both the apo and glutamate bound states. Structural comparisons reveal an allosteric mechanism for ligand discrimination, supported by molecular dynamics and cell-based assays, establishing a mechanism for cystine transport in human cells.


Asunto(s)
Antiportadores/química , Antiportadores/metabolismo , Cistina/metabolismo , Ácido Glutámico/metabolismo , Glutatión/biosíntesis , Sistema de Transporte de Aminoácidos y+/química , Sistema de Transporte de Aminoácidos y+/metabolismo , Antiportadores/genética , Bioquímica , Microscopía por Crioelectrón , Cisteína/metabolismo , Cadena Pesada de la Proteína-1 Reguladora de Fusión/química , Cadena Pesada de la Proteína-1 Reguladora de Fusión/metabolismo , Células HEK293 , Humanos , Neoplasias , Oxidación-Reducción , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba
8.
Sci Adv ; 7(35)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34433568

RESUMEN

The SLC15 family of proton-coupled solute carriers PepT1 and PepT2 play a central role in human physiology as the principal route for acquiring and retaining dietary nitrogen. A remarkable feature of the SLC15 family is their extreme substrate promiscuity, which has enabled the targeting of these transporters for the improvement of oral bioavailability for several prodrug molecules. Although recent structural and biochemical studies on bacterial homologs have identified conserved sites of proton and peptide binding, the mechanism of peptide capture and ligand promiscuity remains unclear for mammalian family members. Here, we present the cryo-electron microscopy structure of the outward open conformation of the rat peptide transporter PepT2 in complex with an inhibitory nanobody. Our structure, combined with molecular dynamics simulations and biochemical and cell-based assays, establishes a framework for understanding peptide and prodrug recognition within this pharmaceutically important transporter family.


Asunto(s)
Profármacos , Simportadores , Animales , Microscopía por Crioelectrón , Mamíferos/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Transportador de Péptidos 1/química , Péptidos/metabolismo , Protones , Ratas
9.
Structure ; 29(10): 1182-1191.e4, 2021 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-34242558

RESUMEN

Tuberculosis (TB) is the leading cause of death from a single infectious agent and in 2019 an estimated 10 million people worldwide contracted the disease. Although treatments for TB exist, continual emergence of drug-resistant variants necessitates urgent development of novel antituberculars. An important new target is the lipid transporter MmpL3, which is required for construction of the unique cell envelope that shields Mycobacterium tuberculosis (Mtb) from the immune system. However, a structural understanding of the mutations in Mtb MmpL3 that confer resistance to the many preclinical leads is lacking, hampering efforts to circumvent resistance mechanisms. Here, we present the cryoelectron microscopy structure of Mtb MmpL3 and use it to comprehensively analyze the mutational landscape of drug resistance. Our data provide a rational explanation for resistance variants local to the central drug binding site, and also highlight a potential alternative route to resistance operating within the periplasmic domain.


Asunto(s)
Proteínas Bacterianas/química , Farmacorresistencia Bacteriana , Proteínas de Transporte de Membrana/química , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Microscopía por Crioelectrón , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Mutación
10.
Elife ; 102021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34137369

RESUMEN

ER proteins of widely differing abundance are retrieved from the Golgi by the KDEL-receptor. Abundant ER proteins tend to have KDEL rather than HDEL signals, whereas ADEL and DDEL are not used in most organisms. Here, we explore the mechanism of selective retrieval signal capture by the KDEL-receptor and how HDEL binds with 10-fold higher affinity than KDEL. Our results show the carboxyl-terminus of the retrieval signal moves along a ladder of arginine residues as it enters the binding pocket of the receptor. Gatekeeper residues D50 and E117 at the entrance of this pocket exclude ADEL and DDEL sequences. D50N/E117Q mutation of human KDEL-receptors changes the selectivity to ADEL and DDEL. However, further analysis of HDEL, KDEL, and RDEL-bound receptor structures shows that affinity differences are explained by interactions between the variable -4 H/K/R position of the signal and W120, rather than D50 or E117. Together, these findings explain KDEL-receptor selectivity, and how signal variants increase dynamic range to support efficient ER retrieval of low and high abundance proteins.


Asunto(s)
Retículo Endoplásmico/metabolismo , Receptores de Péptidos , Aparato de Golgi/metabolismo , Humanos , Mutación/genética , Señales de Clasificación de Proteína/genética , Transporte de Proteínas/genética , Receptores de Péptidos/química , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo
11.
Nature ; 595(7865): 130-134, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34040256

RESUMEN

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.


Asunto(s)
Microscopía por Crioelectrón , Antagonistas del Ácido Fólico/química , Antagonistas del Ácido Fólico/metabolismo , Pemetrexed/química , Pemetrexed/metabolismo , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Transporte Biológico , Humanos , Modelos Moleculares , Transportador de Folato Acoplado a Protón/ultraestructura , Protones
12.
Sci Rep ; 10(1): 21880, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33318549

RESUMEN

Now more than ever there is a demand to understand the mechanisms surrounding antibiotic resistance and look for alternative ways to impact phenotypic antibiotic outcome. Cellular energetics can be impacted by many bacteriostatic and bactericidal antibiotics, which affect metabolism and energy output, resulting in a reduction of cell growth or induction of cell death respectively. In this study, we provide evidence that a mannan rich fraction (MRF) from the cell wall of Saccharomyces cerevisiae modulates growth of antibiotic susceptible and resistant Escherichia coli and potentiates bactericidal antibiotic efficiency through modulation of bacterial cellular respiration. The role of MRF in modulating bactericidal impact and cellular metabolic state were assessed in E. coli by monitoring microbial growth and by measuring oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) using the Seahorse XFe96 Analyser, respectively. This work further illustrates the link between bacterial susceptibility to antibiotics (phenotypic resistance) and resistance through modulation of bacterial metabolism. This is the first example of yeast MRF enabling collateral sensitivity to antibiotics in vitro and supports the search for alternative strategies to promote animal health without contributing to the growing issue of antimicrobial resistance.


Asunto(s)
Antibacterianos , Pared Celular/química , Escherichia coli/crecimiento & desarrollo , Mananos , Saccharomyces cerevisiae/química , Antibacterianos/química , Antibacterianos/farmacología , Mananos/química , Mananos/farmacología
13.
Metallomics ; 12(11): 1729-1734, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-33029604

RESUMEN

The modern world has seen exposure of bacterial communities to toxic metals at selective levels. This manifests itself both intentionally, through medicines and un-intentionally through waste streams. There is growing concern that selective exposure to metals may be linked to microbial resistance to antibiotics. For a microbe to become resistant to a specific metal it must first come in contact with it. The transition metal copper has the ability to enter bacterial cells without need for a copper specific uptake mechanism. Copper is commonly used as an antimicrobial in the healthcare industry, consumer products and as a growth promoter of livestock in the agricultural sector. Here we report a study into the uptake of different organic and inorganic sources of copper. A whole-cell bacterial biosensor was developed to quantify the specific uptake of copper from various sources. Furthermore, a cell-free sensor was utilized to investigate the response to copper sources when uptake is eliminated as a factor. The data within suggest inorganic copper to have greatly reduced uptake compared to organic sources and that there is significant difference between copper oxides, Cu2O and CuO.


Asunto(s)
Técnicas Biosensibles , Cobre/análisis , Compuestos Orgánicos/análisis , Sistema Libre de Células
14.
PLoS One ; 15(6): e0234653, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32579577

RESUMEN

We previously demonstrated that hexokinase II (HK2) dissociation from mitochondria during cardiac ischemia correlates with cytochrome c (cyt-c) loss, oxidative stress and subsequent reperfusion injury. However, whether HK2 release is the primary signal mediating this ischemia-induced mitochondrial dysfunction was not established. To investigate this, we studied the effects of dissociating HK2 from isolated heart mitochondria. Mitochondria isolated from Langendorff-perfused rat hearts before and after 30 min global ischemia ± ischemic preconditioning (IPC) were subject to in vitro dissociation of HK2 by incubation with glucose-6-phosphate at pH 6.3. Prior HK2 dissociation from pre- or end-ischemic heart mitochondria had no effect on their cyt-c release, respiration (± ADP) or mitochondrial permeability transition pore (mPTP) opening. Inner mitochondrial membrane morphology was assessed indirectly by monitoring changes in light scattering (LS) and confirmed by transmission electron microscopy. Although no major ultrastructure differences were detected between pre- and end-ischemia mitochondria, the amplitude of changes in LS was reduced in the latter. This was prevented by IPC but not mimicked in vitro by HK2 dissociation. We also observed more Drp1, a mitochondrial fission protein, in end-ischemia mitochondria. IPC failed to prevent this increase but did decrease mitochondrial-associated dynamin 2. In vitro HK2 dissociation alone cannot replicate ischemia-induced effects on mitochondrial function implying that in vivo dissociation of HK2 modulates end-ischemia mitochondrial function indirectly perhaps involving interaction with mitochondrial fission proteins. The resulting changes in mitochondrial morphology and cristae structure would destabilize outer / inner membrane interactions, increase cyt-c release and enhance mPTP sensitivity to [Ca2+].


Asunto(s)
Hexoquinasa/metabolismo , Mitocondrias Cardíacas/enzimología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Isquemia Miocárdica/enzimología , Animales , Respiración de la Célula/efectos de los fármacos , Dinaminas/metabolismo , Glucosa-6-Fosfato/farmacología , Hemodinámica/efectos de los fármacos , Concentración de Iones de Hidrógeno , Precondicionamiento Isquémico , Ligandos , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/ultraestructura , Dinámicas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , Poro de Transición de la Permeabilidad Mitocondrial , Isquemia Miocárdica/patología , Unión Proteica/efectos de los fármacos , Ratas Wistar
15.
Nat Commun ; 10(1): 4657, 2019 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-31604945

RESUMEN

Nucleotide sugars are the activated form of monosaccharides used by glycosyltransferases during glycosylation. In eukaryotes the SLC35 family of solute carriers are responsible for their selective uptake into the Endoplasmic Reticulum or Golgi apparatus. The structure of the yeast GDP-mannose transporter, Vrg4, revealed a requirement for short chain lipids and a marked difference in transport rate between the nucleotide sugar and nucleoside monophosphate, suggesting a complex network of regulatory elements control transport into these organelles. Here we report the crystal structure of the GMP bound complex of Vrg4, revealing the molecular basis for GMP recognition and transport. Molecular dynamics, combined with biochemical analysis, reveal a lipid mediated dimer interface and mechanism for coordinating structural rearrangements during transport. Together these results provide further insight into how SLC35 family transporters function within the secretory pathway and sheds light onto the role that membrane lipids play in regulating transport across the membrane.


Asunto(s)
Guanosina Monofosfato/química , Membrana Dobles de Lípidos/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/metabolismo , Sitios de Unión , Guanosina Monofosfato/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Modelos Químicos , Dominios Proteicos , Proteínas de Saccharomyces cerevisiae/metabolismo , Especificidad por Sustrato
16.
Curr Opin Struct Biol ; 57: 127-134, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30999236

RESUMEN

The Golgi apparatus plays a central role in the secretory pathway as a hub for posttranslational modification, protein sorting and quality control. To date, there is little structural or biochemical information concerning the function of transporters that reside within this organelle. The SLC35 family of nucleotide sugar transporters link the synthesis of activated sugar molecules and sulfate in the cytoplasm, with the luminal transferases that catalyse their attachment to proteins and lipids during glycosylation and sulfation. A recent crystal structure of the GDP-mannose transporter has revealed key sequence motifs that direct ligand recognition and transport. Further biochemical studies unexpectedly found a requirement for short chain lipids in activating the transporter, suggesting a possible route for transport regulation within the Golgi.


Asunto(s)
Aparato de Golgi/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Nucleótidos/metabolismo , Azúcares/metabolismo , Animales , Humanos , Proteínas de Transporte de Membrana/química
17.
Science ; 363(6431): 1103-1107, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30846601

RESUMEN

Selective export and retrieval of proteins between the endoplasmic reticulum (ER) and Golgi apparatus is indispensable for eukaryotic cell function. An essential step in the retrieval of ER luminal proteins from the Golgi is the pH-dependent recognition of a carboxyl-terminal Lys-Asp-Glu-Leu (KDEL) signal by the KDEL receptor. Here, we present crystal structures of the chicken KDEL receptor in the apo ER state, KDEL-bound Golgi state, and in complex with an antagonistic synthetic nanobody (sybody). These structures show a transporter-like architecture that undergoes conformational changes upon KDEL binding and reveal a pH-dependent interaction network crucial for recognition of the carboxyl terminus of the KDEL signal. Complementary in vitro binding and in vivo cell localization data explain how these features create a pH-dependent retrieval system in the secretory pathway.


Asunto(s)
Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Receptores de Péptidos/química , Animales , Pollos , Cristalografía por Rayos X , Concentración de Iones de Hidrógeno , Ratones , Conformación Proteica , Receptores de Péptidos/metabolismo
18.
JMIR Res Protoc ; 7(4): e108, 2018 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-29691213

RESUMEN

BACKGROUND: Primary care health professionals, especially family physicians, see a variety of wounds, and yet-despite the frequency of providing wound care-many family physicians do not feel confident in wound care management. This is partly due to a lack of formal wound education in Family Medicine programs. While there are numerous electronic wound care resources available in the UK and North America, none were identified that address the specific need in supporting clinical decision-making in wound dressing selection. At the same time, healthcare providers are increasingly using technology in personal and professional contexts, and a logical extension is to use technology for knowledge translation strategies. OBJECTIVE: This work developed a prototype mobile health software application named WounDS, designed to support clinical decision-making in selecting wound dressings. This article presents the development and evaluation plan for the WounDS app. METHODS: WounDS has been developed on the iOS platform. The primary specification included ease of use, in that one of the primary influences in user adoption would be the ability to receive a wound dressing recommendation in under 30 seconds and under 5 taps on the screen. The WounDS app guides users through a series of binary decisions for assessing the wound and provides a wound dressing recommendation. The selection algorithm is based in best practices using the Wound Bed Preparation Paradigm. RESULTS: Current work is underway to examine the implementation needs for WounDS to be most effectively utilized and to pilot test its feasibility and use in clinical care. Data will be collected through user trials, focus groups, and user metadata will be collected within the app. Optimizing these preconditions will enable a subsequent phase of study to determine effects on clinical decision-making and clinical outcomes. CONCLUSIONS: WounDS is designed for knowledge translation, use of technology in clinical decision-making, and continuity of care. The benefits of WounDS include the potential to improve healthcare providers' competency in wound management and to improve wound healing through better alignment with evidence-based best practices in wound dressing selection, consistency in care from primary to community care, and subsequent downstream impacts in quality of life for patients.

19.
Nat Commun ; 9(1): 550, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416041

RESUMEN

Amino acids play essential roles in cell biology as regulators of metabolic pathways. Arginine in particular is a major signalling molecule inside the cell, being a precursor for both l-ornithine and nitric oxide (NO) synthesis and a key regulator of the mTORC1 pathway. In mammals, cellular arginine availability is determined by members of the solute carrier (SLC) 7 family of cationic amino acid transporters. Whereas CAT-1 functions to supply cationic amino acids for cellular metabolism, CAT-2A and -2B are required for macrophage activation and play important roles in regulating inflammation. Here, we present the crystal structure of a close homologue of the mammalian CAT transporters that reveals how these proteins specifically recognise arginine. Our structural and functional data provide a model for cationic amino acid transport in mammalian cells and reveals mechanistic insights into proton-coupled, sodium-independent amino acid transport in the wider APC superfamily.


Asunto(s)
Sistemas de Transporte de Aminoácidos Básicos/química , Sistemas de Transporte de Aminoácidos Básicos/metabolismo , Animales , Arginina/metabolismo , Cristalización , Cristalografía por Rayos X , Óxido Nítrico/biosíntesis , Ornitina/biosíntesis
20.
Proc Natl Acad Sci U S A ; 114(50): 13182-13187, 2017 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-29180426

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Proteínas de Transporte de Membrana/química , Péptidos/metabolismo , Protones , Proteínas Bacterianas/metabolismo , Sitios de Unión , Proteínas de Transporte de Membrana/metabolismo , Simulación de Dinámica Molecular , Unión Proteica , Xanthomonas/química , Xanthomonas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA