Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 117(9): 5059-5066, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32041869

ABSTRACT

The radiation of angiosperms led to the emergence of the vast majority of today's plant species and all our major food crops. Their extraordinary diversification occurred in conjunction with the evolution of a more efficient vascular system for the transport of water, composed of vessel elements. The physical dimensions of these water-conducting specialized cells have played a critical role in angiosperm evolution; they determine resistance to water flow, influence photosynthesis rate, and contribute to plant stature. However, the genetic factors that determine their dimensions are unclear. Here we show that a previously uncharacterized gene, ENLARGED VESSEL ELEMENT (EVE), contributes to the dimensions of vessel elements in Populus, impacting hydraulic conductivity. Our data suggest that EVE is localized in the plasma membrane and is involved in potassium uptake of differentiating xylem cells during vessel development. In plants, EVE first emerged in streptophyte algae, but expanded dramatically among vessel-containing angiosperms. The phylogeny, structure and composition of EVE indicates that it may have been involved in an ancient horizontal gene-transfer event.


Subject(s)
Magnoliopsida/metabolism , Plant Proteins/genetics , Plant Proteins/metabolism , Populus/genetics , Populus/metabolism , Biological Evolution , Cell Membrane , Gene Expression Regulation, Developmental , Gene Expression Regulation, Plant , Photosynthesis , Phycodnaviridae , Plants, Genetically Modified , Potassium/metabolism , Water/metabolism , Xylem/cytology , Xylem/metabolism
2.
Sci Rep ; 7(1): 17996, 2017 12 21.
Article in English | MEDLINE | ID: mdl-29269936

ABSTRACT

About 50% of the world's arable land is strongly acidic (pH ≤ 5). The low pH solubilizes root-toxic ionic aluminium (Al3+) species from clay minerals, driving the evolution of counteractive adaptations in cultivated crops. The food crop Sorghum bicolor upregulates the membrane-embedded transporter protein SbMATE in its roots. SbMATE mediates efflux of the anionic form of the organic acid, citrate, into the soil rhizosphere, chelating Al3+ ions and thereby imparting Al-resistance based on excluding Al+3 from the growing root tip. Here, we use electrophysiological, radiolabeled, and fluorescence-based transport assays in two heterologous expression systems to establish a broad substrate recognition profile of SbMATE, showing the proton and/or sodium-driven transport of 14C-citrate anion, as well as the organic monovalent cation, ethidium, but not its divalent analog, propidium. We further complement our transport assays by measuring substrate binding to detergent-purified SbMATE protein. Finally, we use the purified membrane protein as an antigen to discover native conformation-binding and transport function-altering nanobodies using an animal-free, mRNA/cDNA display technology. Our results demonstrate the utility of using Pichia pastoris as an efficient eukaryotic host to express large quantities of functional plant transporter proteins. The nanobody discovery approach is applicable to other non-immunogenic plant proteins.


Subject(s)
Aluminum/metabolism , Membrane Transport Proteins/metabolism , Plant Proteins/metabolism , Sorghum/metabolism , Membrane Transport Proteins/genetics , Phylogeny , Plant Proteins/genetics , Plant Roots/metabolism , Sorghum/genetics , Substrate Specificity
3.
Acta Crystallogr D Biol Crystallogr ; 71(Pt 3): 732-41, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25760620

ABSTRACT

P-glycoprotein (P-gp) is a transporter of great clinical and pharmacological significance. Several structural studies of P-gp and its homologs have provided insights into its transport cycle, but questions remain regarding how P-gp recognizes diverse substrates and how substrate binding is coupled to ATP hydrolysis. Here, four new P-gp co-crystal structures with a series of rationally designed ligands are presented. It is observed that the binding of certain ligands, including an ATP-hydrolysis stimulator, produces a large conformational change in the fourth transmembrane helix, which is positioned to potentially transmit a signal to the nucleotide-binding domains. A new ligand-binding site on the surface of P-gp facing the inner leaflet of the membrane is also described, providing vital insights regarding the entry mechanism of hydrophobic drugs and lipids into P-gp. These results represent significant advances in the understanding of how P-gp and related transporters bind and export a plethora of metabolites, antibiotics and clinically approved and pipeline drugs.


Subject(s)
Adenosine Triphosphate/chemistry , ATP Binding Cassette Transporter, Subfamily B/chemistry , Crystallography, X-Ray , Humans , Protein Structure, Secondary , Protein Structure, Tertiary
4.
Proc Natl Acad Sci U S A ; 110(33): 13386-91, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23901103

ABSTRACT

P-glycoprotein (P-gp) is one of the best-known mediators of drug efflux-based multidrug resistance in many cancers. This validated therapeutic target is a prototypic, plasma membrane resident ATP-Binding Cassette transporter that pumps xenobiotic compounds out of cells. The large, polyspecific drug-binding pocket of P-gp recognizes a variety of structurally unrelated compounds. The transport of these drugs across the membrane is coincident with changes in the size and shape of this pocket during the course of the transport cycle. Here, we present the crystal structures of three inward-facing conformations of mouse P-gp derived from two different crystal forms. One structure has a nanobody bound to the C-terminal side of the first nucleotide-binding domain. This nanobody strongly inhibits the ATP hydrolysis activity of mouse P-gp by hindering the formation of a dimeric complex between the ATP-binding domains, which is essential for nucleotide hydrolysis. Together, these inward-facing conformational snapshots of P-gp demonstrate a range of flexibility exhibited by this transporter, which is likely an essential feature for the binding and transport of large, diverse substrates. The nanobody-bound structure also reveals a unique epitope on P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Drug Delivery Systems/methods , Models, Molecular , Protein Conformation , Animals , Epitope Mapping , Mice , Single-Domain Antibodies/chemistry
5.
Nature ; 467(7318): 991-4, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20861838

ABSTRACT

Transporter proteins from the MATE (multidrug and toxic compound extrusion) family are vital in metabolite transport in plants, directly affecting crop yields worldwide. MATE transporters also mediate multiple-drug resistance (MDR) in bacteria and mammals, modulating the efficacy of many pharmaceutical drugs used in the treatment of a variety of diseases. MATE transporters couple substrate transport to electrochemical gradients and are the only remaining class of MDR transporters whose structure has not been determined. Here we report the X-ray structure of the MATE transporter NorM from Vibrio cholerae determined to 3.65 Å, revealing an outward-facing conformation with two portals open to the outer leaflet of the membrane and a unique topology of the predicted 12 transmembrane helices distinct from any other known MDR transporter. We also report a cation-binding site in close proximity to residues previously deemed critical for transport. This conformation probably represents a stage of the transport cycle with high affinity for monovalent cations and low affinity for substrates.


Subject(s)
Antiporters/chemistry , Antiporters/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Vibrio cholerae/chemistry , Antiporters/genetics , Bacterial Proteins/genetics , Binding Sites , Cations/chemistry , Cations/metabolism , Crystallography, X-Ray , Cysteine/genetics , Cysteine/metabolism , Ion Transport , Models, Molecular , Protein Conformation , Reproducibility of Results , Static Electricity , Substrate Specificity
6.
Science ; 312(5774): 741-4, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16675700

ABSTRACT

EmrD is a multidrug transporter from the Major Facilitator Superfamily that expels amphipathic compounds across the inner membrane of Escherichia coli. Here, we report the x-ray structure of EmrD determined to a resolution of 3.5 angstroms. The structure reveals an interior that is composed mostly of hydrophobic residues, which is consistent with its role transporting amphipathic molecules. Two long loops extend into the inner leaflet side of the cell membrane. This region can serve to recognize and bind substrate directly from the lipid bilayer. We propose that multisubstrate specificity, binding, and transport are facilitated by these loop regions and the internal cavity.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/chemistry , Membrane Transport Proteins/chemistry , Amino Acid Sequence , Biological Transport , Carbonyl Cyanide m-Chlorophenyl Hydrazone/metabolism , Cell Membrane/chemistry , Crystallography, X-Ray , Cytoplasm/chemistry , Dimerization , Drug Resistance, Multiple, Bacterial , Escherichia coli/drug effects , Escherichia coli Proteins/metabolism , Hydrophobic and Hydrophilic Interactions , Lipid Bilayers , Membrane Transport Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Structure, Secondary , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL