1
|
Tataru C, Peras M, Rutherford E, Dunlap K, Yin X, Chrisman BS, DeSantis TZ, Wall DP, Iwai S, David MM. Topic modeling for multi-omic integration in the human gut microbiome and implications for Autism. Sci Rep 2023; 13:11353. [PMID: 37443184 PMCID: PMC10345091 DOI: 10.1038/s41598-023-38228-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
While healthy gut microbiomes are critical to human health, pertinent microbial processes remain largely undefined, partially due to differential bias among profiling techniques. By simultaneously integrating multiple profiling methods, multi-omic analysis can define generalizable microbial processes, and is especially useful in understanding complex conditions such as Autism. Challenges with integrating heterogeneous data produced by multiple profiling methods can be overcome using Latent Dirichlet Allocation (LDA), a promising natural language processing technique that identifies topics in heterogeneous documents. In this study, we apply LDA to multi-omic microbial data (16S rRNA amplicon, shotgun metagenomic, shotgun metatranscriptomic, and untargeted metabolomic profiling) from the stool of 81 children with and without Autism. We identify topics, or microbial processes, that summarize complex phenomena occurring within gut microbial communities. We then subset stool samples by topic distribution, and identify metabolites, specifically neurotransmitter precursors and fatty acid derivatives, that differ significantly between children with and without Autism. We identify clusters of topics, deemed "cross-omic topics", which we hypothesize are representative of generalizable microbial processes observable regardless of profiling method. Interpreting topics, we find each represents a particular diet, and we heuristically label each cross-omic topic as: healthy/general function, age-associated function, transcriptional regulation, and opportunistic pathogenesis.
Collapse
Affiliation(s)
- Christine Tataru
- Department of Microbiology, Oregon State University, SW Campus Way, Corvallis, USA.
| | - Marie Peras
- Second Genome Inc, 1000 Marina Blvd, Suite 500, Brisbane, CA, 94005, USA
| | - Erica Rutherford
- Second Genome Inc, 1000 Marina Blvd, Suite 500, Brisbane, CA, 94005, USA
| | - Kaiti Dunlap
- Department of Bioengineering, Serra Mall, Stanford, USA
| | - Xiaochen Yin
- Second Genome Inc, 1000 Marina Blvd, Suite 500, Brisbane, CA, 94005, USA
| | | | - Todd Z DeSantis
- Second Genome Inc, 1000 Marina Blvd, Suite 500, Brisbane, CA, 94005, USA
| | - Dennis P Wall
- Department of Biomedical Data Science, Serra Mall, Stanford, USA
- Department of Pediatrics (Systems Medicine), Stanford, 1265 Welch Road, Stanford, USA
| | - Shoko Iwai
- Second Genome Inc, 1000 Marina Blvd, Suite 500, Brisbane, CA, 94005, USA
| | - Maude M David
- Department of Microbiology, Oregon State University, SW Campus Way, Corvallis, USA.
- School of Pharmacy, Oregon State University, SW Campus Way, Corvallis, USA.
| |
Collapse
|
2
|
Bazzone A, Tesmer L, Kurt D, Kaback HR, Fendler K, Madej MG. Investigation of sugar binding kinetics of the E. coli sugar/H + symporter XylE using solid supported membrane-based electrophysiology. J Biol Chem 2021; 298:101505. [PMID: 34929170 PMCID: PMC8784342 DOI: 10.1016/j.jbc.2021.101505] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 12/19/2022] Open
Abstract
Bacterial transporters are difficult to study using conventional electrophysiology because of their low transport rates and the small size of bacterial cells. Here, we applied solid-supported membrane–based electrophysiology to derive kinetic parameters of sugar translocation by the Escherichia coli xylose permease (XylE), including functionally relevant mutants. Many aspects of the fucose permease (FucP) and lactose permease (LacY) have also been investigated, which allow for more comprehensive conclusions regarding the mechanism of sugar translocation by transporters of the major facilitator superfamily. In all three of these symporters, we observed sugar binding and transport in real time to determine KM, Vmax, KD, and kobs values for different sugar substrates. KD and kobs values were attainable because of a conserved sugar-induced electrogenic conformational transition within these transporters. We also analyzed interactions between the residues in the available X-ray sugar/H+ symporter structures obtained with different bound sugars. We found that different sugars induce different conformational states, possibly correlating with different charge displacements in the electrophysiological assay upon sugar binding. Finally, we found that mutations in XylE altered the kinetics of glucose binding and transport, as Q175 and L297 are necessary for uncoupling H+ and d-glucose translocation. Based on the rates for the electrogenic conformational transition upon sugar binding (>300 s−1) and for sugar translocation (2 s−1 − 30 s−1 for different substrates), we propose a multiple-step mechanism and postulate an energy profile for sugar translocation. We also suggest a mechanism by which d-glucose can act as an inhibitor for XylE.
Collapse
Affiliation(s)
- Andre Bazzone
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - Laura Tesmer
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - Derya Kurt
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - H Ronald Kaback
- University of California, Department of Physiology and Department of Microbiology, Immunology, Molecular Genetics, Molecular Biology Institute in Los Angeles CA, USA
| | - Klaus Fendler
- Max-Planck-Institute of Biophysics, Department of Biophysical Chemistry in Frankfurt/M, Germany
| | - M Gregor Madej
- Institute of Biophysics and Biophysical Chemistry, Department of Structural Biology, University of Regensburg, Universitätsstr. 31, 95053 Regensburg, Germany; Institute of Biophysics, Department of Structural Biology, Saarland University, Center of Human and Molecular Biology, Building 60, 66421 Homburg, Germany
| |
Collapse
|
3
|
Damaraju VL, Aminpour M, Kuzma M, Winter P, Preto J, Tuszynski J, McEwan ABJ, Sawyer MB. Tyrosine Kinase Inhibitors Reduce Glucose Uptake by Binding to an Exofacial Site on hGLUT-1: Influence on 18 F-FDG PET Uptake. Clin Transl Sci 2020; 14:847-858. [PMID: 33278334 PMCID: PMC8212708 DOI: 10.1111/cts.12943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023] Open
Abstract
Positron emission tomography (PET) using 2‐deoxy‐2‐[18F]fluoro‐d‐glucose ([18F]FDG), a marker of energy metabolism and cell proliferation, is routinely used in the clinic to assess patient response to chemotherapy and to monitor tumor growth. Treatment with some tyrosine kinase inhibitors (TKIs) causes changes in blood glucose levels in both nondiabetic and diabetic patients. We evaluated the interaction of several classes of TKIs with human glucose transporter‐1 (hGLUT‐1) in FaDu and GIST‐1 cells by measuring [3H]2‐deoxy‐d‐glucose ([3H]2‐DG) and [3H]FDG uptake. Uptake of both was inhibited to varying extents by the TKIs, and representative TKIs from each class showed competitive inhibition of [3H]2‐DG uptake. In GIST‐1 cells, [3H]FDG uptake inhibition by temsirolimus and nilotinib was irreversible, whereas inhibition by imatinib, gefitinib, and pazopanib was reversible. Molecular modeling studies showed that TKIs form multiple hydrogen bonds with polar residues of the sugar binding site (i.e., Q161, Q282, Q283, N288, N317, and W388), and van der Waals interactions with the H‐pocket site. Our results showed interaction of TKIs with amino acid residues at the glucose binding site to inhibit glucose uptake by hGLUT‐1. We hypothesize that inhibition of hGLUT‐1 by TKIs could alter glucose levels in patients treated with TKIs, leading to hypoglycemia and fatigue, although further studies are required to evaluate roles of other SLC2 and SLC5 members. In addition, TKIs could affect tumor [18F]FDG uptake, increasingly used as a marker of tumor response. The hGLUT‐1 inhibition by TKIs may have implications for routine [18F]FDG‐PET monitoring of tumor response in patients.
Collapse
Affiliation(s)
- Vijaya L Damaraju
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Maral Aminpour
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Michelle Kuzma
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Philip Winter
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Jordane Preto
- Department of Physics, University of Alberta, Edmonton, Alberta, Canada.,DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi, Torino, Italy
| | - Jack Tuszynski
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Physics, University of Alberta, Edmonton, Alberta, Canada
| | - Alexander B J McEwan
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Michael B Sawyer
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada.,Department of Medical Oncology, Cross Cancer Institute, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
Meyer MJ, Tuerkova A, Römer S, Wenzel C, Seitz T, Gaedcke J, Oswald S, Brockmöller J, Zdrazil B, Tzvetkov MV. Differences in Metformin and Thiamine Uptake between Human and Mouse Organic Cation Transporter 1: Structural Determinants and Potential Consequences for Intrahepatic Concentrations. Drug Metab Dispos 2020; 48:1380-1392. [PMID: 33037045 DOI: 10.1124/dmd.120.000170] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/28/2020] [Indexed: 12/23/2022] Open
Abstract
The most commonly used oral antidiabetic drug, metformin, is a substrate of the hepatic uptake transporter OCT1 (gene name SLC22A1). However, OCT1 deficiency leads to more pronounced reductions of metformin concentrations in mouse than in human liver. Similarly, the effects of OCT1 deficiency on the pharmacokinetics of thiamine were reported to differ between human and mouse. Here, we compared the uptake characteristics of metformin and thiamine between human and mouse OCT1 using stably transfected human embryonic kidney 293 cells. The affinity for metformin was 4.9-fold lower in human than in mouse OCT1, resulting in a 6.5-fold lower intrinsic clearance. Therefore, the estimated liver-to-blood partition coefficient is only 3.34 in human compared with 14.4 in mouse and may contribute to higher intrahepatic concentrations in mice. Similarly, the affinity for thiamine was 9.5-fold lower in human than in mouse OCT1. Using human-mouse chimeric OCT1, we showed that simultaneous substitution of transmembrane helices TMH2 and TMH3 resulted in the reversal of affinity for metformin. Using homology modeling, we suggest several explanations, of which a different interaction of Leu155 (human TMH2) compared with Val156 (mouse TMH2) with residues in TMH3 had the strongest experimental support. In conclusion, the contribution of human OCT1 to the cellular uptake of thiamine and especially of metformin may be much lower than that of mouse OCT1. This may lead to an overestimation of the effects of OCT1 on hepatic concentrations in humans when using mouse as a model. In addition, comparative analyses of human and mouse orthologs may help reveal mechanisms of OCT1 transport. SIGNIFICANCE STATEMENT: OCT1 is a major hepatic uptake transporter of metformin and thiamine, but this study reports strong differences in the affinity for both compounds between human and mouse OCT1. Consequently, intrahepatic metformin concentrations could be much higher in mice than in humans, impacting metformin actions and representing a strong limitation of using rodent animal models for predictions of OCT1-related pharmacokinetics and efficacy in humans. Furthermore, OCT1 transmembrane helices TMH2 and TMH3 were identified to confer the observed species-specific differences in metformin affinity.
Collapse
Affiliation(s)
- Marleen J Meyer
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Alzbeta Tuerkova
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Sarah Römer
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Christoph Wenzel
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Tina Seitz
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Jochen Gaedcke
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Stefan Oswald
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Jürgen Brockmöller
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Barbara Zdrazil
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| | - Mladen V Tzvetkov
- Institute of Pharmacology, Center of Drug Absorption and Transport (C_DAT), University Medicine Greifswald, Greifswald, Germany (M.J.M., S.R., C.W., S.O., M.V.T.); Department of Pharmaceutical Chemistry, Division of Drug Design and Medicinal Chemistry, University of Vienna, Vienna, Austria (A.T., B.Z.); and Department of General, Visceral, and Pediatric Surgery (J.G.) and Institute of Clinical Pharmacology (T.S., J.B.), University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
5
|
Pereira D, McDonald BA, Croll D. The Genetic Architecture of Emerging Fungicide Resistance in Populations of a Global Wheat Pathogen. Genome Biol Evol 2020; 12:2231-2244. [PMID: 32986802 PMCID: PMC7846115 DOI: 10.1093/gbe/evaa203] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2020] [Indexed: 12/22/2022] Open
Abstract
Containing fungal diseases often depends on the application of fungicidal compounds. Fungicides can rapidly lose effectiveness due to the rise of resistant individuals in populations. However, the lack of knowledge about resistance mutations beyond known target genes challenges investigations into pathways to resistance. We used whole-genome sequencing data and association mapping to reveal the multilocus genetic architecture of fungicide resistance in a global panel of 159 isolates of Parastagonospora nodorum, an important fungal pathogen of wheat. We found significant differences in azole resistance among global field populations. The populations evolved distinctive combinations of resistance alleles which can interact when co-occurring in the same genetic background. We identified 34 significantly associated single nucleotide polymorphisms located in close proximity to genes associated with fungicide resistance in other fungi, including a major facilitator superfamily transporter. Using fungal colony growth rates and melanin production at different temperatures as fitness proxies, we found no evidence that resistance was constrained by genetic trade-offs. Our study demonstrates how genome-wide association studies of a global collection of pathogen strains can recapitulate the emergence of fungicide resistance. The distinct complement of resistance mutations found among populations illustrates how the evolutionary trajectory of fungicide adaptation can be complex and challenging to predict.
Collapse
Affiliation(s)
- Danilo Pereira
- Plant Pathology, Institute of Integrative Biology, ETH Zürich, Zürich, Switzerland
| | - Bruce A McDonald
- Plant Pathology, Institute of Integrative Biology, ETH Zürich, Zürich, Switzerland
| | - Daniel Croll
- Laboratory of Evolutionary Genetics, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| |
Collapse
|
6
|
Nuclear Ssr4 Is Required for the In Vitro and In Vivo Asexual Cycles and Global Gene Activity of Beauveria bassiana. mSystems 2020; 5:5/2/e00677-19. [PMID: 32317391 PMCID: PMC7174636 DOI: 10.1128/msystems.00677-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Ssr4 is known to serve as a cosubunit of chromatin-remodeling SWI/SNF and RSC complexes in yeasts but has not been functionally characterized in fungi. This study unveils for the first time the pleiotropic effects caused by deletion of ssr4 and its role in mediating global gene expression in a fungal insect pathogen. Our findings confirm an essential role of Ssr4 in hydrophobin biosynthesis and assembly required for growth, differentiation, and development of aerial hyphae for conidiation and conidial adhesion to insect surface and its essentiality for insect pathogenicity and virulence-related cellular events. Importantly, Ssr4 can regulate nearly one-fourth of all genes in the fungal genome in direct and indirect manners, including dozens involved in gene activity and hundreds involved in metabolism and/or transport of carbohydrates, amino acids, lipids, and/or inorganic ions. These findings highlight a significance of Ssr4 for filamentous fungal lifestyle. Ssr4 serves as a cosubunit of chromatin-remodeling SWI/SNF and RSC complexes in yeasts but remains functionally uncharacterized due to its essentiality for yeast viability. Here, we report pleiotropic effects of the deletion of the ssr4 ortholog nonessential for cell viability in Beauveria bassiana, an asexual insect mycopathogen. The deletion of ssr4 resulted in severe growth defects on different carbon/nitrogen sources, increased hyphal hydrophilicity, blocked hyphal differentiation, and 98% reduced conidiation capacity compared to a wild-type standard. The limited Δssr4 conidia featured an impaired coat with disordered or obscure hydrophobin rodlet bundles, decreased hydrophobicity, increased size, and lost insect pathogenicity via normal cuticle infection and 90% of virulence via intrahemocoel injection. The expression of genes required for hydrophobin biosynthesis and assembly of the rodlet layer was drastically repressed in more hydrophilic Δssr4 cells. Transcriptomic analysis revealed 2,517 genes differentially expressed in the Δssr4 mutant, including 1,505 downregulated genes and 1,012 upregulated genes. The proteins encoded by hundreds of repressed genes were involved in metabolism and/or transport of carbohydrates, amino acids, and lipids, inorganic ion transport and energy production or conversion, including dozens involved in DNA replication, transcription, translation, and posttranslational modifications. However, purified Ssr4 samples showed no DNA-binding activity, implying that the role of Ssr4 in genome-wide gene regulation could rely upon its acting as a cosubunit of the two complexes. These findings provide the first insight into an essential role of Ssr4 in the asexual cycle in vitro and in vivo of B. bassiana and highlights its importance for the filamentous fungal lifestyle. IMPORTANCE Ssr4 is known to serve as a cosubunit of chromatin-remodeling SWI/SNF and RSC complexes in yeasts but has not been functionally characterized in fungi. This study unveils for the first time the pleiotropic effects caused by deletion of ssr4 and its role in mediating global gene expression in a fungal insect pathogen. Our findings confirm an essential role of Ssr4 in hydrophobin biosynthesis and assembly required for growth, differentiation, and development of aerial hyphae for conidiation and conidial adhesion to insect surface and its essentiality for insect pathogenicity and virulence-related cellular events. Importantly, Ssr4 can regulate nearly one-fourth of all genes in the fungal genome in direct and indirect manners, including dozens involved in gene activity and hundreds involved in metabolism and/or transport of carbohydrates, amino acids, lipids, and/or inorganic ions. These findings highlight a significance of Ssr4 for filamentous fungal lifestyle.
Collapse
|
7
|
Nijland JG, Driessen AJM. Engineering of Pentose Transport in Saccharomyces cerevisiae for Biotechnological Applications. Front Bioeng Biotechnol 2020; 7:464. [PMID: 32064252 PMCID: PMC7000353 DOI: 10.3389/fbioe.2019.00464] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 12/19/2019] [Indexed: 01/05/2023] Open
Abstract
Lignocellulosic biomass yields after hydrolysis, besides the hexose D-glucose, D-xylose, and L-arabinose as main pentose sugars. In second generation bioethanol production utilizing the yeast Saccharomyces cerevisiae, it is critical that all three sugars are co-consumed to obtain an economically feasible and robust process. Since S. cerevisiae is unable to metabolize pentose sugars, metabolic pathway engineering has been employed to introduce the respective pathways for D-xylose and L-arabinose metabolism. However, S. cerevisiae lacks specific pentose transporters, and these sugars enter the cell with low affinity via glucose transporters of the Hxt family. Therefore, in the presence of D-glucose, utilization of D-xylose and L-arabinose is poor as the Hxt transporters prefer D-glucose. To solve this problem, heterologous expression of pentose transporters has been attempted but often with limited success due to poor expression and stability, and/or low turnover. A more successful approach is the engineering of the endogenous Hxt transporter family and evolutionary selection for D-glucose insensitive growth on pentose sugars. This has led to the identification of a critical and conserved asparagine residue in Hxt transporters that, when mutated, reduces the D-glucose affinity while leaving the D-xylose affinity mostly unaltered. Likewise, mutant Gal2 transporter have been selected supporting specific uptake of L-arabinose. In fermentation experiments, the transporter mutants support efficient uptake and consumption of pentose sugars, and even co-consumption of D-xylose and D-glucose when used at industrial concentrations. Further improvements are obtained by interfering with the post-translational inactivation of Hxt transporters at high or low D-glucose concentrations. Transporter engineering solved major limitations in pentose transport in yeast, now allowing for co-consumption of sugars that is limited only by the rates of primary metabolism. This paves the way for a more economical second-generation biofuels production process.
Collapse
Affiliation(s)
- Jeroen G Nijland
- Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology, University of Groningen, Groningen, Netherlands
| | - Arnold J M Driessen
- Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology, University of Groningen, Groningen, Netherlands
| |
Collapse
|
8
|
Liu Y, Liu B, Yang P, Wang T, Chang Z, Wang J, Wang Q, Li W, Wu J, Huang D, Jiang L, Yang B. LysR-type transcriptional regulator OvrB encoded in O island 9 drives enterohemorrhagic Escherichia coli O157:H7 virulence. Virulence 2019; 10:783-792. [PMID: 31502495 PMCID: PMC6768210 DOI: 10.1080/21505594.2019.1661721] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Enterohemorrhagic Escherichia coli (EHEC) O157:H7 (O157) is a major foodborne pathogen that causes severe illness in humans worldwide. The genome of O157 contains 177 genomic islands known as O islands (OIs), including Shiga toxin-converting phages (OI-45 and OI-93) and the locus for enterocyte effacement (LEE) pathogenicity island (OI-148). However, most genes in OIs are uncharacterized and code for unknown functions. In this study, we demonstrated, for the first time, that OI-9 encodes a novel transcriptional activator, Z0346 (named OvrB), which is required for bacterial adherence to host cells and LEE gene expression in O157. OvrB directly binds to the promoter region of LEE1 and activates the transcription of ler (encoding a master regulator of LEE genes), which in turn activates LEE1–5 genes to promote O157 adherence. Furthermore, mouse oral infection assays showed that OvrB promotes O157 colonization in the mouse intestine. Finally, OvrB is shown to be a widespread transcriptional activator of virulence genes in other enterohemorrhagic and enteropathogenic Escherichia coli serotypes. Our work significantly expands the understanding of bacterial virulence control and provides new evidence suggesting that horizontally transferred regulator genes mediate LEE gene expression.
Collapse
Affiliation(s)
- Yutao Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Bin Liu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Pan Yang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Ting Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Zhanhe Chang
- School of Biomedical Engineering, Tianjin Medical University , Tianjin , P. R. China
| | - Junyue Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Qian Wang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Wendi Li
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Jialin Wu
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Di Huang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Lingyan Jiang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| | - Bin Yang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA , Tianjin , P. R. China.,The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education , Tianjin , P. R. China
| |
Collapse
|
9
|
Shao W, Cai Q, Tong SM, Ying SH, Feng MG. Rei1-like protein regulates nutritional metabolism and transport required for the asexual cycle in vitro and in vivo of a fungal insect pathogen. Environ Microbiol 2019; 21:2772-2786. [PMID: 30932324 DOI: 10.1111/1462-2920.14616] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 03/31/2019] [Indexed: 12/12/2022]
Abstract
Rei1 is a cytoplasm-specific pre-60S subunit export factor that functions exclusively in cold-sensitive yeast growth but remains unexplored in filamentous fungi. Here, we report that Rei1-like BbRei1 is localized in both cytoplasm and nucleus and acts as a vital regulator in Beauveria bassiana. Deletion of BbRei1 resulted in delayed conidial germination, abnormally polarized germlings, severe growth defects on various carbon/nitrogen sources and reduced conidiation capacity as well as low temperature-sensitive growth. In ΔBbrei1, greatly attenuated virulence correlated with reduced activities of enzymes secreted for cuticular penetration and blocked formation of hyphal bodies in vivo essential for facilitation of host mummification. Revealed by transcriptomic analysis, 560 and 840 genes were significantly up- and down-regulated in ΔBbrei1 versus wild-type respectively, representing 13.5% of the fungal genome. Many repressed genes were involved in metabolism and transport of carbohydrates and amino acids. However, electrophoretic mobility shift assays presented no interactions of purified BbRei1 with 14 promoter DNA fragments. Conclusively, BbRei1 plays a pivotal role in gene expression and metabolism of nutrients and energy essential for the asexual cycle in vitro and in vivo of B. bassiana and functions much beyond the role for the yeast Rei1 in cold-sensitive cell growth.
Collapse
Affiliation(s)
- Wei Shao
- MOE Laboratory of Biosystems Homeostasis & Protection, Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qing Cai
- MOE Laboratory of Biosystems Homeostasis & Protection, Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Sen-Miao Tong
- MOE Laboratory of Biosystems Homeostasis & Protection, Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China.,College of Agricultural and Food Science, Zhejiang A&F University, Lin'an, Zhejiang 311300, China
| | - Sheng-Hua Ying
- MOE Laboratory of Biosystems Homeostasis & Protection, Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ming-Guang Feng
- MOE Laboratory of Biosystems Homeostasis & Protection, Institute of Microbiology, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
10
|
Abstract
Transport of solutes across biological membranes is essential for cellular life. This process is mediated by membrane transport proteins which move nutrients, waste products, certain drugs and ions into and out of cells. Secondary active transporters couple the transport of substrates against their concentration gradients with the transport of other solutes down their concentration gradients. The alternating access model of membrane transporters and the coupling mechanism of secondary active transporters are introduced in this book chapter. Structural studies have identified typical protein folds for transporters that we exemplify by the major facilitator superfamily (MFS) and LeuT folds. Finally, substrate binding and substrate translocation of the transporters LacY of the MFS and AdiC of the amino acid-polyamine-organocation (APC) superfamily are described.
Collapse
Affiliation(s)
- Patrick D Bosshart
- Swiss National Centre of Competence in Research (NCCR) TransCure, Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland
| | - Dimitrios Fotiadis
- Swiss National Centre of Competence in Research (NCCR) TransCure, Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012, Bern, Switzerland.
| |
Collapse
|
11
|
Bai X, Moraes TF, Reithmeier RAF. Structural biology of solute carrier (SLC) membrane transport proteins. Mol Membr Biol 2018; 34:1-32. [PMID: 29651895 DOI: 10.1080/09687688.2018.1448123] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The human solute carriers (SLCs) comprise over 400 different transporters, organized into 65 families ( http://slc.bioparadigms.org/ ) based on their sequence homology and transport function. SLCs are responsible for transporting extraordinarily diverse solutes across biological membranes, including inorganic ions, amino acids, lipids, sugars, neurotransmitters and drugs. Most of these membrane proteins function as coupled symporters (co-transporters) utilizing downhill ion (H+ or Na+) gradients as the driving force for the transport of substrate against its concentration gradient into cells. Other members work as antiporters (exchangers) that typically contain a single substrate-binding site with an alternating access mode of transport, while a few members exhibit channel-like properties. Dysfunction of SLCs is correlated with numerous human diseases and therefore they are potential therapeutic drug targets. In this review, we identified all of the SLC crystal structures that have been determined, most of which are from prokaryotic species. We further sorted all the SLC structures into four main groups with different protein folds and further discuss the well-characterized MFS (major facilitator superfamily) and LeuT (leucine transporter) folds. This review provides a systematic analysis of the structure, molecular basis of substrate recognition and mechanism of action in different SLC family members.
Collapse
Affiliation(s)
- Xiaoyun Bai
- a Department of Biochemistry , University of Toronto , Toronto , Canada
| | - Trevor F Moraes
- a Department of Biochemistry , University of Toronto , Toronto , Canada
| | | |
Collapse
|
12
|
Majd H, King MS, Palmer SM, Smith AC, Elbourne LDH, Paulsen IT, Sharples D, Henderson PJF, Kunji ERS. Screening of candidate substrates and coupling ions of transporters by thermostability shift assays. eLife 2018; 7:38821. [PMID: 30320551 PMCID: PMC6211832 DOI: 10.7554/elife.38821] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 10/11/2018] [Indexed: 02/02/2023] Open
Abstract
Substrates of most transport proteins have not been identified, limiting our understanding of their role in physiology and disease. Traditional identification methods use transport assays with radioactive compounds, but they are technically challenging and many compounds are unavailable in radioactive form or are prohibitively expensive, precluding large-scale trials. Here, we present a high-throughput screening method that can identify candidate substrates from libraries of unlabeled compounds. The assay is based on the principle that transport proteins recognize substrates through specific interactions, which lead to enhanced stabilization of the transporter population in thermostability shift assays. Representatives of three different transporter (super)families were tested, which differ in structure as well as transport and ion coupling mechanisms. In each case, the substrates were identified correctly from a large set of chemically related compounds, including stereo-isoforms. In some cases, stabilization by substrate binding was enhanced further by ions, providing testable hypotheses on energy coupling mechanisms.
Collapse
Affiliation(s)
- Homa Majd
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUnited Kingdom
| | - Martin S King
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUnited Kingdom
| | - Shane M Palmer
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUnited Kingdom
| | - Anthony C Smith
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUnited Kingdom
| | - Liam DH Elbourne
- Department of Molecular SciencesMacquarie UniversitySydneyAustralia
| | - Ian T Paulsen
- Department of Molecular SciencesMacquarie UniversitySydneyAustralia
| | - David Sharples
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsUnited Kingdom,School of Biomedical SciencesUniversity of LeedsLeedsUnited Kingdom
| | - Peter JF Henderson
- Astbury Centre for Structural Molecular BiologyUniversity of LeedsLeedsUnited Kingdom,School of Biomedical SciencesUniversity of LeedsLeedsUnited Kingdom
| | - Edmund RS Kunji
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
13
|
Affinity and path of binding xylopyranose unto E. coli xylose permease. Biochem Biophys Res Commun 2017; 494:202-206. [PMID: 29032199 DOI: 10.1016/j.bbrc.2017.10.053] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 10/11/2017] [Indexed: 11/20/2022]
Abstract
Glucose transporters (GLUTs), expressed in all types of human cells, are responsible for the uptake of sugars as the primary energy source for the normal functions of good cells and for the abnormal growth of cancer cells. The E. coli xylose permease (XylE), a homologue of human GLUTs, has been investigated more thoroughly than other major facilitator proteins in the current literature. In this paper, we present a molecular dynamics (MD) study of an all-atom model system to elucidate the atomistic details and the free-energy landscape along the path of binding a xylopyranose (XYP) from the extracellular space to the inside of the transporter protein XylE. From the MD simulations, the Gibbs free energy of binding was found to be -4.4kcal/mol in agreement with the experimental value of -4.7kcal/mol. The accuracy of our study is further shown in the computed hydration energy of XYP of -14.6kcal/mol in comparison with the experimental data of -15.0kcal/mol. Along the binding path, the Gibbs free energy of the XYP-XylE complex first rises from zero in the dissociated state to approximately 4 kcal/mol in the transition state (when XylE slightly increases its opening toward the extracellular side to accommodate XYP) before dropping down to -9.0 kcal/mol in the bound state. These quantitative insights indicate the fast equilibration between the bound and the unbound states of XylE and XYP. They also serve as an atomistic-dynamic corroboration of the experimental conclusion that XylE is a high-affinity sugar transporter.
Collapse
|
14
|
Veith K, Martinez Molledo M, Almeida Hernandez Y, Josts I, Nitsche J, Löw C, Tidow H. Lipid-like Peptides can Stabilize Integral Membrane Proteins for Biophysical and Structural Studies. Chembiochem 2017; 18:1735-1742. [PMID: 28603929 PMCID: PMC5601290 DOI: 10.1002/cbic.201700235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Indexed: 12/30/2022]
Abstract
A crucial bottleneck in membrane protein structural biology is the difficulty in identifying a detergent that can maintain the stability and functionality of integral membrane proteins (IMPs). Detergents are poor membrane mimics, and their common use in membrane protein crystallography may be one reason for the challenges in obtaining high-resolution crystal structures of many IMP families. Lipid-like peptides (LLPs) have detergent-like properties and have been proposed as alternatives for the solubilization of G protein-coupled receptors and other membrane proteins. Here, we systematically analyzed the stabilizing effect of LLPs on integral membrane proteins of different families. We found that LLPs could significantly stabilize detergent-solubilized IMPs in vitro. This stabilizing effect depended on the chemical nature of the LLP and the intrinsic stability of a particular IMP in the detergent. Our results suggest that screening a subset of LLPs is sufficient to stabilize a particular IMP, which can have a substantial impact on the crystallization and quality of the crystal.
Collapse
Affiliation(s)
- Katharina Veith
- The Hamburg Centre for Ultrafast ImagingDepartment of ChemistryInstitute for Biochemistry and Molecular BiologyUniversity of HamburgMartin-Luther-King-Platz 620146HamburgGermany
| | - Maria Martinez Molledo
- Centre for Structural Systems Biology (CSSB)DESY and European Molecular Biology Laboratory HamburgNotkestrasse 8522607HamburgGermany
| | - Yasser Almeida Hernandez
- The Hamburg Centre for Ultrafast ImagingDepartment of ChemistryInstitute for Biochemistry and Molecular BiologyUniversity of HamburgMartin-Luther-King-Platz 620146HamburgGermany
| | - Inokentijs Josts
- The Hamburg Centre for Ultrafast ImagingDepartment of ChemistryInstitute for Biochemistry and Molecular BiologyUniversity of HamburgMartin-Luther-King-Platz 620146HamburgGermany
| | - Julius Nitsche
- The Hamburg Centre for Ultrafast ImagingDepartment of ChemistryInstitute for Biochemistry and Molecular BiologyUniversity of HamburgMartin-Luther-King-Platz 620146HamburgGermany
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB)DESY and European Molecular Biology Laboratory HamburgNotkestrasse 8522607HamburgGermany
- Department of Medical Biochemistry and BiophysicsKarolinska InstitutetScheeles väg 217177StockholmSweden
| | - Henning Tidow
- The Hamburg Centre for Ultrafast ImagingDepartment of ChemistryInstitute for Biochemistry and Molecular BiologyUniversity of HamburgMartin-Luther-King-Platz 620146HamburgGermany
| |
Collapse
|
15
|
Li P, Gu Y, Li J, Xie L, Li X, Xie J. Mycobacterium tuberculosis Major Facilitator Superfamily Transporters. J Membr Biol 2017; 250:573-585. [DOI: 10.1007/s00232-017-9982-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/18/2017] [Indexed: 01/26/2023]
|
16
|
Hou J, Qiu C, Shen Y, Li H, Bao X. Engineering of Saccharomyces cerevisiae for the efficient co-utilization of glucose and xylose. FEMS Yeast Res 2017; 17:3861258. [DOI: 10.1093/femsyr/fox034] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/02/2017] [Indexed: 11/14/2022] Open
Affiliation(s)
- Jin Hou
- State Key Laboratory of Microbial Technology, The School of Life Science, Shandong University, Jinan, 250100, China
| | - Chenxi Qiu
- State Key Laboratory of Microbial Technology, The School of Life Science, Shandong University, Jinan, 250100, China
| | - Yu Shen
- State Key Laboratory of Microbial Technology, The School of Life Science, Shandong University, Jinan, 250100, China
| | - Hongxing Li
- State Key Laboratory of Microbial Technology, The School of Life Science, Shandong University, Jinan, 250100, China
- Shandong Provincial Key Laboratory of Microbial Engineering, Qi Lu University of Technology, Jinan, 250353, China
| | - Xiaoming Bao
- State Key Laboratory of Microbial Technology, The School of Life Science, Shandong University, Jinan, 250100, China
- Shandong Provincial Key Laboratory of Microbial Engineering, Qi Lu University of Technology, Jinan, 250353, China
| |
Collapse
|
17
|
Quistgaard EM, Martinez Molledo M, Löw C. Structure determination of a major facilitator peptide transporter: Inward facing PepTSt from Streptococcus thermophilus crystallized in space group P3121. PLoS One 2017; 12:e0173126. [PMID: 28264013 PMCID: PMC5338821 DOI: 10.1371/journal.pone.0173126] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/15/2017] [Indexed: 12/03/2022] Open
Abstract
Major facilitator superfamily (MFS) peptide transporters (typically referred to as PepT, POT or PTR transporters) mediate the uptake of di- and tripeptides, and so play an important dietary role in many organisms. In recent years, a better understanding of the molecular basis for this process has emerged, which is in large part due to a steep increase in structural information. Yet, the conformational transitions underlying the transport mechanism are still not fully understood, and additional data is therefore needed. Here we report in detail the detergent screening, crystallization, experimental MIRAS phasing, and refinement of the peptide transporter PepTSt from Streptococcus thermophilus. The space group is P3121, and the protein is crystallized in a monomeric inward facing form. The binding site is likely to be somewhat occluded, as the lobe encompassing transmembrane helices 10 and 11 is markedly bent towards the central pore of the protein, but the extent of this potential occlusion could not be determined due to disorder at the apex of the lobe. Based on structural comparisons with the seven previously determined P212121 and C2221 structures of inward facing PepTSt, the structural flexibility as well as the conformational changes mediating transition between the inward open and inward facing occluded states are discussed. In conclusion, this report improves our understanding of the structure and conformational cycle of PepTSt, and can furthermore serve as a case study, which may aid in supporting future structure determinations of additional MFS transporters or other integral membrane proteins.
Collapse
Affiliation(s)
- Esben M. Quistgaard
- Centre for Structural Systems Biology (CSSB), DESY and European Molecular Biology Laboratory Hamburg, Hamburg, Germany
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Maria Martinez Molledo
- Centre for Structural Systems Biology (CSSB), DESY and European Molecular Biology Laboratory Hamburg, Hamburg, Germany
| | - Christian Löw
- Centre for Structural Systems Biology (CSSB), DESY and European Molecular Biology Laboratory Hamburg, Hamburg, Germany
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
18
|
Mechanism of inhibition of human glucose transporter GLUT1 is conserved between cytochalasin B and phenylalanine amides. Proc Natl Acad Sci U S A 2016; 113:4711-6. [PMID: 27078104 PMCID: PMC4855560 DOI: 10.1073/pnas.1603735113] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Cancerous cells have an acutely increased demand for energy, leading to increased levels of human glucose transporter 1 (hGLUT1). This up-regulation suggests hGLUT1 as a target for therapeutic inhibitors addressing a multitude of cancer types. Here, we present three inhibitor-bound, inward-open structures of WT-hGLUT1 crystallized with three different inhibitors: cytochalasin B, a nine-membered bicyclic ring fused to a 14-membered macrocycle, which has been described extensively in the literature of hGLUTs, and two previously undescribed Phe amide-derived inhibitors. Despite very different chemical backbones, all three compounds bind in the central cavity of the inward-open state of hGLUT1, and all binding sites overlap the glucose-binding site. The inhibitory action of the compounds was determined for hGLUT family members, hGLUT1-4, using cell-based assays, and compared with homology models for these hGLUT members. This comparison uncovered a probable basis for the observed differences in inhibition between family members. We pinpoint regions of the hGLUT proteins that can be targeted to achieve isoform selectivity, and show that these same regions are used for inhibitors with very distinct structural backbones. The inhibitor cocomplex structures of hGLUT1 provide an important structural insight for the design of more selective inhibitors for hGLUTs and hGLUT1 in particular.
Collapse
|
19
|
Wang M, Yu C, Zhao H. Identification of an important motif that controls the activity and specificity of sugar transporters. Biotechnol Bioeng 2016; 113:1460-7. [PMID: 26724683 DOI: 10.1002/bit.25926] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/09/2015] [Accepted: 12/28/2015] [Indexed: 01/15/2023]
Abstract
Efficient glucose-xylose co-utilization is critical for economical biofuel production from lignocellulosic biomass. To enable glucose-xylose co-utilization, a highly active xylose specific transporter without glucose inhibition is desirable. However, our understanding of the structure-activity/specificity relationship of sugar transporters in general is limited, which hinders our ability to engineer xylose-specific transporters. In this study, via homology modeling and analysis of hexose sugar transporter HXT14 mutants, we identified a highly conserved YYX(T/P) motif that plays an important role in controlling the activity and specificity of sugar transporters. We demonstrated that mutating the two tyrosine residues of the motif to phenylalanine, respectively, improved glucose transport capacity across several different sugar transporters. Furthermore, we illustrated that by engineering the fourth position in the YYX(T/P) motif, the sugar specificity of transporters was significantly altered or even reversed towards xylose. Finally, using the engineered sugar transporter, genuine glucose-xylose co-fermentation was achieved. Biotechnol. Bioeng. 2016;113: 1460-1467. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Meng Wang
- Department of Chemical and Biomolecular Engineering, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Chenzhao Yu
- School of Molecular and Cellular Biology, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Huimin Zhao
- Department of Chemical and Biomolecular Engineering, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois. .,Department of Biochemistry, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois. .,Departments of Chemistry and Bioengineering, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana 61801, Illinois.
| |
Collapse
|
20
|
Understanding transport by the major facilitator superfamily (MFS): structures pave the way. Nat Rev Mol Cell Biol 2016; 17:123-32. [PMID: 26758938 DOI: 10.1038/nrm.2015.25] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Members of the major facilitator superfamily (MFS) of transport proteins are essential for the movement of a wide range of substrates across biomembranes. As this transport requires a series of conformational changes, structures of MFS transporters captured in different conformational states are needed to decipher the transport mechanism. Recently, a large number of MFS transporter structures have been determined, which has provided us with an unprecedented opportunity to understand general aspects of the transport mechanism. We propose an updated model for the conformational cycle of MFS transporters, the 'clamp-and-switch model', and discuss the role of so-called 'gating residues' and the substrate in modulating these conformational changes.
Collapse
|
21
|
Abstract
The ancient and ubiquitous major facilitator superfamily (MFS) represents the largest secondary transporter family and plays a crucial role in a multitude of physiological processes. MFS proteins transport a broad spectrum of ions and solutes across membranes via facilitated diffusion, symport, or antiport. In recent years, remarkable advances in understanding the structural biology of the MFS transporters have been made. This article reviews the history, classification, and general features of the MFS proteins; summarizes recent structural progress with a focus on the sugar porter family transporters exemplified by GLUT1; and discusses the molecular mechanisms of substrate binding, alternating access, and cotransport coupling.
Collapse
Affiliation(s)
- Nieng Yan
- State Key Laboratory of Bio-membrane and Membrane Biotechnology, Center for Structural Biology, School of Medicine, Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China;
| |
Collapse
|
22
|
Deng D, Yan N. GLUT, SGLT, and SWEET: Structural and mechanistic investigations of the glucose transporters. Protein Sci 2016; 25:546-58. [PMID: 26650681 DOI: 10.1002/pro.2858] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/20/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022]
Abstract
Glucose is the primary fuel to life on earth. Cellular uptake of glucose is a fundamental process for metabolism, growth, and homeostasis. Three families of secondary glucose transporters have been identified in human, including the major facilitator superfamily glucose facilitators GLUTs, the sodium-driven glucose symporters SGLTs, and the recently identified SWEETs. Structures of representative members or their prokaryotic homologs of all three families were obtained. This review focuses on the recent advances in the structural elucidation of the glucose transporters and the mechanistic insights derived from these structures, including the molecular basis for substrate recognition, alternating access, and stoichiometric coupling of co-transport.
Collapse
Affiliation(s)
- Dong Deng
- State Key Laboratory of Bio-Membrane and Membrane Biotechnology, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences and School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Nieng Yan
- State Key Laboratory of Bio-Membrane and Membrane Biotechnology, Center for Structural Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences and School of Medicine, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
23
|
Molecular cloning and functional characterization of a glucose transporter (CsGLUT) in Clonorchis sinensis. Parasitol Res 2015; 115:347-54. [PMID: 26450594 DOI: 10.1007/s00436-015-4754-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/28/2015] [Indexed: 10/22/2022]
Abstract
A complementary DNA (cDNA) encoding a glucose transporter of Clonorchis sinensis (CsGLUT) was isolated from the adult C. sinensis cDNA library. The open reading frame of CsGLUT cDNA consists of 1653 base pairs that encode a 550-amino acid residue protein. Hydropathy analysis suggested that CsGLUT possess 12 putative membrane-spanning domains. The Northern blot analysis result using poly(A)(+)RNA showed a strong band at ~2.1 kb for CsGLUT. When expressed in Xenopus oocytes, CsGLUT mediated the transport of radiolabeled deoxy-D-glucose in a time-dependent but sodium-independent manner. Concentration-dependency results showed saturable kinetics and followed the Michaelis-Menten equation. Nonlinear regression analyses yielded a Km value of 588.5 ± 53.0 μM and a Vmax value of 1500.0 ± 67.5 pmol/oocyte/30 min for [1,2-(3)H]2-deoxy-D-glucose. No trans-uptakes of bile acid (taurocholic acid), amino acids (tryptophan and arginine), or p-aminohippuric acid were observed. CsGLUT-mediated transport of deoxyglucose was significantly and concentration-dependently inhibited by radio-unlabeled deoxyglucose and D-glucose. 3-O-Methylglucose at 10 and 100 μM inhibited deoxyglucose uptake by ~50 % without concentration dependence. No inhibitory effects by galactose, mannose, and fructose were observed. This work may contribute to the molecular biological study of carbohydrate metabolism and new drug development of C. sinensis.
Collapse
|
24
|
Wang M, Yu C, Zhao H. Directed evolution of xylose specific transporters to facilitate glucose-xylose co-utilization. Biotechnol Bioeng 2015; 113:484-91. [DOI: 10.1002/bit.25724] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 08/06/2015] [Accepted: 08/09/2015] [Indexed: 01/19/2023]
Affiliation(s)
- Meng Wang
- Department of Chemical and Biomolecular Engineering; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
| | - Chenzhao Yu
- School of Molecular and Cellular Biology; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
| | - Huimin Zhao
- Department of Chemical and Biomolecular Engineering; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
- Department of Biochemistry; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
- Departments of Chemistry, Bioengineering, and Institute for Genomic Biology; University of Illinois at Urbana-Champaign; Urbana Illinois 61801
| |
Collapse
|
25
|
Sandhu P, Akhter Y. The internal gene duplication and interrupted coding sequences in the MmpL genes of Mycobacterium tuberculosis: Towards understanding the multidrug transport in an evolutionary perspective. Int J Med Microbiol 2015; 305:413-23. [PMID: 25841626 DOI: 10.1016/j.ijmm.2015.03.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/18/2014] [Accepted: 03/10/2015] [Indexed: 11/25/2022] Open
Abstract
The multidrug resistance has emerged as a major problem in the treatment of many of the infectious diseases. Tuberculosis (TB) is one of such disease caused by Mycobacterium tuberculosis. There is short term chemotherapy to treat the infection, but the main hurdle is the development of the resistance to antibiotics. This resistance is primarily due to the impermeable mycolic acid rich cell wall of the bacteria and other factors such as efflux of antibiotics from the bacterial cell. The MmpL (Mycobacterial Membrane Protein Large) proteins of mycobacteria are involved in the lipid transport and antibiotic efflux as indicated by the preliminary reports. We present here, comprehensive comparative sequence and structural analysis, which revealed topological signatures shared by the MmpL proteins and RND (Resistance Nodulation Division) multidrug efflux transporters. This provides evidence in support of the notion that they belong to the extended RND permeases superfamily. In silico modelled tertiary structures are in homology with an integral membrane component present in all of the RND efflux pumps. We document internal gene duplication and gene splitting events happened in the MmpL genes, which further elucidate the molecular functions of these putative transporters in an evolutionary perspective.
Collapse
Affiliation(s)
- Padmani Sandhu
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Shahpur District, Kangra 176206, Himachal Pradesh, India
| | - Yusuf Akhter
- Centre for Computational Biology and Bioinformatics, School of Life Sciences, Central University of Himachal Pradesh, Shahpur District, Kangra 176206, Himachal Pradesh, India.
| |
Collapse
|
26
|
Andersen JL, He GX, Kakarla P, K C R, Kumar S, Lakra WS, Mukherjee MM, Ranaweera I, Shrestha U, Tran T, Varela MF. Multidrug efflux pumps from Enterobacteriaceae, Vibrio cholerae and Staphylococcus aureus bacterial food pathogens. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:1487-547. [PMID: 25635914 PMCID: PMC4344678 DOI: 10.3390/ijerph120201487] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/15/2015] [Indexed: 02/07/2023]
Abstract
Foodborne illnesses caused by bacterial microorganisms are common worldwide and constitute a serious public health concern. In particular, microorganisms belonging to the Enterobacteriaceae and Vibrionaceae families of Gram-negative bacteria, and to the Staphylococcus genus of Gram-positive bacteria are important causative agents of food poisoning and infection in the gastrointestinal tract of humans. Recently, variants of these bacteria have developed resistance to medically important chemotherapeutic agents. Multidrug resistant Escherichia coli, Salmonella enterica, Vibrio cholerae, Enterobacter spp., and Staphylococcus aureus are becoming increasingly recalcitrant to clinical treatment in human patients. Of the various bacterial resistance mechanisms against antimicrobial agents, multidrug efflux pumps comprise a major cause of multiple drug resistance. These multidrug efflux pump systems reside in the biological membrane of the bacteria and actively extrude antimicrobial agents from bacterial cells. This review article summarizes the evolution of these bacterial drug efflux pump systems from a molecular biological standpoint and provides a framework for future work aimed at reducing the conditions that foster dissemination of these multidrug resistant causative agents through human populations.
Collapse
Affiliation(s)
- Jody L Andersen
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Gui-Xin He
- Department of Clinical Laboratory and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA.
| | - Prathusha Kakarla
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Ranjana K C
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Sanath Kumar
- QC Laboratory, Harvest and Post-Harvest Technology Division, Central Institute of Fisheries Education (CIFE), Seven Bungalows, Versova, Andheri (W), Mumbai 400061, India.
| | - Wazir Singh Lakra
- QC Laboratory, Harvest and Post-Harvest Technology Division, Central Institute of Fisheries Education (CIFE), Seven Bungalows, Versova, Andheri (W), Mumbai 400061, India.
| | - Mun Mun Mukherjee
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Indrika Ranaweera
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Ugina Shrestha
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| | - Thuy Tran
- Department of Clinical Laboratory and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA.
| | - Manuel F Varela
- Department of Biology, Eastern New Mexico University, Portales, NM 88130, USA.
| |
Collapse
|
27
|
Farwick A, Bruder S, Schadeweg V, Oreb M, Boles E. Engineering of yeast hexose transporters to transport D-xylose without inhibition by D-glucose. Proc Natl Acad Sci U S A 2014; 111:5159-64. [PMID: 24706835 PMCID: PMC3986176 DOI: 10.1073/pnas.1323464111] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
All known D-xylose transporters are competitively inhibited by D-glucose, which is one of the major reasons hampering simultaneous fermentation of D-glucose and D-xylose, two primary sugars present in lignocellulosic biomass. We have set up a yeast growth-based screening system for mutant D-xylose transporters that are insensitive to the presence of D-glucose. All of the identified variants had a mutation at either a conserved asparagine residue in transmembrane helix 8 or a threonine residue in transmembrane helix 5. According to a homology model of the yeast hexose transporter Gal2 deduced from the crystal structure of the D-xylose transporter XylE from Escherichia coli, both residues are found in the same region of the protein and are positioned slightly to the extracellular side of the central sugar-binding pocket. Therefore, it is likely that alterations sterically prevent D-glucose but not D-xylose from entering the pocket. In contrast, changing amino acids that are supposed to directly interact with the C6 hydroxymethyl group of D-glucose negatively affected transport of both D-glucose and D-xylose. Determination of kinetic properties of the mutant transporters revealed that Gal2-N376F had the highest affinity for D-xylose, along with a moderate transport velocity, and had completely lost the ability to transport hexoses. These transporter versions should prove valuable for glucose-xylose cofermentation in lignocellulosic hydrolysates by Saccharomyces cerevisiae and other biotechnologically relevant organisms. Moreover, our data contribute to the mechanistic understanding of sugar transport because the decisive role of the conserved asparagine residue for determining sugar specificity has not been recognized before.
Collapse
Affiliation(s)
- Alexander Farwick
- Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Stefan Bruder
- Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Virginia Schadeweg
- Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Mislav Oreb
- Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| | - Eckhard Boles
- Institute of Molecular Biosciences, Goethe University Frankfurt, 60438 Frankfurt am Main, Germany
| |
Collapse
|
28
|
Västermark A, Lunt B, Saier M. Major facilitator superfamily porters, LacY, FucP and XylE of Escherichia coli appear to have evolved positionally dissimilar catalytic residues without rearrangement of 3-TMS repeat units. J Mol Microbiol Biotechnol 2014; 24:82-90. [PMID: 24603210 DOI: 10.1159/000358429] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Based on alleged functional residue correspondences between FucP and LacY, a recent study has resulted in a proposed model of 3-TMS unit rearrangements [Madej et al.: Proc Natl Acad Sci USA 2013;110:5870-5874]. We rebut this theory, using 7 different lines of evidence. Our observations suggest that these two transporters are homologous throughout their lengths, having evolved from a common ancestor without repeat unit rearrangements. We exploit the availability of the high-resolution XylE crystal structures in multiple conformations including the inward-facing state to render possible direct comparisons with LacY. Based on a Δdistance map, we confirm the conclusion of Quistgaard et al. [Nat Struct Mol Biol 2013;20:766-768] that the N-terminal 6 TMS halves of these transporters are internally less mobile than the second halves during the conformational transition from the outward occluded state to the inward occluded state and inward occluded state to inward open state. These observations, together with those of Madej et al. [2013], lead to the suggestion that functionally equivalent catalytic residues involved in substrate binding and transport catalysis have evolved in dissimilar positions, but apparently often in similar positions in the putative 3-TMS repeat units, from a single structural scaffold without intragenic rearrangement.
Collapse
Affiliation(s)
- Ake Västermark
- Department of Molecular Biology, University of California at San Diego, La Jolla, Calif., USA
| | | | | |
Collapse
|
29
|
Abstract
The Major Facilitator Superfamily (MFS) is a diverse group of secondary transporters with over 10,000 members, found in all kingdoms of life, including Homo sapiens. One objective of determining crystallographic models of the bacterial representatives is identification and physical localization of residues important for catalysis in transporters with medical relevance. The recently solved crystallographic models of the D-xylose permease XylE from Escherichia coli and GlcP from Staphylococcus epidermidus, homologs of the human D-glucose transporters, the GLUTs (SLC2), provide information about the structure of these transporters. The goal of this work is to examine general concepts derived from the bacterial XylE, GlcP, and other MFS transporters for their relevance to the GLUTs by comparing conservation of functionally critical residues. An energy landscape for symport and uniport is presented. Furthermore, the substrate selectivity of XylE is compared with GLUT1 and GLUT5, as well as a XylE mutant that transports D-glucose.
Collapse
|
30
|
Modulation of Bacterial Multidrug Resistance Efflux Pumps of the Major Facilitator Superfamily. INTERNATIONAL JOURNAL OF BACTERIOLOGY 2013; 2013. [PMID: 25750934 PMCID: PMC4347946 DOI: 10.1155/2013/204141] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial infections pose a serious public health concern, especially when an infectious disease has a multidrug resistant causative agent. Such multidrug resistant bacteria can compromise the clinical utility of major chemotherapeutic antimicrobial agents. Drug and multidrug resistant bacteria harbor several distinct molecular mechanisms for resistance. Bacterial antimicrobial agent efflux pumps represent a major mechanism of clinical resistance. The major facilitator superfamily (MFS) is one of the largest groups of solute transporters to date and includes a significant number of bacterial drug and multidrug efflux pumps. We review recent work on the modulation of multidrug efflux pumps, paying special attention to those transporters belonging primarily to the MFS.
Collapse
|
31
|
Cura AJ, Carruthers A. Role of monosaccharide transport proteins in carbohydrate assimilation, distribution, metabolism, and homeostasis. Compr Physiol 2013; 2:863-914. [PMID: 22943001 DOI: 10.1002/cphy.c110024] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The facilitated diffusion of glucose, galactose, fructose, urate, myoinositol, and dehydroascorbicacid in mammals is catalyzed by a family of 14 monosaccharide transport proteins called GLUTs. These transporters may be divided into three classes according to sequence similarity and function/substrate specificity. GLUT1 appears to be highly expressed in glycolytically active cells and has been coopted in vitamin C auxotrophs to maintain the redox state of the blood through transport of dehydroascorbate. Several GLUTs are definitive glucose/galactose transporters, GLUT2 and GLUT5 are physiologically important fructose transporters, GLUT9 appears to be a urate transporter while GLUT13 is a proton/myoinositol cotransporter. The physiologic substrates of some GLUTs remain to be established. The GLUTs are expressed in a tissue specific manner where affinity, specificity, and capacity for substrate transport are paramount for tissue function. Although great strides have been made in characterizing GLUT-catalyzed monosaccharide transport and mapping GLUT membrane topography and determinants of substrate specificity, a unifying model for GLUT structure and function remains elusive. The GLUTs play a major role in carbohydrate homeostasis and the redistribution of sugar-derived carbons among the various organ systems. This is accomplished through a multiplicity of GLUT-dependent glucose sensing and effector mechanisms that regulate monosaccharide ingestion, absorption,distribution, cellular transport and metabolism, and recovery/retention. Glucose transport and metabolism have coevolved in mammals to support cerebral glucose utilization.
Collapse
Affiliation(s)
- Anthony J Cura
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | |
Collapse
|
32
|
de la Paz Celorio-Mancera M, Wheat CW, Vogel H, Söderlind L, Janz N, Nylin S. Mechanisms of macroevolution: polyphagous plasticity in butterfly larvae revealed by RNA-Seq. Mol Ecol 2013; 22:4884-95. [DOI: 10.1111/mec.12440] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/20/2013] [Accepted: 06/26/2013] [Indexed: 12/15/2022]
Affiliation(s)
| | - Christopher W. Wheat
- Department of Zoology Ecology; Stockholm University; Svante Arrheniusväg 18 B, 106 91 Stockholm Sweden
| | - Heiko Vogel
- Department of Entomology; Max Planck Institute for Chemical Ecology; Beutenberg Campus Hans-Knöll Straβe 8 07745 Jena Germany
| | - Lina Söderlind
- Department of Zoology Ecology; Stockholm University; Svante Arrheniusväg 18 B, 106 91 Stockholm Sweden
| | - Niklas Janz
- Department of Zoology Ecology; Stockholm University; Svante Arrheniusväg 18 B, 106 91 Stockholm Sweden
| | - Sören Nylin
- Department of Zoology Ecology; Stockholm University; Svante Arrheniusväg 18 B, 106 91 Stockholm Sweden
| |
Collapse
|
33
|
Yan N. Structural investigation of the proton-coupled secondary transporters. Curr Opin Struct Biol 2013; 23:483-91. [DOI: 10.1016/j.sbi.2013.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 04/17/2013] [Accepted: 04/19/2013] [Indexed: 11/16/2022]
|
34
|
Yan N. Structural advances for the major facilitator superfamily (MFS) transporters. Trends Biochem Sci 2013; 38:151-9. [PMID: 23403214 DOI: 10.1016/j.tibs.2013.01.003] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 01/03/2013] [Accepted: 01/07/2013] [Indexed: 01/18/2023]
Abstract
The major facilitator superfamily (MFS) is one of the largest groups of secondary active transporters conserved from bacteria to humans. MFS proteins selectively transport a wide spectrum of substrates across biomembranes and play a pivotal role in multiple physiological processes. Despite intense investigation, only seven MFS proteins from six subfamilies have been structurally elucidated. These structures were captured in distinct states during a transport cycle involving alternating access to binding sites from either side of the membrane. This review discusses recent progress in MFS structure analysis and focuses on the molecular basis for substrate binding, co-transport coupling, and alternating access.
Collapse
Affiliation(s)
- Nieng Yan
- State Key Laboratory of Bio-membrane and Membrane Biotechnology, Center for Structural Biology, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
35
|
Sun L, Zeng X, Yan C, Sun X, Gong X, Rao Y, Yan N. Crystal structure of a bacterial homologue of glucose transporters GLUT1-4. Nature 2012; 490:361-6. [PMID: 23075985 DOI: 10.1038/nature11524] [Citation(s) in RCA: 348] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 08/17/2012] [Indexed: 12/22/2022]
Abstract
Glucose transporters are essential for metabolism of glucose in cells of diverse organisms from microbes to humans, exemplified by the disease-related human proteins GLUT1, 2, 3 and 4. Despite rigorous efforts, the structural information for GLUT1-4 or their homologues remains largely unknown. Here we report three related crystal structures of XylE, an Escherichia coli homologue of GLUT1-4, in complex with d-xylose, d-glucose and 6-bromo-6-deoxy-D-glucose, at resolutions of 2.8, 2.9 and 2.6 Å, respectively. The structure consists of a typical major facilitator superfamily fold of 12 transmembrane segments and a unique intracellular four-helix domain. XylE was captured in an outward-facing, partly occluded conformation. Most of the important amino acids responsible for recognition of D-xylose or d-glucose are invariant in GLUT1-4, suggesting functional and mechanistic conservations. Structure-based modelling of GLUT1-4 allows mapping and interpretation of disease-related mutations. The structural and biochemical information reported here constitutes an important framework for mechanistic understanding of glucose transporters and sugar porters in general.
Collapse
Affiliation(s)
- Linfeng Sun
- State Key Laboratory of Bio-membrane and Membrane Biotechnology, Center for Structural Biology, Tsinghua University, Beijing 100084, China
| | | | | | | | | | | | | |
Collapse
|
36
|
On the evolution of hexose transporters in kinetoplastid Protozoans [corrected]. PLoS One 2012; 7:e36303. [PMID: 22567148 PMCID: PMC3342237 DOI: 10.1371/journal.pone.0036303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 04/02/2012] [Indexed: 11/24/2022] Open
Abstract
Glucose, an almost universally used energy and carbon source, is processed through several well-known metabolic pathways, primarily glycolysis. Glucose uptake is considered to be the first step in glycolysis. In kinetoplastids, a protozoan group that includes relevant human pathogens, the importance of glucose uptake in different phases of the life cycles is well established, and hexose transporters have been proposed as targets for therapeutic drugs. However, little is known about the evolutionary history of these hexose transporters. Hexose transporters contain an intracellular N- and C- termini, and 12 transmembrane spans connected by alternate intracellular and extracellular loops. In the present work we tested the hypothesis that the evolutionary rate of the transmembrane span is different from that of the whole sequence and that it is possible to define evolutionary units inside the sequence. The phylogeny of whole molecules was compared to that of their transmembrane spans and the loops connecting the transmembrane spans. We show that the evolutionary units in these proteins primarily consist of clustered rather than individual transmembrane spans. These analyses demonstrate that there are evolutionary constraints on the organization of these proteins; more specifically, the order of the transmembrane spans along the protein is highly conserved. Finally, we defined a signature sequence for the identification of kinetoplastid hexose transporters.
Collapse
|
37
|
Biochemistry of bacterial multidrug efflux pumps. Int J Mol Sci 2012; 13:4484-4495. [PMID: 22605991 PMCID: PMC3344227 DOI: 10.3390/ijms13044484] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 03/09/2012] [Accepted: 03/15/2012] [Indexed: 11/17/2022] Open
Abstract
Bacterial pathogens that are multi-drug resistant compromise the effectiveness of treatment when they are the causative agents of infectious disease. These multi-drug resistance mechanisms allow bacteria to survive in the presence of clinically useful antimicrobial agents, thus reducing the efficacy of chemotherapy towards infectious disease. Importantly, active multi-drug efflux is a major mechanism for bacterial pathogen drug resistance. Therefore, because of their overwhelming presence in bacterial pathogens, these active multi-drug efflux mechanisms remain a major area of intense study, so that ultimately measures may be discovered to inhibit these active multi-drug efflux pumps.
Collapse
|
38
|
Novel transporter required for biogenesis of cbb3-type cytochrome c oxidase in Rhodobacter capsulatus. mBio 2012; 3:mBio.00293-11. [PMID: 22294680 PMCID: PMC3266609 DOI: 10.1128/mbio.00293-11] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
UNLABELLED The acquisition, delivery, and incorporation of metals into their respective metalloproteins are important cellular processes. These processes are tightly controlled in order to prevent exposure of cells to free-metal concentrations that could yield oxidative damage. Copper (Cu) is one such metal that is required as a cofactor in a variety of proteins. However, when present in excessive amounts, Cu is toxic due to its oxidative capability. Cytochrome c oxidases (Coxs) are among the metalloproteins whose assembly and activity require the presence of Cu in their catalytic subunits. In this study, we focused on the acquisition of Cu for incorporation into the heme-Cu binuclear center of the cbb(3)-type Cox (cbb(3)-Cox) in the facultative phototroph Rhodobacter capsulatus. Genetic screens identified a cbb(3)-Cox defective mutant that requires Cu(2+) supplementation to produce an active cbb(3)-Cox. Complementation of this mutant using wild-type genomic libraries unveiled a novel gene (ccoA) required for cbb(3)-Cox biogenesis. In the absence of CcoA, the cellular Cu content decreases and cbb(3)-Cox assembly and activity become defective. CcoA shows homology to major facilitator superfamily (MFS)-type transporter proteins. Members of this family are known to transport small solutes or drugs, but so far, no MFS protein has been implicated in cbb(3)-Cox biogenesis. These findings provide novel insights into the maturation and assembly of membrane-integral metalloproteins and on a hitherto-unknown function(s) of MFS-type transporters in bacterial Cu acquisition. IMPORTANCE Biogenesis of energy-transducing membrane-integral enzymes, like the heme copper-containing cytochrome c oxidases, and the acquisition of transition metals, like copper, as their catalytic cofactors are vital processes for all cells. These widespread and well-controlled processes are poorly understood in all organisms, including bacteria. Defects in these processes lead to severe mitochondrial diseases in humans and poor crop yields in plants. In this study, using the facultative phototroph Rhodobacter capsulatus as a model organism, we report on the discovery of a novel major facilitator superfamily (MFS)-type transporter (CcoA) that affects cellular copper content and cbb(3)-type cytochrome c oxidase production in bacteria.
Collapse
|
39
|
Magnetic resonance imaging of tumors colonized with bacterial ferritin-expressing Escherichia coli. PLoS One 2011; 6:e25409. [PMID: 21984917 PMCID: PMC3184983 DOI: 10.1371/journal.pone.0025409] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2011] [Accepted: 09/02/2011] [Indexed: 12/31/2022] Open
Abstract
Background Recent studies have shown that human ferritin can be used as a reporter of gene expression for magnetic resonance imaging (MRI). Bacteria also encode three classes of ferritin-type molecules with iron accumulation properties. Methods and Findings Here, we investigated whether these bacterial ferritins can also be used as MRI reporter genes and which of the bacterial ferritins is the most suitable reporter. Bacterial ferritins were overexpressed in probiotic E. coli Nissle 1917. Cultures of these bacteria were analyzed and those generating highest MRI contrast were further investigated in tumor bearing mice. Among members of three classes of bacterial ferritin tested, bacterioferritin showed the most promise as a reporter gene. Although all three proteins accumulated similar amounts of iron when overexpressed individually, bacterioferritin showed the highest contrast change. By site-directed mutagenesis we also show that the heme iron, a unique part of the bacterioferritin molecule, is not critical for MRI contrast change. Tumor-specific induction of bacterioferritin-expression in colonized tumors resulted in contrast changes within the bacteria-colonized tumors. Conclusions Our data suggest that colonization and gene expression by live vectors expressing bacterioferritin can be monitored by MRI due to contrast changes.
Collapse
|
40
|
Malle E, Zhou H, Neuhold J, Spitzenberger B, Klepsch F, Pollak T, Bergner O, Ecker GF, Stolt-Bergner PC. Random mutagenesis of the prokaryotic peptide transporter YdgR identifies potential periplasmic gating residues. J Biol Chem 2011; 286:23121-31. [PMID: 21558271 DOI: 10.1074/jbc.m111.239657] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The peptide transporter (PTR) family represents a group of proton-coupled secondary transporters responsible for bulk uptake of amino acids in the form of di- and tripeptides, an essential process employed across species ranging from bacteria to humans. To identify amino acids critical for peptide transport in a prokaryotic PTR member, we have screened a library of mutants of the Escherichia coli peptide transporter YdgR using a high-throughput substrate uptake assay. We have identified 35 single point mutations that result in a full or partial loss of transport activity. Additional analysis, including homology modeling based on the crystal structure of the Shewanella oneidensis peptide transporter PepT(so), identifies Glu(56) and Arg(305) as potential periplasmic gating residues. In addition to providing new insights into transport by members of the PTR family, these mutants provide valuable tools for further study of the mechanism of peptide transport.
Collapse
Affiliation(s)
- Elisabeth Malle
- Research Institute of Molecular Pathology, Dr. Bohr-gasse 7, 1030 Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Govind G, Mittapalli O, Griebel T, Allmann S, Böcker S, Baldwin IT. Unbiased transcriptional comparisons of generalist and specialist herbivores feeding on progressively defenseless Nicotiana attenuata plants. PLoS One 2010; 5:e8735. [PMID: 20090945 PMCID: PMC2806910 DOI: 10.1371/journal.pone.0008735] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Accepted: 12/20/2009] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Herbivore feeding elicits dramatic increases in defenses, most of which require jasmonate (JA) signaling, and against which specialist herbivores are thought to be better adapted than generalist herbivores. Unbiased transcriptional analyses of how neonate larvae cope with these induced plant defenses are lacking. METHODOLOGY/PRINCIPAL FINDINGS We created cDNA microarrays for Manduca sexta and Heliothis virescens separately, by spotting normalized midgut-specific cDNA libraries created from larvae that fed for 24 hours on MeJA-elicited wild-type (WT) Nicotiana attenuata plants. These microarrays were hybridized with labeled probes from neonates that fed for 24 hours on WT and isogenic plants progressively silenced in JA-mediated defenses (N: nicotine; N/PI: N and trypsin protease inhibitors; JA: all JA-mediated defenses). H. virescens neonates regulated 16 times more genes than did M. sexta neonates when they fed on plants silenced in JA-mediated defenses, and for both species, the greater the number of defenses silenced in the host plant (JA > N/PI > N), the greater were the number of transcripts regulated in the larvae. M. sexta larvae tended to down-regulate while H. virescens larvae up- and down-regulated transcripts from the same functional categories of genes. M. sexta larvae regulated transcripts in a diet-specific manner, while H. virescens larvae regulated a similar suite of transcripts across all diet types. CONCLUSIONS/SIGNIFICANCE The observations are consistent with the expectation that specialists are better adapted than generalist herbivores to the defense responses elicited in their host plants by their feeding. While M. sexta larvae appear to be better adapted to N. attenuata's defenses, some of the elicited responses remain effective defenses against both herbivore species. The regulated genes provide novel insights into larval adaptations to N. attenuata's induced defenses, and represent potential targets for plant-mediated RNAi to falsify hypotheses about the process of adaptation.
Collapse
Affiliation(s)
- Geetha Govind
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | | | - Thasso Griebel
- Faculty of Mathematics and Computer Science, Friedrich Schiller University of Jena, Jena, Germany
| | - Silke Allmann
- Max Planck Institute for Chemical Ecology, Jena, Germany
| | - Sebastian Böcker
- Faculty of Mathematics and Computer Science, Friedrich Schiller University of Jena, Jena, Germany
| | | |
Collapse
|
43
|
Corda D, Zizza P, Varone A, Filippi BM, Mariggiò S. The glycerophosphoinositols: cellular metabolism and biological functions. Cell Mol Life Sci 2009; 66:3449-67. [PMID: 19669618 PMCID: PMC11115907 DOI: 10.1007/s00018-009-0113-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Revised: 06/26/2009] [Accepted: 07/16/2009] [Indexed: 12/25/2022]
Abstract
The glycerophosphoinositols are cellular products of phospholipase A(2) and lysolipase activities on the membrane phosphoinositides. Their intracellular concentrations can vary upon oncogenic transformation, cell differentiation and hormonal stimulation. Specific glycerophosphodiester phosphodiesterases are involved in their catabolism, which, as with their formation, is under hormonal regulation. With their mechanisms of action including modulation of adenylyl cyclase, intracellular calcium levels, and Rho-GTPases, the glycerophosphoinositols have diverse effects in multiple cell types: induction of cell proliferation in thyroid cells; modulation of actin cytoskeleton organisation in fibroblasts; and reduction of the invasive potential of tumour cell lines. More recent investigations include their effects in inflammatory and immune responses. Indeed, the glycerophosphoinositols enhance cytokine-dependent chemotaxis in T-lymphocytes induced by SDF-1alpha-receptor activation, indicating roles for these compounds as modulators of T-cell signalling and T-cell responses.
Collapse
Affiliation(s)
- Daniela Corda
- Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
| | - Pasquale Zizza
- Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
| | - Alessia Varone
- Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
| | - Beatrice Maria Filippi
- Present Address: MRC Protein Phosphorylation Unit, College of Life Sciences, University of Dundee, Dundee, Scotland, UK
| | - Stefania Mariggiò
- Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro, Chieti Italy
| |
Collapse
|
44
|
Tse YM, Yu M, Tsang JSH. Topological analysis of a haloacid permease of a Burkholderia sp. bacterium with a PhoA-LacZ reporter. BMC Microbiol 2009; 9:233. [PMID: 19878597 PMCID: PMC2777183 DOI: 10.1186/1471-2180-9-233] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 10/31/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND 2-Haloacids can be found in the natural environment as degradative products of natural and synthetic halogenated compounds. They can also be generated by disinfection of water and have been shown to be mutagenic and to inhibit glyceraldehyde-3-phosphate dehydrogenase activity. We have recently identified a novel haloacid permease Deh4p from a bromoacetate-degrading bacterium Burkholderia sp. MBA4. Comparative analyses suggested that Deh4p is a member of the Major Facilitator Superfamily (MFS), which includes thousands of membrane transporter proteins. Members of the MFS usually possess twelve putative transmembrane segments (TMS). Deh4p was predicted to have twelve TMS. In this study we characterized the topology of Deh4p with a PhoA-LacZ dual reporters system. RESULTS Thirty-six Deh4p-reporter recombinants were constructed and expressed in E. coli. Both PhoA and LacZ activities were determined in these cells. Strength indices were calculated to determine the locations of the reporters. The results mainly agree with the predicted model. However, two of the TMS were not verified. This lack of confirmation of the TMS, using a reporter, has been reported previously. Further comparative analysis of Deh4p has assigned it to the Metabolite:H+ Symporter (MHS) 2.A.1.6 family with twelve TMS. Deh4p exhibits many common features of the MHS family proteins. Deh4p is apparently a member of the MFS but with some atypical features. CONCLUSION The PhoA-LacZ reporter system is convenient for analysis of the topology of membrane proteins. However, due to the limitation of the biological system, verification of some of the TMS of the protein was not successful. The present study also makes use of bioinformatic analysis to verify that the haloacid permease Deh4p of Burkholderia sp. MBA4 is a MFS protein but with atypical features.
Collapse
Affiliation(s)
- Yuk Man Tse
- Molecular Microbiology Laboratory, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong.
| | | | | |
Collapse
|
45
|
Leandro MJ, Fonseca CÃ, Gonçalves P. Hexose and pentose transport in ascomycetous yeasts: an overview. FEMS Yeast Res 2009; 9:511-25. [DOI: 10.1111/j.1567-1364.2009.00509.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
46
|
Szakonyi G, Leng D, Ma P, Bettaney KE, Saidijam M, Ward A, Zibaei S, Gardiner AT, Cogdell RJ, Butaye P, Kolsto AB, O'reilly J, Hope RJ, Rutherford NG, Hoyle CJ, Henderson PJF. A genomic strategy for cloning, expressing and purifying efflux proteins of the major facilitator superfamily. J Antimicrob Chemother 2007; 59:1265-70. [PMID: 17412722 DOI: 10.1093/jac/dkm036] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A genomic strategy for the overexpression of bacterial multidrug and antibiotic resistance membrane efflux proteins in Escherichia coli is described. Expression is amplified so that the encoded proteins from a range of Gram-positive and Gram-negative bacteria comprise 5% to 35% of E. coli inner membrane protein. Depending upon their topology, proteins are produced with RGS(His)(6)-tag or a Strep-tag at the C terminus. These tags facilitate the purification of the overexpressed proteins in milligram quantities for structural studies. The strategy is illustrated for the bicyclomycin resistance efflux protein, Bcr, of E. coli.
Collapse
Affiliation(s)
- Gerda Szakonyi
- Astbury Centre for Structural Molecular Biology, Faculty of Biological Sciences, Institute for Membrane and Systems Biology, University of Leeds, Leeds, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Mariggiò S, Iurisci C, Sebastià J, Patton-Vogt J, Corda D. Molecular characterization of a glycerophosphoinositol transporter in mammalian cells. FEBS Lett 2006; 580:6789-96. [PMID: 17141226 DOI: 10.1016/j.febslet.2006.11.039] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Revised: 11/04/2006] [Accepted: 11/13/2006] [Indexed: 11/20/2022]
Abstract
The glycerophosphoinositols are ubiquitous phosphoinositide metabolites involved in the control of several cell functions. They exert their actions both intracellularly and by rapidly equilibrating across the plasma membrane when added to cells, implying the existence of a transporter for their membrane permeation. Such a transporter, GIT1, has been cloned in yeast. By PSI-BLAST analysis, we have identified the Glut2 transporter as a human-genome candidate ortholog of GIT1. This was supported directly through the use of inhibitors, siRNAs and competition studies of specific uptake of GroPIns in HeLa cells over-expressing human Glut2. These data identify Glut2 as a GroPIns transporter in mammals, and define a physiologically relevant cell-permeation mechanism.
Collapse
Affiliation(s)
- Stefania Mariggiò
- Department of Cell Biology and Oncology, Consorzio Mario Negri Sud, Via Nazionale 8A, 66030 Santa Maria Imbaro (Chieti), Italy.
| | | | | | | | | |
Collapse
|
48
|
Sobczak I, Lolkema JS. The 2-hydroxycarboxylate transporter family: physiology, structure, and mechanism. Microbiol Mol Biol Rev 2006; 69:665-95. [PMID: 16339740 PMCID: PMC1306803 DOI: 10.1128/mmbr.69.4.665-695.2005] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The 2-hydroxycarboxylate transporter family is a family of secondary transporters found exclusively in the bacterial kingdom. They function in the metabolism of the di- and tricarboxylates malate and citrate, mostly in fermentative pathways involving decarboxylation of malate or oxaloacetate. These pathways are found in the class Bacillales of the low-CG gram-positive bacteria and in the gamma subdivision of the Proteobacteria. The pathways have evolved into a remarkable diversity in terms of the combinations of enzymes and transporters that built the pathways and of energy conservation mechanisms. The transporter family includes H+ and Na+ symporters and precursor/product exchangers. The proteins consist of a bundle of 11 transmembrane helices formed from two homologous domains containing five transmembrane segments each, plus one additional segment at the N terminus. The two domains have opposite orientations in the membrane and contain a pore-loop or reentrant loop structure between the fourth and fifth transmembrane segments. The two pore-loops enter the membrane from opposite sides and are believed to be part of the translocation site. The binding site is located asymmetrically in the membrane, close to the interface of membrane and cytoplasm. The binding site in the translocation pore is believed to be alternatively exposed to the internal and external media. The proposed structure of the 2HCT transporters is different from any known structure of a membrane protein and represents a new structural class of secondary transporters.
Collapse
Affiliation(s)
- Iwona Sobczak
- Molecular Microbiology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Kerklaan 30, 9751 NN Haren, The Netherlands
| | | |
Collapse
|
49
|
Schivell AE, Mochida S, Kensel-Hammes P, Custer KL, Bajjalieh SM. SV2A and SV2C contain a unique synaptotagmin-binding site. Mol Cell Neurosci 2005; 29:56-64. [PMID: 15866046 DOI: 10.1016/j.mcn.2004.12.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2004] [Revised: 12/22/2004] [Accepted: 12/31/2004] [Indexed: 10/25/2022] Open
Abstract
SV2 (Synaptic Vesicle Protein 2) is expressed in neurons and endocrine cells where it is required for normal calcium-evoked neurosecretion. In mammals, there are three SV2 genes, denoted SV2A, B and C. SV2A interacts with synaptotagmin, the prime candidate for the calcium sensor in exocytosis. Here, we report that all isoforms of native SV2 bind synaptotagmin and that binding is inhibited by calcium, indicating that all isoforms contain a common calcium-inhibited synaptotagmin-binding site. The isolated amino termini of SV2A and SV2C supported an additional interaction with synaptotagmin, and binding at this site was stimulated by calcium. The amino-terminal binding site was mapped to the first 57 amino acids of SV2A, and removal of this domain decreased calcium-mediated inhibition of binding to synaptotagmin, suggesting that it modulates calcium's effect on the SV2-synaptotagmin interaction. Introduction of the amino terminus of SV2A or SV2C into cultured superior cervical ganglion neurons inhibited neurotransmission, whereas the amino terminus of SV2B did not. These observations implicate the SV2-synaptotagmin interaction in regulated exocytosis and suggest that SV2A and SV2C, via their additional synaptotagmin binding site, function differently than SV2B.
Collapse
Affiliation(s)
- Amanda E Schivell
- Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
50
|
Lolkema JS, Sobczak I, Slotboom DJ. Secondary transporters of the 2HCT family contain two homologous domains with inverted membrane topology and trans re-entrant loops. FEBS J 2005; 272:2334-44. [PMID: 15853816 DOI: 10.1111/j.1742-4658.2005.04665.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The 2-hydroxycarboxylate transporter (2HCT) family of secondary transporters belongs to a much larger structural class of secondary transporters termed ST3 which contains about 2000 transporters in 32 families. The transporters of the 2HCT family are among the best studied in the class. Here we detect weak sequence similarity between the N- and C-terminal halves of the proteins using a sensitive method which uses a database containing the N- and C-terminal halves of all the sequences in ST3 and involves blast searches of each sequence in the database against the whole database. Unrelated families of secondary transporters of the same length and composition were used as controls. The sequence similarity involved major parts of the N- and C-terminal halves and not just a small stretch. The membrane topology of the homologous N- and C-terminal domains was deduced from the experimentally determined topology of the members of the 2HCT family. The domains consist of five transmembrane segments each and have opposite orientations in the membrane. The N terminus of the N-terminal domain is extracellular, while the N terminus of the C-terminal domain is cytoplasmic. The loops between the fourth and fifth transmembrane segment in each domain are well conserved throughout the class and contain a high fraction of residues with small side chains, Gly, Ala and Ser. Experimental work on the citrate transporter CitS in the 2HCT family indicates that the loops are re-entrant or pore loops. The re-entrant loops in the N- and C-terminal domains enter the membrane from opposite sides (trans-re-entrant loops). The combination of inverted membrane topology and trans-re-entrant loops represents a new fold for secondary transporters and resembles the structure of aquaporins and models proposed for Na+/Ca2+ exchangers.
Collapse
Affiliation(s)
- Juke S Lolkema
- Molecular Microbiology, Biomolecular Sciences and Biotechnology Institute, University of Groningen, the Netherlands.
| | | | | |
Collapse
|