Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Biol ; 21(5): e3002091, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37192172

RESUMO

The streptothricin natural product mixture (also known as nourseothricin) was discovered in the early 1940s, generating intense initial interest because of excellent gram-negative activity. Here, we establish the activity spectrum of nourseothricin and its main components, streptothricin F (S-F, 1 lysine) and streptothricin D (S-D, 3 lysines), purified to homogeneity, against highly drug-resistant, carbapenem-resistant Enterobacterales (CRE) and Acinetobacter baumannii. For CRE, the MIC50 and MIC90 for S-F and S-D were 2 and 4 µM, and 0.25 and 0.5 µM, respectively. S-F and nourseothricin showed rapid, bactericidal activity. S-F and S-D both showed approximately 40-fold greater selectivity for prokaryotic than eukaryotic ribosomes in in vitro translation assays. In vivo, delayed renal toxicity occurred at >10-fold higher doses of S-F compared with S-D. Substantial treatment effect of S-F in the murine thigh model was observed against the otherwise pandrug-resistant, NDM-1-expressing Klebsiella pneumoniae Nevada strain with minimal or no toxicity. Cryo-EM characterization of S-F bound to the A. baumannii 70S ribosome defines extensive hydrogen bonding of the S-F steptolidine moiety, as a guanine mimetic, to the 16S rRNA C1054 nucleobase (Escherichia coli numbering) in helix 34, and the carbamoylated gulosamine moiety of S-F with A1196, explaining the high-level resistance conferred by corresponding mutations at the residues identified in single rrn operon E. coli. Structural analysis suggests that S-F probes the A-decoding site, which potentially may account for its miscoding activity. Based on unique and promising activity, we suggest that the streptothricin scaffold deserves further preclinical exploration as a potential therapeutic for drug-resistant, gram-negative pathogens.


Assuntos
Antibacterianos , Estreptotricinas , Animais , Camundongos , Antibacterianos/farmacologia , Estreptotricinas/química , Estreptotricinas/farmacologia , Escherichia coli/genética , RNA Ribossômico 16S/genética , Bactérias Gram-Negativas , Carbapenêmicos/farmacologia , Ribossomos , Testes de Sensibilidade Microbiana
2.
Mol Cell Proteomics ; 22(12): 100666, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37839702

RESUMO

The application of integrated systems biology to the field of structural biology is a promising new direction, although it is still in the infant stages of development. Here we report the use of single particle cryo-EM to identify multiple proteins from three enriched heterogeneous fractions prepared from human liver mitochondrial lysate. We simultaneously identify and solve high-resolution structures of nine essential mitochondrial enzymes with key metabolic functions, including fatty acid catabolism, reactive oxidative species clearance, and amino acid metabolism. Our methodology also identified multiple distinct members of the acyl-CoA dehydrogenase family. This work highlights the potential of cryo-EM to explore tissue proteomics at the atomic level.


Assuntos
Mitocôndrias , Proteômica , Humanos , Mitocôndrias/metabolismo , Fígado/metabolismo , Oxirredução
3.
Nat Methods ; 18(1): 69-75, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33408407

RESUMO

Single-particle cryo-electron microscopy (cryo-EM) has become a powerful technique in the field of structural biology. However, the inability to reliably produce pure, homogeneous membrane protein samples hampers the progress of their structural determination. Here, we develop a bottom-up iterative method, Build and Retrieve (BaR), that enables the identification and determination of cryo-EM structures of a variety of inner and outer membrane proteins, including membrane protein complexes of different sizes and dimensions, from a heterogeneous, impure protein sample. We also use the BaR methodology to elucidate structural information from Escherichia coli K12 crude membrane and raw lysate. The findings demonstrate that it is possible to solve high-resolution structures of a number of relatively small (<100 kDa) and less abundant (<10%) unidentified membrane proteins within a single, heterogeneous sample. Importantly, these results highlight the potential of cryo-EM for systems structural proteomics.


Assuntos
Proteínas de Bactérias/química , Burkholderia pseudomallei/metabolismo , Membrana Celular/metabolismo , Microscopia Crioeletrônica/métodos , Proteínas de Membrana/química , Proteínas de Bactérias/metabolismo , Membrana Celular/química , Escherichia coli/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Modelos Moleculares , Estrutura Molecular , Conformação Proteica
4.
PLoS Biol ; 19(8): e3001370, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34383749

RESUMO

The mycobacterial membrane protein large 3 (MmpL3) transporter is essential and required for shuttling the lipid trehalose monomycolate (TMM), a precursor of mycolic acid (MA)-containing trehalose dimycolate (TDM) and mycolyl arabinogalactan peptidoglycan (mAGP), in Mycobacterium species, including Mycobacterium tuberculosis and Mycobacterium smegmatis. However, the mechanism that MmpL3 uses to facilitate the transport of fatty acids and lipidic elements to the mycobacterial cell wall remains elusive. Here, we report 7 structures of the M. smegmatis MmpL3 transporter in its unbound state and in complex with trehalose 6-decanoate (T6D) or TMM using single-particle cryo-electron microscopy (cryo-EM) and X-ray crystallography. Combined with calculated results from molecular dynamics (MD) and target MD simulations, we reveal a lipid transport mechanism that involves a coupled movement of the periplasmic domain and transmembrane helices of the MmpL3 transporter that facilitates the shuttling of lipids to the mycobacterial cell wall.


Assuntos
Proteínas de Bactérias/metabolismo , Fatores Corda/metabolismo , Metabolismo dos Lipídeos , Proteínas de Membrana Transportadoras/metabolismo , Mycobacterium smegmatis/metabolismo , Proteínas de Bactérias/ultraestrutura , Microscopia Crioeletrônica , Decanoatos/metabolismo , Escherichia coli , Proteínas de Membrana Transportadoras/ultraestrutura , Simulação de Dinâmica Molecular , Mycobacterium smegmatis/ultraestrutura , Trealose/metabolismo
5.
EMBO Rep ; 22(3): e51628, 2021 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-33471955

RESUMO

The intra-erythrocyte stage of P. falciparum relies primarily on glycolysis to generate adenosine triphosphate (ATP) and the energy required to support growth and reproduction. Lactic acid, a metabolic byproduct of glycolysis, is potentially toxic as it lowers the pH inside the parasite. Plasmodium falciparum formate-nitrite transporter (PfFNT), a 34-kDa transmembrane protein, has been identified as a novel drug target as it exports lactate from inside the parasite to the surrounding parasitophorous vacuole within the erythrocyte cytosol. The structure and detailed molecular mechanism of this membrane protein are not yet available. Here we present structures of PfFNT in the absence and presence of the functional inhibitor MMV007839 at resolutions of 2.56 Å and 2.78 Å using single-particle cryo-electron microscopy. Genetic analysis and transport assay indicate that PfFNT is able to transfer lactate across the membrane. Combined, our data suggest a stepwise displacement mechanism for substrate transport. The PfFNT membrane protein is capable of picking up lactate ions from the parasite's cytosol, converting them to lactic acids and then exporting these acids into the extracellular space.


Assuntos
Nitritos , Plasmodium falciparum , Microscopia Crioeletrônica , Formiatos , Plasmodium falciparum/genética , Proteínas de Protozoários/genética
6.
Chem Rev ; 121(9): 5378-5416, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33211490

RESUMO

Multidrug resistant (MDR) bacteria are a global threat with many common infections becoming increasingly difficult to eliminate. While significant effort has gone into the development of potent biocides, the effectiveness of many first-line antibiotics has been diminished due to adaptive resistance mechanisms. Bacterial membrane proteins belonging to the resistance-nodulation-cell division (RND) superfamily play significant roles in mediating bacterial resistance to antimicrobials. They participate in multidrug efflux and cell wall biogenesis to transform bacterial pathogens into "superbugs" that are resistant even to last resort antibiotics. In this review, we summarize the RND superfamily of efflux transporters with a primary focus on the assembly and function of the inner membrane pumps. These pumps are critical for extrusion of antibiotics from the cell as well as the transport of lipid moieties to the outer membrane to establish membrane rigidity and stability. We analyze recently solved structures of bacterial inner membrane efflux pumps as to how they bind and transport their substrates. Our cumulative data indicate that these RND membrane proteins are able to utilize different oligomerization states to achieve particular activities, including forming MDR pumps and cell wall remodeling machineries, to ensure bacterial survival. This mechanistic insight, combined with simulated docking techniques, allows for the design and optimization of new efflux pump inhibitors to more effectively treat infections that today are difficult or impossible to cure.


Assuntos
Bactérias/metabolismo , Proteínas da Membrana Bacteriana Externa/química , Proteínas da Membrana Bacteriana Externa/metabolismo , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Animais , Antibacterianos/química , Antibacterianos/metabolismo , Antibacterianos/farmacologia , Bactérias/química , Bactérias/efeitos dos fármacos , Farmacorresistência Bacteriana Múltipla , Humanos , Simulação de Dinâmica Molecular , Relação Estrutura-Atividade
7.
PLoS Pathog ; 16(12): e1009119, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33290418

RESUMO

Infections caused by Gram-negative bacteria are difficult to fight because these pathogens exclude or expel many clinical antibiotics and host defense molecules. However, mammals have evolved a substantial immune arsenal that weakens pathogen defenses, suggesting the feasibility of developing therapies that work in concert with innate immunity to kill Gram-negative bacteria. Using chemical genetics, we recently identified a small molecule, JD1, that kills Salmonella enterica serovar Typhimurium (S. Typhimurium) residing within macrophages. JD1 is not antibacterial in standard microbiological media, but rapidly inhibits growth and curtails bacterial survival under broth conditions that compromise the outer membrane or reduce efflux pump activity. Using a combination of cellular indicators and super resolution microscopy, we found that JD1 damaged bacterial cytoplasmic membranes by increasing fluidity, disrupting barrier function, and causing the formation of membrane distortions. We quantified macrophage cell membrane integrity and mitochondrial membrane potential and found that disruption of eukaryotic cell membranes required approximately 30-fold more JD1 than was needed to kill bacteria in macrophages. Moreover, JD1 preferentially damaged liposomes with compositions similar to E. coli inner membranes versus mammalian cell membranes. Cholesterol, a component of mammalian cell membranes, was protective in the presence of neutral lipids. In mice, intraperitoneal administration of JD1 reduced tissue colonization by S. Typhimurium. These observations indicate that during infection, JD1 gains access to and disrupts the cytoplasmic membrane of Gram-negative bacteria, and that neutral lipids and cholesterol protect mammalian membranes from JD1-mediated damage. Thus, it may be possible to develop therapeutics that exploit host innate immunity to gain access to Gram-negative bacteria and then preferentially damage the bacterial cell membrane over host membranes.


Assuntos
Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Bactérias Gram-Negativas/efeitos dos fármacos , Infecções por Bactérias Gram-Negativas , Imunidade Inata , Animais , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Macrófagos/microbiologia , Lipídeos de Membrana , Camundongos , Camundongos Endogâmicos C57BL
8.
Proc Natl Acad Sci U S A ; 116(23): 11241-11246, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31113875

RESUMO

The cell envelope of Mycobacterium tuberculosis is notable for the abundance of mycolic acids (MAs), essential to mycobacterial viability, and of other species-specific lipids. The mycobacterial cell envelope is extremely hydrophobic, which contributes to virulence and antibiotic resistance. However, exactly how fatty acids and lipidic elements are transported across the cell envelope for cell-wall biosynthesis is unclear. Mycobacterial membrane protein Large 3 (MmpL3) is essential and required for transport of trehalose monomycolates (TMMs), precursors of MA-containing trehalose dimycolates (TDM) and mycolyl arabinogalactan peptidoglycan, but the exact function of MmpL3 remains elusive. Here, we report a crystal structure of Mycobacterium smegmatis MmpL3 at a resolution of 2.59 Å, revealing a monomeric molecule that is structurally distinct from all known bacterial membrane proteins. A previously unknown MmpL3 ligand, phosphatidylethanolamine (PE), was discovered inside this transporter. We also show, via native mass spectrometry, that MmpL3 specifically binds both TMM and PE, but not TDM, in the micromolar range. These observations provide insight into the function of MmpL3 and suggest a possible role for this protein in shuttling a variety of lipids to strengthen the mycobacterial cell wall.


Assuntos
Proteínas de Bactérias/metabolismo , Fatores Corda/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Fosfatidiletanolaminas/metabolismo , Transporte Biológico/fisiologia , Membrana Celular/metabolismo , Parede Celular/metabolismo , Mycobacterium smegmatis/metabolismo , Ácidos Micólicos/metabolismo
9.
Plant J ; 102(6): 1107-1126, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32168387

RESUMO

Microalgae and cyanobacteria contribute roughly half of the global photosynthetic carbon assimilation. Faced with limited access to CO2 in aquatic environments, which can vary daily or hourly, these microorganisms have evolved use of an efficient CO2 concentrating mechanism (CCM) to accumulate high internal concentrations of inorganic carbon (Ci ) to maintain photosynthetic performance. For eukaryotic algae, a combination of molecular, genetic and physiological studies using the model organism Chlamydomonas reinhardtii, have revealed the function and molecular characteristics of many CCM components, including active Ci uptake systems. Fundamental to eukaryotic Ci uptake systems are Ci transporters/channels located in membranes of various cell compartments, which together facilitate the movement of Ci from the environment into the chloroplast, where primary CO2 assimilation occurs. Two putative plasma membrane Ci transporters, HLA3 and LCI1, are reportedly involved in active Ci uptake. Based on previous studies, HLA3 clearly plays a meaningful role in HCO3- transport, but the function of LCI1 has not yet been thoroughly investigated so remains somewhat obscure. Here we report a crystal structure of the full-length LCI1 membrane protein to reveal LCI1 structural characteristics, as well as in vivo physiological studies in an LCI1 loss-of-function mutant to reveal the Ci species preference for LCI1. Together, these new studies demonstrate LCI1 plays an important role in active CO2 uptake and that LCI1 likely functions as a plasma membrane CO2 channel, possibly a gated channel.


Assuntos
Proteínas de Algas/metabolismo , Dióxido de Carbono/metabolismo , Membrana Celular/metabolismo , Chlamydomonas reinhardtii/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Algas/química , Proteínas de Membrana Transportadoras/química , Simulação de Dinâmica Molecular , Estrutura Terciária de Proteína
10.
J Biol Chem ; 294(43): 15711-15723, 2019 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-31471317

RESUMO

The mycobacterial cell envelope is crucial to host-pathogen interactions as a barrier against antibiotics and the host immune response. In addition, cell envelope lipids are mycobacterial virulence factors. Cell envelope lipid biosynthesis is the target of a number of frontline tuberculosis treatments and has been the focus of much research. However, the transport mechanisms by which these lipids reach the mycomembrane remain poorly understood. Many envelope lipids are exported from the cytoplasm to the periplasmic space via the mycobacterial membrane protein large (MmpL) family of proteins. In other bacteria, lipoproteins can contribute to outer membrane biogenesis through direct binding of substrates and/or protein-protein associations with extracytoplasmic biosynthetic enzymes. In this report, we investigate whether the lipoprotein LpqN plays a similar role in mycobacteria. Using a genetic two-hybrid approach, we demonstrate that LpqN interacts with periplasmic loop domains of the MmpL3 and MmpL11 transporters that export mycolic acid-containing cell envelope lipids. We observe that LpqN also interacts with secreted cell envelope biosynthetic enzymes such as Ag85A via pulldown assays. The X-ray crystal structures of LpqN and LpqN bound to dodecyl-trehalose suggest that LpqN directly binds trehalose monomycolate, the MmpL3 and Ag85A substrate. Finally, we observe altered lipid profiles of the ΔlpqN mutant during biofilm maturation, pointing toward a possible physiological role for the protein. The results of this study suggest that LpqN may act as a membrane fusion protein, connecting MmpL transporters with periplasmic proteins, and provide general insight into the role of lipoproteins in Mycobacterium tuberculosis cell envelope biogenesis.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Parede Celular/metabolismo , Lipoproteínas/química , Lipoproteínas/metabolismo , Mycobacterium tuberculosis/metabolismo , Sítios de Ligação , Biofilmes , Transporte Biológico , Vias Biossintéticas , Ligantes , Modelos Moleculares , Ácidos Micólicos/metabolismo , Ligação Proteica
11.
PLoS Pathog ; 14(6): e1007115, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29879224

RESUMO

Bacterial efflux pumps transport small molecules from the cytoplasm or periplasm outside the cell. Efflux pump activity is typically increased in multi-drug resistant (MDR) pathogens; chemicals that inhibit efflux pumps may have potential for antibiotic development. Using an in-cell screen, we identified three efflux pump modulators (EPMs) from a drug diversity library. The screening platform uses macrophages infected with the human Gram-negative pathogen Salmonella enterica (Salmonella) to identify small molecules that prevent bacterial replication or survival within the host environment. A secondary screen for hit compounds that increase the accumulation of an efflux pump substrate, Hoechst 33342, identified three small molecules with activity comparable to the known efflux pump inhibitor PAßN (Phe-Arg ß-naphthylamide). The three putative EPMs demonstrated significant antibacterial activity against Salmonella within primary and cell culture macrophages and within a human epithelial cell line. Unlike traditional antibiotics, the three compounds did not inhibit bacterial growth in standard microbiological media. The three compounds prevented energy-dependent efflux pump activity in Salmonella and bound the AcrB subunit of the AcrAB-TolC efflux system with KDs in the micromolar range. Moreover, the EPMs display antibacterial synergy with antimicrobial peptides, a class of host innate immune defense molecules present in body fluids and cells. The EPMs also had synergistic activity with antibiotics exported by AcrAB-TolC in broth and in macrophages and inhibited efflux pump activity in MDR Gram-negative ESKAPE clinical isolates. Thus, an in-cell screening approach identified EPMs that synergize with innate immunity to kill bacteria and have potential for development as adjuvants to antibiotics.


Assuntos
Antibacterianos/farmacologia , Carga Bacteriana/efeitos dos fármacos , Dipeptídeos/farmacologia , Ensaios de Triagem em Larga Escala , Macrófagos/efeitos dos fármacos , Salmonella enterica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Transporte Biológico , Células Cultivadas , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Macrófagos/microbiologia , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Testes de Sensibilidade Microbiana
12.
Proc Natl Acad Sci U S A ; 114(25): 6557-6562, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28584102

RESUMO

Strains of the Burkholderia cepacia complex (Bcc) are Gram-negative opportunisitic bacteria that are capable of causing serious diseases, mainly in immunocompromised individuals. Bcc pathogens are intrinsically resistant to multiple antibiotics, including ß-lactams, aminoglycosides, fluoroquinolones, and polymyxins. They are major pathogens in patients with cystic fibrosis (CF) and can cause severe necrotizing pneumonia, which is often fatal. Hopanoid biosynthesis is one of the major mechanisms involved in multiple antimicrobial resistance of Bcc pathogens. The hpnN gene of B. multivorans encodes an integral membrane protein of the HpnN family of transporters, which is responsible for shuttling hopanoids to the outer membrane. Here, we report crystal structures of B. multivorans HpnN, revealing a dimeric molecule with an overall butterfly shape. Each subunit of the transporter contains 12 transmembrane helices and two periplasmic loops that suggest a plausible pathway for substrate transport. Further analyses indicate that HpnN is capable of shuttling hopanoid virulence factors from the outer leaflet of the inner membrane to the periplasm. Taken together, our data suggest that the HpnN transporter is critical for multidrug resistance and cell wall remodeling in Burkholderia.


Assuntos
Complexo Burkholderia cepacia/química , Proteínas de Membrana Transportadoras/química , Cristalografia por Raios X/métodos , Periplasma/química , Fatores de Virulência/química
15.
Nature ; 470(7335): 558-62, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21350490

RESUMO

Gram-negative bacteria, such as Escherichia coli, expel toxic chemicals through tripartite efflux pumps that span both the inner and outer membrane. The three parts are an inner membrane, substrate-binding transporter; a membrane fusion protein; and an outer-membrane-anchored channel. The fusion protein connects the transporter to the channel within the periplasmic space. A crystallographic model of this tripartite efflux complex has been unavailable because co-crystallization of the various components of the system has proven to be extremely difficult. We previously described the crystal structures of both the inner membrane transporter CusA and the membrane fusion protein CusB of the CusCBA efflux system of E. coli. Here we report the co-crystal structure of the CusBA efflux complex, showing that the transporter (or pump) CusA, which is present as a trimer, interacts with six CusB protomers and that the periplasmic domain of CusA is involved in these interactions. The six CusB molecules seem to form a continuous channel. The affinity of the CusA and CusB interaction was found to be in the micromolar range. Finally, we have predicted a three-dimensional structure for the trimeric CusC outer membrane channel and developed a model of the tripartite efflux assemblage. This CusC(3)-CusB(6)-CusA(3) model shows a 750-kilodalton efflux complex that spans the entire bacterial cell envelope and exports Cu I and Ag I ions.


Assuntos
Proteínas de Escherichia coli/química , Escherichia coli/química , Proteínas de Membrana Transportadoras/química , Metais Pesados/metabolismo , Complexos Multiproteicos/química , Cobre/metabolismo , Cristalização , Cristalografia por Raios X , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Ligação Proteica , Multimerização Proteica , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Prata/metabolismo , Eletricidade Estática
16.
J Biol Chem ; 290(47): 28559-28574, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26396194

RESUMO

The mycobacterial cell wall is critical to the virulence of these pathogens. Recent work shows that the MmpL (mycobacterial membrane protein large) family of transporters contributes to cell wall biosynthesis by exporting fatty acids and lipidic elements of the cell wall. The expression of the Mycobacterium tuberculosis MmpL proteins is controlled by a complex regulatory network, including the TetR family transcriptional regulators Rv3249c and Rv1816. Here we report the crystal structures of these two regulators, revealing dimeric, two-domain molecules with architecture consistent with the TetR family of regulators. Buried extensively within the C-terminal regulatory domains of Rv3249c and Rv1816, we found fortuitous bound ligands, which were identified as palmitic acid (a fatty acid) and isopropyl laurate (a fatty acid ester), respectively. Our results suggest that fatty acids may be the natural ligands of these regulatory proteins. Using fluorescence polarization and electrophoretic mobility shift assays, we demonstrate the recognition of promoter and intragenic regions of multiple mmpL genes by these proteins. Binding of palmitic acid renders these regulators incapable of interacting with their respective operator DNAs, which will result in derepression of the corresponding mmpL genes. Taken together, these experiments provide new perspectives on the regulation of the MmpL family of transporters.


Assuntos
Proteínas de Bactérias/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Mycobacterium tuberculosis/metabolismo , Proteínas de Bactérias/química , Cristalografia por Raios X , Proteínas de Membrana Transportadoras/química , Conformação Proteica
17.
Nature ; 467(7314): 484-8, 2010 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-20865003

RESUMO

Gram-negative bacteria, such as Escherichia coli, frequently use tripartite efflux complexes in the resistance-nodulation-cell division (RND) family to expel various toxic compounds from the cell. The efflux system CusCBA is responsible for extruding biocidal Cu(I) and Ag(I) ions. No previous structural information was available for the heavy-metal efflux (HME) subfamily of the RND efflux pumps. Here we describe the crystal structures of the inner-membrane transporter CusA in the absence and presence of bound Cu(I) or Ag(I). These CusA structures provide new structural information about the HME subfamily of RND efflux pumps. The structures suggest that the metal-binding sites, formed by a three-methionine cluster, are located within the cleft region of the periplasmic domain. This cleft is closed in the apo-CusA form but open in the CusA-Cu(I) and CusA-Ag(I) structures, which directly suggests a plausible pathway for ion export. Binding of Cu(I) and Ag(I) triggers significant conformational changes in both the periplasmic and transmembrane domains. The crystal structure indicates that CusA has, in addition to the three-methionine metal-binding site, four methionine pairs-three located in the transmembrane region and one in the periplasmic domain. Genetic analysis and transport assays suggest that CusA is capable of actively picking up metal ions from the cytosol, using these methionine pairs or clusters to bind and export metal ions. These structures suggest a stepwise shuttle mechanism for transport between these sites.


Assuntos
Cobre/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Escherichia coli/química , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/metabolismo , Metionina/metabolismo , Prata/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Sítios de Ligação , Membrana Celular/metabolismo , Cobre/química , Cristalografia por Raios X , Citosol/metabolismo , Transporte de Íons , Modelos Biológicos , Modelos Moleculares , Periplasma/metabolismo , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Prata/química , Relação Estrutura-Atividade
18.
J Biol Chem ; 289(23): 16526-40, 2014 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-24737322

RESUMO

Recent work demonstrates that the MmpL (mycobacterial membrane protein large) transporters are dedicated to the export of mycobacterial lipids for cell wall biosynthesis. An MmpL transporter frequently works with an accessory protein, belonging to the MmpS (mycobacterial membrane protein small) family, to transport these key virulence factors. One such efflux system in Mycobacterium tuberculosis is the MmpS5-MmpL5 transporter. The expression of MmpS5-MmpL5 is controlled by the MarR-like transcriptional regulator Rv0678, whose open reading frame is located downstream of the mmpS5-mmpL5 operon. To elucidate the structural basis of Rv0678 regulation, we have determined the crystal structure of this regulator, to 1.64 Å resolution, revealing a dimeric two-domain molecule with an architecture similar to members of the MarR family of transcriptional regulators. Rv0678 is distinct from other MarR regulators in that its DNA-binding and dimerization domains are clustered together. These two domains seemingly cooperate to bind an inducing ligand that we identified as 2-stearoylglycerol, which is a fatty acid glycerol ester. The structure also suggests that the conformational change leading to substrate-mediated derepression is primarily caused by a rigid body rotational motion of the entire DNA-binding domain of the regulator toward the dimerization domain. This movement results in a conformational state that is incompatible with DNA binding. We demonstrate using electrophoretic mobility shift assays that Rv0678 binds to the mmpS5-mmpL5, mmpS4-mmpL4, and the mmpS2-mmpL2 promoters. Binding by Rv0678 was reversed upon the addition of the ligand. These findings provide new insight into the mechanisms of gene regulation in the MarR family of regulators.


Assuntos
Mycobacterium tuberculosis/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Cristalografia por Raios X , Primers do DNA , Dimerização , Dados de Sequência Molecular , Mycobacterium tuberculosis/química , Reação em Cadeia da Polimerase , Homologia de Sequência de Aminoácidos
19.
Nucleic Acids Res ; 40(18): 9340-55, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22821564

RESUMO

The Mmr multidrug efflux pump recognizes and actively extrudes a broad range of antimicrobial agents, and promotes the intrinsic resistance to these antimicrobials in Mycobacterium tuberculosis. The expression of Mmr is controlled by the TetR-like transcriptional regulator Rv3066, whose open reading frame is located downstream of the mmr operon. To understand the structural basis of Rv3066 regulation, we have determined the crystal structures of Rv3066, both in the absence and presence of bound ethidium, revealing an asymmetric homodimeric two-domain molecule with an entirely helical architecture. The structures underscore the flexibility and plasticity of the regulator essential for multidrug recognition. Comparison of the apo-Rv3066 and Rv3066-ethidium crystal structures suggests that the conformational changes leading to drug-mediated derepression is primarily due to a rigid body rotational motion within the dimer interface of the regulator. The Rv3066 regulator creates a multidrug-binding pocket, which contains five aromatic residues. The bound ethidium is found buried within the multidrug-binding site, where extensive aromatic stacking interactions seemingly govern the binding. In vitro studies reveal that the dimeric Rv3066 regulator binds to a 14-bp palindromic inverted repeat sequence in the nanomolar range. These findings provide new insight into the mechanisms of ligand binding and Rv3066 regulation.


Assuntos
Proteínas de Bactérias/química , Mycobacterium tuberculosis , Proteínas Repressoras/química , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Cromatografia em Gel , Pegada de DNA , Proteínas de Ligação a DNA/química , Farmacorresistência Bacteriana Múltipla , Ensaio de Desvio de Mobilidade Eletroforética , Etídio/química , Polarização de Fluorescência , Sequências Repetidas Invertidas , Proteínas de Membrana Transportadoras/genética , Modelos Moleculares , Simulação de Acoplamento Molecular , Mycobacterium smegmatis/efeitos dos fármacos , Mycobacterium smegmatis/genética , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/genética , Regiões Promotoras Genéticas , Conformação Proteica , Proteínas Repressoras/metabolismo , Transcrição Gênica/efeitos dos fármacos
20.
Microbiol Mol Biol Rev ; 88(2): e0019823, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38551344

RESUMO

SUMMARYInfectious bacteria have both intrinsic and acquired mechanisms to combat harmful biocides that enter the cell. Through adaptive pressures, many of these pathogens have become resistant to many, if not all, of the current antibiotics used today to treat these often deadly infections. One prominent mechanism is the upregulation of efflux systems, especially the resistance-nodulation-cell division class of exporters. These tripartite systems consist of an inner membrane transporter coupled with a periplasmic adaptor protein and an outer membrane channel to efficiently transport a diverse array of substrates from inside the cell to the extracellular space. Detailed mechanistic insight into how these inner membrane transporters recognize and shuttle their substrates can ultimately inform both new antibiotic and efflux pump inhibitor design. This review examines the structural basis of substrate recognition of these pumps and the molecular mechanisms underlying multidrug extrusion, which in turn mediate antimicrobial resistance in bacterial pathogens.


Assuntos
Antibacterianos , Proteínas de Membrana Transportadoras , Proteínas de Membrana Transportadoras/metabolismo , Antibacterianos/farmacologia , Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Divisão Celular , Farmacorresistência Bacteriana Múltipla , Transporte Biológico , Modelos Moleculares , Farmacorresistência Bacteriana
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA