Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 277
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 603(7901): 515-521, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35264792

RESUMO

Nitrogen availability is a growth-limiting factor in many habitats1, and the global nitrogen cycle involves prokaryotes and eukaryotes competing for this precious resource. Only some bacteria and archaea can fix elementary nitrogen; all other organisms depend on the assimilation of mineral or organic nitrogen. The nitrogen-rich compound guanidine occurs widely in nature2-4, but its utilization is impeded by pronounced resonance stabilization5, and enzymes catalysing hydrolysis of free guanidine have not been identified. Here we describe the arginase family protein GdmH (Sll1077) from Synechocystis sp. PCC 6803 as a Ni2+-dependent guanidine hydrolase. GdmH is highly specific for free guanidine. Its activity depends on two accessory proteins that load Ni2+ instead of the typical Mn2+ ions into the active site. Crystal structures of GdmH show coordination of the dinuclear metal cluster in a geometry typical for arginase family enzymes and allow modelling of the bound substrate. A unique amino-terminal extension and a tryptophan residue narrow the substrate-binding pocket and identify homologous proteins in further cyanobacteria, several other bacterial taxa and heterokont algae as probable guanidine hydrolases. This broad distribution suggests notable ecological relevance of guanidine hydrolysis in aquatic habitats.


Assuntos
Hidrolases , Synechocystis , Arginase/metabolismo , Proteínas de Bactérias/metabolismo , Guanidina/metabolismo , Hidrolases/metabolismo , Nitrogênio/metabolismo
2.
Nature ; 566(7742): 94-99, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30728519

RESUMO

Small molecules containing the N-nitroso group, such as the bacterial natural product streptozotocin, are prominent carcinogens1,2 and important cancer chemotherapeutics3,4. Despite the considerable importance of this functional group to human health, enzymes dedicated to the assembly of the N-nitroso unit have not been identified. Here we show that SznF, a metalloenzyme from the biosynthesis of streptozotocin, catalyses an oxidative rearrangement of the guanidine group of Nω-methyl-L-arginine to generate an N-nitrosourea product. Structural characterization and mutagenesis of SznF reveal two separate active sites that promote distinct steps in this transformation using different iron-containing metallocofactors. This biosynthetic reaction, which has little precedent in enzymology or organic synthesis, expands the catalytic capabilities of non-haem-iron-dependent enzymes to include N-N bond formation. We find that biosynthetic gene clusters that encode SznF homologues are widely distributed among bacteria-including environmental organisms, plant symbionts and human pathogens-which suggests an unexpectedly diverse and uncharacterized microbial reservoir of bioactive N-nitroso metabolites.


Assuntos
Metaloproteínas/metabolismo , Estreptozocina/biossíntese , Estreptozocina/química , Arginina/análogos & derivados , Domínio Catalítico/genética , Coenzimas/metabolismo , Cristalografia por Raios X , Guanidina/metabolismo , Ferro/metabolismo , Metaloproteínas/química , Metaloproteínas/genética , Modelos Moleculares , Família Multigênica , Compostos de Nitrosoureia/metabolismo , Streptomyces/enzimologia , Streptomyces/genética
3.
Mol Cell ; 65(2): 220-230, 2017 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-27989440

RESUMO

The guanidyl moiety is a component of fundamental metabolites, including the amino acid arginine, the energy carrier creatine, and the nucleobase guanine. Curiously, reports regarding the importance of free guanidine in biology are sparse, and no biological receptors that specifically recognize this compound have been previously identified. We report that many members of the ykkC motif RNA, the longest unresolved riboswitch candidate, naturally sense and respond to guanidine. This RNA is found throughout much of the bacterial domain of life, where it commonly controls the expression of proteins annotated as urea carboxylases and multidrug efflux pumps. Our analyses reveal that these proteins likely function as guanidine carboxylases and guanidine transporters, respectively. Furthermore, we demonstrate that bacteria are capable of endogenously producing guanidine. These and related findings demonstrate that free guanidine is a biologically relevant compound, and several gene families that can alleviate guanidine toxicity exist.


Assuntos
Bactérias/metabolismo , Proteínas de Bactérias/metabolismo , Carbono-Nitrogênio Ligases/metabolismo , Guanidina/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , RNA Bacteriano/metabolismo , RNA Mensageiro/metabolismo , Riboswitch , Bactérias/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Sítios de Ligação , Carbono-Nitrogênio Ligases/química , Carbono-Nitrogênio Ligases/genética , Regulação Bacteriana da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Proteínas de Membrana Transportadoras/química , Proteínas de Membrana Transportadoras/genética , Conformação de Ácido Nucleico , Motivos de Nucleotídeos , RNA Bacteriano/química , RNA Bacteriano/genética , RNA Mensageiro/química , RNA Mensageiro/genética , Especificidade por Substrato
4.
Magn Reson Med ; 91(4): 1512-1527, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38098305

RESUMO

PURPOSE: Guanidinium CEST is sensitive to metabolic changes and pH variation in ischemia, and it can offer advantages over conventional pH-sensitive amide proton transfer (APT) imaging by providing hyperintense contrast in stroke lesions. However, quantifying guanidinium CEST is challenging due to multiple overlapping components and a close frequency offset from water. This study aims to evaluate the applicability of a new rapid and model-free CEST quantification method using double saturation power, termed DSP-CEST, for isolating the guanidinium CEST effect from confounding factors in ischemia. To further reduce acquisition time, the DSP-CEST was combined with a quasi-steady state (QUASS) CEST technique to process non-steady-state CEST signals. METHODS: The specificity and accuracy of the DSP-CEST method in quantifying the guanidinium CEST effect were assessed by comparing simulated CEST signals with/without the contribution from confounding factors. The feasibility of this method for quantifying guanidinium CEST was evaluated in a rat model of global ischemia induced by cardiac arrest and compared to a conventional multiple-pool Lorentzian fit method. RESULTS: The DSP-CEST method was successful in removing all confounding components and quantifying the guanidinium CEST signal increase in ischemia. This suggests that the DSP-CEST has the potential to provide hyperintense contrast in stroke lesions. Additionally, the DSP-CEST was shown to be a rapid method that does not require the acquisition of the entire or a portion of the CEST Z-spectrum that is required in conventional model-based fitting approaches. CONCLUSION: This study highlights the potential of DSP-CEST as a valuable tool for rapid and specific detection of viable tissues.


Assuntos
Encéfalo , Acidente Vascular Cerebral , Ratos , Animais , Encéfalo/metabolismo , Imageamento por Ressonância Magnética/métodos , Guanidina/metabolismo , Roedores , Isquemia/diagnóstico por imagem , Isquemia/metabolismo , Amidas/metabolismo
5.
Nature ; 557(7706): 590-594, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769724

RESUMO

Potassium-sensitive hypokalaemic and normokalaemic periodic paralysis are inherited skeletal muscle diseases characterized by episodes of flaccid muscle weakness1,2. They are caused by single mutations in positively charged residues ('gating charges') in the S4 transmembrane segment of the voltage sensor of the voltage-gated sodium channel Nav1.4 or the calcium channel Cav1.11,2. Mutations of the outermost gating charges (R1 and R2) cause hypokalaemic periodic paralysis1,2 by creating a pathogenic gating pore in the voltage sensor through which cations leak in the resting state3,4. Mutations of the third gating charge (R3) cause normokalaemic periodic paralysis 5 owing to cation leak in both activated and inactivated states 6 . Here we present high-resolution structures of the model bacterial sodium channel NavAb with the analogous gating-charge mutations7,8, which have similar functional effects as in the human channels. The R2G and R3G mutations have no effect on the backbone structures of the voltage sensor, but they create an aqueous cavity near the hydrophobic constriction site that controls gating charge movement through the voltage sensor. The R3G mutation extends the extracellular aqueous cleft through the entire length of the activated voltage sensor, creating an aqueous path through the membrane. Conversely, molecular modelling shows that the R2G mutation creates a continuous aqueous path through the membrane only in the resting state. Crystal structures of NavAb(R2G) in complex with guanidinium define a potential drug target site. Molecular dynamics simulations illustrate the mechanism of Na+ permeation through the mutant gating pore in concert with conformational fluctuations of the gating charge R4. Our results reveal pathogenic mechanisms of periodic paralysis at the atomic level and suggest designs of drugs that may prevent ionic leak and provide symptomatic relief from hypokalaemic and normokalaemic periodic paralysis.


Assuntos
Ativação do Canal Iônico , Canal de Sódio Disparado por Voltagem NAV1.4/química , Canal de Sódio Disparado por Voltagem NAV1.4/metabolismo , Paralisias Periódicas Familiares/metabolismo , Sítios de Ligação , Condutividade Elétrica , Guanidina/metabolismo , Humanos , Paralisia Periódica Hipopotassêmica/genética , Paralisia Periódica Hipopotassêmica/metabolismo , Ativação do Canal Iônico/genética , Simulação de Dinâmica Molecular , Mutação , Canal de Sódio Disparado por Voltagem NAV1.4/genética , Paralisias Periódicas Familiares/genética , Sódio/metabolismo , Termodinâmica
6.
Ecotoxicol Environ Saf ; 272: 116084, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38350217

RESUMO

Polyhexamethylene guanidine (PHMG) is manufactured and applied extensively due to its superior disinfectant capabilities. However, the inhalatory exposure to PHMG aerosols is increasingly recognized as a potential instigator of pulmonary fibrosis, prompting an urgent call for elucidation of the underlying pathophysiological mechanisms. Within this context, alveolar macrophages play a pivotal role in the primary immune defense in the respiratory tract. Dysregulated lipid metabolism within alveolar macrophages leads to the accumulation of foam cells, a process that is intimately linked with the pathogenesis of pulmonary fibrosis. Therefore, this study examines PHMG's effects on alveolar macrophage foaminess and its underlying mechanisms. We conducted a 3-week inhalation exposure followed by a 3-week recovery period in C57BL/6 J mice using a whole-body exposure system equipped with a disinfection aerosol generator (WESDAG). The presence of lipid-laden alveolar macrophages and downregulation of pulmonary tissue lipid transport proteins ABCA1 and ABCG1 were observed in mice. In cell culture models involving lipid-loaded macrophages, we demonstrated that PHMG promotes foam cell formation by inhibiting lipid efflux in mouse alveolar macrophages. Furthermore, PHMG-induced foam cells were found to promote an increase in the release of TGF-ß1, fibronectin deposition, and collagen remodeling. In vivo interventions were subsequently implemented on mice exposed to PHMG aerosols, aiming to restore macrophage lipid efflux function. Remarkably, this intervention demonstrated the potential to retard the progression of pulmonary fibrosis. In conclusion, this study underscores the pivotal role of macrophage foaming in the pathogenesis of PHMG disinfectants-induced pulmonary fibrosis. Moreover, it provides compelling evidence to suggest that the regulation of macrophage efflux function holds promise for mitigating the progression of pulmonary fibrosis, thereby offering novel insights into the mechanisms underlying inhaled PHMG disinfectants-induced pulmonary fibrosis.


Assuntos
Desinfetantes , Fibrose Pulmonar , Camundongos , Animais , Fibrose Pulmonar/metabolismo , Guanidina/toxicidade , Guanidina/metabolismo , Camundongos Endogâmicos C57BL , Aerossóis e Gotículas Respiratórios , Pulmão , Guanidinas/metabolismo , Macrófagos , Desinfetantes/farmacologia , Lipídeos
7.
Magn Reson Med ; 89(1): 177-191, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36063502

RESUMO

PURPOSE: To extract guanidinium (Guan) and amide CEST on the human brain at 3 T MRI with the high spectral resolution (HSR) CEST combined with the polynomial Lorentzian line-shape fitting (PLOF). METHODS: Continuous wave (cw) turbo spin-echo (TSE) CEST was implemented to obtain the optimum saturation parameters. Both Guan and amide CEST peaks were extracted and quantified using the PLOF method. The NMR spectra on the egg white phantoms were acquired to reveal the fitting range and the contributions to the amide and GuanCEST. Two types of CEST approaches, including cw gradient- and spin-echo (cwGRASE) and steady state EPI (ssEPI), were implemented to acquire multi-slice HSR-CEST. RESULTS: GuanCEST can be extracted with the PLOF method at 3 T, and the optimum B 1 = 0.6 µ T $$ {\mathrm{B}}_1=0.6\kern0.2em \upmu \mathrm{T} $$ was determined for GuanCEST in white matter (WM) and 1.0 µT in gray matter (GM). The optimum B1  = 0.8-1 µT was found for amideCEST. AmideCEST is lower in both WM and GM collected with ssEPI compared to those by cwGRASE (ssEPI = [1.27-1.63]%; cwGRASE = [2.19-2.25]%). The coefficients of variation (COV) of the amide and Guan CEST in both WM and GM for ssEPI (COV: 28.6-33.4%) are significantly higher than those of cwGRASE (COV: 8.6-18.8%). Completely different WM/GM contrasts for Guan and amide CEST were observed between ssEPI and cwGRASE. The amideCEST was found to have originated from the unstructured amide protons as suggested by the NMR spectrum of the unfolded proteins in egg white. CONCLUSION: Guan and amide CEST mapping can be achieved by the HSR-CEST at 3 T combing with the PLOF method.


Assuntos
Amidas , Encéfalo , Humanos , Guanidina/metabolismo , Amidas/química , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Imageamento por Ressonância Magnética/métodos , Substância Cinzenta
8.
RNA Biol ; 20(1): 10-19, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36548032

RESUMO

Riboswitches are regulatory RNAs that specifically bind a small molecule or ion. Like metabolite-binding proteins, riboswitches can evolve new ligand specificities, and some examples of this phenomenon have been validated. As part of work based on comparative genomics to discover novel riboswitches, we encountered a candidate riboswitch with striking similarities to the recently identified guanidine-IV riboswitch. This candidate riboswitch, the Gd4v motif, is predicted in four distinct bacterial phyla, thus almost as widespread as the guanidine-IV riboswitch. Bioinformatic and experimental analysis suggest that the Gd4v motif is a riboswitch that binds a ligand other than guanidine. It is found associated with gene classes that differ from genes regulated by confirmed guanidine riboswitches. In inline-probing assays, we showed that free guanidine binds only weakly to one of the tested sequences of the variant. Further tested compounds did not show binding, attenuation of transcription termination, or activation of a genetic reporter construct. We characterized an N-acetyltransferase frequently associated with the Gd4v motif and compared its substrate preference to an N-acetyltransferase that occurs under control of guanidine-IV riboswitches. The substrates of this Gd4v-motif-associated enzyme did not show activity for Gd4v RNA binding or transcription termination. Hence, the ligand of the candidate riboswitch motif remains unidentified. The variant RNA motif is predominantly found in gut metagenome sequences, hinting at a ligand that is highly relevant in this environment. This finding is a first step to determining the identity of this unknown ligand, and understanding how guanidine-IV-riboswitch-like structures can evolve to bind different ligands.


Assuntos
Riboswitch , Guanidina/química , Guanidina/metabolismo , Conformação de Ácido Nucleico , Ligantes , Guanidinas/metabolismo , Acetiltransferases/genética , Acetiltransferases/metabolismo
9.
J Cell Mol Med ; 26(14): 3816-3827, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35678269

RESUMO

Radix puerariae, a traditional Chinese herbal medication, has been used to treat patients with diabetic kidney disease (DKD). Our previous studies demonstrated that puerarin, the active compound of radix puerariae, improves podocyte injury in type 1 DKD mice. However, the direct molecular target of puerarin and its underlying mechanisms in DKD remain unknown. In this study, we confirmed that puerarin also improved DKD in type 2 diabetic db/db mice. Through RNA-sequencing odf isolated glomeruli, we found that differentially expressed genes (DEGs) that were altered in the glomeruli of these diabetic mice but reversed by puerarin treatment were involved mostly in oxidative stress, inflammatory and fibrosis. Further analysis of these reversed DEGs revealed protein kinase A (PKA) was among the top pathways. By utilizing the drug affinity responsive target stability method combined with mass spectrometry analysis, we identified guanine nucleotide-binding protein Gi alpha-1 (Gnai1) as the direct binding partner of puerarin. Gnai1 is an inhibitor of cAMP production which is known to have protection against podocyte injury. In vitro, we showed that puerarin not only interacted with Gnai1 but also increased cAMP production in human podocytes and mouse diabetic kidney in vivo. Puerarin also enhanced CREB phosphorylation, a downstream transcription factor of cAMP/PKA. Overexpression of CREB reduced high glucose-induced podocyte apoptosis. Inhibition of PKA by Rp-cAMP also diminished the effects of puerarin on high glucose-induced podocyte apoptosis. We conclude that the renal protective effects of puerarin are likely through inhibiting Gnai1 to activate cAMP/PKA/CREB pathway in podocytes.


Assuntos
Diabetes Mellitus Experimental , Nefropatias Diabéticas , Podócitos , Animais , Apoptose , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/metabolismo , Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/farmacologia , Glucose/metabolismo , Guanidina/metabolismo , Guanidina/farmacologia , Guanidina/uso terapêutico , Humanos , Isoflavonas , Camundongos , Nucleotídeos/metabolismo , Podócitos/metabolismo
10.
Mol Microbiol ; 116(1): 200-210, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33590553

RESUMO

Guanidine is sensed by at least four different classes of riboswitches that are widespread in bacteria. However, only very few insights into physiological roles of guanidine exist. Genes predominantly regulated by guanidine riboswitches are Gdx transporters exporting the compound from the bacterial cell. In addition, urea/guanidine carboxylases and associated hydrolases and ABC transporters are often found combined in guanidine-inducible operons. We noted that the associated ABC transporters are configured to function as importers, challenging the current view that riboswitches solely control the detoxification of guanidine in bacteria. We demonstrate that the carboxylase pathway enables utilization of guanidine as sole nitrogen source. We isolated three enterobacteria (Raoultella terrigena, Klebsiella michiganensis, and Erwinia rhapontici) that utilize guanidine efficiently as N-source. Proteome analyses show that the expression of a carboxylase, associated hydrolases and transport genes is strongly induced by guanidine. Finding two urea/guanidine carboxylase enzymes in E. rhapontici, we demonstrate that the riboswitch-controlled carboxylase displays specificity toward guanidine, whereas the other enzyme prefers urea. We characterize the distribution of riboswitch-associated carboxylases and Gdx exporters in bacterial habitats by analyzing available metagenome data. The findings represent a paradigm shift from riboswitch-controlled detoxification of guanidine to the uptake and assimilation of this enigmatic nitrogen-rich compound.


Assuntos
Enterobacteriaceae/metabolismo , Erwinia/metabolismo , Guanidina/metabolismo , Klebsiella/metabolismo , Riboswitch/genética , Carbono-Nitrogênio Ligases/genética , Metabolismo Energético/genética , Regulação Bacteriana da Expressão Gênica/genética , Hidrolases/metabolismo , Proteínas de Membrana Transportadoras/genética
11.
Chembiochem ; 23(3): e202100564, 2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-34847270

RESUMO

Riboswitches are regulatory RNA elements that undergo functionally important allosteric conformational switching upon binding of specific ligands. The here investigated guanidine-II riboswitch binds the small cation, guanidinium, and forms a kissing loop-loop interaction between its P1 and P2 hairpins. We investigated the structural changes to support previous studies regarding the binding mechanism. Using NMR spectroscopy, we confirmed the structure as observed in crystal structures and we characterized the kissing loop interaction upon addition of Mg2+ and ligand for the riboswitch aptamer from Escherichia coli. We further investigated closely related mutant constructs providing further insight into functional differences between the two (different) hairpins P1 and P2. Formation of intermolecular interactions were probed by small-angle X-ray scattering (SAXS) and NMR DOSY data. All data are consistent and show the formation of oligomeric states of the riboswitch induced by Mg2+ and ligand binding.


Assuntos
Escherichia coli/química , Guanidina/química , Guanidina/metabolismo , Espectroscopia de Ressonância Magnética , Riboswitch , Espalhamento a Baixo Ângulo , Difração de Raios X
12.
Bioorg Med Chem Lett ; 76: 129017, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36209968

RESUMO

Despite the enormous potential of siRNAs to transcriptionally downregulate disease causing proteins in many genetic diseases, efficient delivery and endosomal escape are the two bottlenecks that have resulted in only a handful of FDA approved drugs. In this report, we have successfully delivered siRNA against Nanog with the help of pentafluorobenzyl modified Internal Oligo-guanidinium transporter (IGT) that has previously shown promising results in peptide and antisense morpholino delivery. Nanog downregulation in prostate cancer cell line DU145 in serum containing media led to suppression of associated proteins such as KLF4, FAK and cMyc and also enhanced the chemosensitivity of Epirubicin, an anthracycline based drug, in DU145 cells by associated MDR-1 downregulation in vitro. These results show that IGT is a promising candidate for siRNA delivery and its conjugation with stable siRNAs could enhance the chemotherapeutic efficiency of siRNAs alone and in combination with small molecule-based drugs.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Epirubicina , Proteína Homeobox Nanog , Proteínas de Transporte de Cátions Orgânicos , Neoplasias da Próstata , RNA Interferente Pequeno , Humanos , Masculino , Linhagem Celular Tumoral , Epirubicina/farmacologia , Guanidina/metabolismo , Morfolinos , Proteína Homeobox Nanog/genética , Peptídeos/administração & dosagem , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , RNA Interferente Pequeno/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/genética
13.
Nucleic Acids Res ; 48(22): 12889-12899, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33237283

RESUMO

Riboswitches are RNAs that specifically sense a small molecule and regulate genes accordingly. The recent discovery of guanidine-binding riboswitches revealed the biological significance of this compound, and uncovered genes related to its biology. For example, certain sugE genes encode guanidine exporters and are activated by the riboswitches to reduce toxic levels of guanidine in the cell. In order to study guanidine biology and riboswitches, we applied a bioinformatics strategy for discovering additional guanidine riboswitches by searching for new candidate motifs associated with sugE genes. Based on in vitro and in vivo experiments, we determined that one of our six best candidates is a new structural class of guanidine riboswitches. The expression of a genetic reporter was induced 80-fold in response to addition of 5 mM guanidine in Staphylococcus aureus. This new class, called the guanidine-IV riboswitch, reveals additional guanidine-associated protein domains that are extremely rarely or never associated with previously established guanidine riboswitches. Among these protein domains are two transporter families that are structurally distinct from SugE, and could represent novel types of guanidine exporters. These results establish a new metabolite-binding RNA, further validate a bioinformatics method for finding riboswitches and suggest substrate specificities for as-yet uncharacterized transporter proteins.


Assuntos
Proteínas de Escherichia coli/genética , Proteínas de Membrana/genética , Chaperonas Moleculares/genética , RNA Bacteriano/genética , RNA/genética , Riboswitch/genética , Biologia Computacional , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/genética , Guanidina/metabolismo , Proteínas de Membrana Transportadoras/genética , Conformação de Ácido Nucleico , Domínios Proteicos/genética , Staphylococcus aureus/genética
14.
Nucleic Acids Res ; 48(18): 10518-10526, 2020 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-32857846

RESUMO

Riboswitches regulate genes by adopting different structures in responds to metabolite binding. The guanidine-II riboswitch is the smallest representative of the ykkC class with the mechanism of its function being centred on the idea that its two stem loops P1 and P2 form a kissing hairpin interaction upon binding of guanidinium (Gdm+). This mechanism is based on in-line probing experiments with the full-length riboswitch and crystal structures of the truncated stem loops P1 and P2. However, the crystal structures reveal only the formation of the homodimers P1 | P1 and P2 | P2 but not of the proposed heterodimer P1 | P2. Here, site-directed spin labeling (SDSL) in combination with Pulsed Electron-Electron Double Resonance (PELDOR or DEER) is used to study their structures in solution and how they change upon binding of Gdm+. It is found that both hairpins adopt different structures in solution and that binding of Gdm+ does indeed lead to the formation of the heterodimer but alongside the homodimers in a statistical 1:2:1 fashion. These results do thus support the proposed switching mechanism.


Assuntos
Guanidina/química , Metaboloma/genética , Conformação de Ácido Nucleico , Riboswitch/genética , Simulação por Computador , Cristalografia por Raios X , Dimerização , Espectroscopia de Ressonância de Spin Eletrônica , Escherichia coli/genética , Escherichia coli/ultraestrutura , Guanidina/metabolismo , Ligação de Hidrogênio , Sequências Repetidas Invertidas/genética , Ligantes , Ligação Proteica/genética , Riboswitch/efeitos dos fármacos , Marcadores de Spin
15.
J Am Chem Soc ; 143(27): 10083-10087, 2021 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-34181406

RESUMO

Guanidine prenylation is an outstanding modification in alkaloid and peptide biosynthesis, but its enzymatic basis has remained elusive. We report the isolation of argicyclamides, a new class of cyanobactins with unique mono- and bis-prenylations on guanidine moieties, from Microcystis aeruginosa NIES-88. The genetic basis of argicyclamide biosynthesis was established by the heterologous expression and in vitro characterization of biosynthetic enzymes including AgcF, a new guanidine prenyltransferase. This study provides important insight into the biosynthesis of prenylated guanidines and offers a new toolkit for peptide modification.


Assuntos
Guanidina/química , Guanidina/metabolismo , Microcystis/metabolismo , Peptídeos Cíclicos/química , Peptídeos Cíclicos/metabolismo , Estrutura Molecular , Prenilação
16.
Biochem Biophys Res Commun ; 577: 110-115, 2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34509722

RESUMO

Phosphoryl guanidine oligonucleotides (PGOs) are promising uncharged analogs of nucleic acids and are used in a variety of applications. The importance of hydration is frequently ignored during the design of modified nucleic acid probes. Such hydrophobic modifications (phosphoryl guanidine) are expected to have a significant impact on the structure and thermal stability of the affected oligo with complementary nucleic acids. Here we aimed to investigate (by the osmotic stress method) hydration changes upon the formation of a duplex of a PGO with complementary DNA. According to our results, the presence of phosphoryl guanidines in one or both strands of a duplex only minimally affects hydration alterations under crowding conditions. The secondary structure of native and modified duplexes did not change significantly in the presence of ethanol, ethylene glycol, polyethylene glycol 200, or polyethylene glycol 1000. After the addition of a cosolvent, the thermodynamic stability of the PGO complexes changed in the same manner as that seen in a corresponding DNA duplex. The findings reported here and our previous studies form the basis for efficient use of PGOs in basic research and a variety of applications.


Assuntos
DNA/química , Guanidina/química , Conformação de Ácido Nucleico , Hibridização de Ácido Nucleico/métodos , Oligonucleotídeos/química , Termodinâmica , Dicroísmo Circular/métodos , DNA/genética , DNA/metabolismo , Etanol/química , Guanidina/metabolismo , Modelos Moleculares , Simulação de Dinâmica Molecular , Desnaturação de Ácido Nucleico , Ácidos Nucleicos/química , Ácidos Nucleicos/genética , Ácidos Nucleicos/metabolismo , Oligonucleotídeos/genética , Oligonucleotídeos/metabolismo , Polietilenoglicóis/química , Soluções/química
17.
Appl Environ Microbiol ; 87(11)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33741630

RESUMO

The widely prescribed pharmaceutical metformin and its main metabolite, guanylurea, are currently two of the most common contaminants in surface and wastewater. Guanylurea often accumulates and is poorly, if at all, biodegraded in wastewater treatment plants. This study describes Pseudomonas mendocina strain GU, isolated from a municipal wastewater treatment plant, using guanylurea as its sole nitrogen source. The genome was sequenced with 36-fold coverage and mined to identify guanylurea degradation genes. The gene encoding the enzyme initiating guanylurea metabolism was expressed, and the enzyme was purified and characterized. Guanylurea hydrolase, a newly described enzyme, was shown to transform guanylurea to one equivalent (each) of ammonia and guanidine. Guanidine also supports growth as a sole nitrogen source. Cell yields from growth on limiting concentrations of guanylurea revealed that metabolism releases all four nitrogen atoms. Genes encoding complete metabolic transformation were identified bioinformatically, defining the pathway as follows: guanylurea to guanidine to carboxyguanidine to allophanate to ammonia and carbon dioxide. The first enzyme, guanylurea hydrolase, is a member of the isochorismatase-like hydrolase protein family, which includes biuret hydrolase and triuret hydrolase. Although homologs, the three enzymes show distinct substrate specificities. Pairwise sequence comparisons and the use of sequence similarity networks allowed fine structure discrimination between the three homologous enzymes and provided insights into the evolutionary origins of guanylurea hydrolase.IMPORTANCE Metformin is a pharmaceutical most prescribed for type 2 diabetes and is now being examined for potential benefits to COVID-19 patients. People taking the drug pass it largely unchanged, and it subsequently enters wastewater treatment plants. Metformin has been known to be metabolized to guanylurea. The levels of guanylurea often exceed that of metformin, leading to the former being considered a "dead-end" metabolite. Metformin and guanylurea are water pollutants of emerging concern, as they persist to reach nontarget aquatic life and humans, the latter if it remains in treated water. The present study has identified a Pseudomonas mendocina strain that completely degrades guanylurea. The genome was sequenced, and the genes involved in guanylurea metabolism were identified in three widely separated genomic regions. This knowledge advances the idea that guanylurea is not a dead-end product and will allow for bioinformatic identification of the relevant genes in wastewater treatment plant microbiomes and other environments subjected to metagenomic sequencing.


Assuntos
Proteínas de Bactérias/metabolismo , Guanidina/análogos & derivados , Hidrolases/metabolismo , Redes e Vias Metabólicas , Metformina/metabolismo , Ureia/análogos & derivados , Poluentes Químicos da Água/metabolismo , Amônia/metabolismo , Proteínas de Bactérias/genética , Biodegradação Ambiental , Biomineralização , Genoma Bacteriano/genética , Guanidina/metabolismo , Hidrolases/genética , Família Multigênica , Pseudomonas mendocina/genética , Pseudomonas mendocina/isolamento & purificação , Pseudomonas mendocina/metabolismo , Especificidade por Substrato , Ureia/metabolismo , Águas Residuárias/microbiologia
18.
Proc Natl Acad Sci U S A ; 115(12): 3060-3065, 2018 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-29507227

RESUMO

The small multidrug resistance (SMR) family of membrane proteins is prominent because of its rare dual topology architecture, simplicity, and small size. Its best studied member, EmrE, is an important model system in several fields related to membrane protein biology, from evolution to mechanism. But despite decades of work on these multidrug transporters, the native function of the SMR family has remained a mystery, and many highly similar SMR homologs do not transport drugs at all. Here we establish that representative SMR proteins, selected from each of the major clades in the phylogeny, function as guanidinium ion exporters. Drug-exporting SMRs are all clustered in a single minority clade. Using membrane transport experiments, we show that these guanidinium exporters, which we term Gdx, are very selective for guanidinium and strictly and stoichiometrically couple its export with the import of two protons. These findings draw important mechanistic distinctions with the notably promiscuous and weakly coupled drug exporters like EmrE.


Assuntos
Antiporters/metabolismo , Proteínas de Bactérias/metabolismo , Guanidina/metabolismo , Sequência de Aminoácidos , Antiporters/química , Bactérias/química , Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Transporte Biológico , Evolução Molecular , Filogenia , Riboswitch
19.
J Bacteriol ; 202(23)2020 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-32928929

RESUMO

Members of the small multidrug resistance (SMR) efflux pump family known as SugE (recently renamed Gdx) are known for their narrow substrate selectivity to small guanidinium (Gdm+) compounds and disinfectant quaternary ammonium compounds (QACs). Gdx members have been identified on multidrug resistance plasmids in Gram-negative bacilli, but their functional role remains unclear, as few have been characterized. Here, we conducted a survey of sequenced proteobacterial plasmids that encoded one or more SugE/Gdx sequences in an effort to (i) identify the most frequently represented Gdx member(s) on these plasmids and their sequence diversity, (ii) verify if Gdx sequences possess a Gdm+ riboswitch that regulates their translation similarly to chromosomally encoded Gdx members, and (iii) determine the antimicrobial susceptibility profile of the most predominate Gdx member to various QACs and antibiotics in Escherichia coli strains BW25113 and KAM32. The results of this study determined 14 unique SugE sequences, but only one Gdx sequence, annotated as "SugE(p)," predominated among the >140 plasmids we surveyed. Enterobacterales plasmids carrying sugE(p) possessed a guanidine II riboswitch similar to the upstream region of E. coligdx Cloning and expression of sugE(p), gdx, and emrE sequences into a low-copy-number expression vector (pMS119EH) revealed significant increases in QAC resistance to a limited range of detergent-like QACs only when gdx and sugE(p) transformants were grown as biofilms. These findings suggest that sugE(p) presence on proteobacterial plasmids may be driven by species that frequently encounter Gdm+ and QAC exposure.IMPORTANCE This study characterized the function of antimicrobial-resistant phenotypes attributed to plasmid-encoded guanidinium-selective small multidrug resistance (Gdm/SugE) efflux pumps. These sequences are frequently monitored as biocide resistance markers in antimicrobial resistance surveillance studies. Our findings reveal that enterobacterial gdm sequences transmitted on plasmids possess a guanidine II riboswitch, which restricts transcript translation in the presence of guanidinium. Cloning and overexpression of this gdm sequence revealed that it confers higher resistance to quaternary ammonium compound (QAC) disinfectants (which possess guanidium moieties) when grown as biofilms. Since biofilms are commonly eradicated with QAC-containing compounds, the presence of this gene on plasmids and its biofilm-specific resistance are a growing concern for clinical and food safety prevention measures.


Assuntos
Biofilmes/efeitos dos fármacos , Desinfetantes/farmacologia , Escherichia coli/efeitos dos fármacos , Guanidina/metabolismo , Plasmídeos/genética , Proteobactérias/genética , Compostos de Amônio Quaternário/farmacologia , Riboswitch/efeitos dos fármacos , Farmacorresistência Bacteriana/efeitos dos fármacos , Escherichia coli/genética , Escherichia coli/fisiologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Testes de Sensibilidade Microbiana , Plasmídeos/metabolismo
20.
Biochemistry ; 59(35): 3258-3270, 2020 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-32786413

RESUMO

Free guanidine is increasingly recognized as a relevant molecule in biological systems. Recently, it was reported that urea carboxylase acts preferentially on guanidine, and consequently, it was considered to participate directly in guanidine biodegradation. Urea carboxylase combines with allophanate hydrolase to comprise the activity of urea amidolyase, an enzyme predominantly found in bacteria and fungi that catalyzes the carboxylation and subsequent hydrolysis of urea to ammonia and carbon dioxide. Here, we demonstrate that urea carboxylase and allophanate hydrolase from Pseudomonas syringae are insufficient to catalyze the decomposition of guanidine. Rather, guanidine is decomposed to ammonia through the combined activities of urea carboxylase, allophanate hydrolase, and two additional proteins of the DUF1989 protein family, expansively annotated as urea carboxylase-associated family proteins. These proteins comprise the subunits of a heterodimeric carboxyguanidine deiminase (CgdAB), which hydrolyzes carboxyguanidine to N-carboxyurea (allophanate). The genes encoding CgdAB colocalize with genes encoding urea carboxylase and allophanate hydrolase. However, 25% of urea carboxylase genes, including all fungal urea amidolyases, do not colocalize with cgdAB. This subset of urea carboxylases correlates with a notable Asp to Asn mutation in the carboxyltransferase active site. Consistent with this observation, we demonstrate that fungal urea amidolyase retains a strong substrate preference for urea. The combined activities of urea carboxylase, carboxyguanidine deiminase and allophanate hydrolase represent a newly recognized pathway for the biodegradation of guanidine. These findings reinforce the relevance of guanidine as a biological metabolite and reveal a broadly distributed group of enzymes that act on guanidine in bacteria.


Assuntos
Guanidina/metabolismo , Hidrolases/metabolismo , Nitrogênio/metabolismo , Pseudomonas syringae/enzimologia , Ureia/metabolismo , Alofanato Hidrolase/química , Alofanato Hidrolase/metabolismo , Amônia/metabolismo , Carbono-Nitrogênio Ligases/química , Carbono-Nitrogênio Ligases/metabolismo , Catálise , Citrulinação/fisiologia , Hidrolases/química , Redes e Vias Metabólicas/fisiologia , Anotação de Sequência Molecular/normas , Subunidades Proteicas/química , Subunidades Proteicas/metabolismo , Pseudomonas syringae/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA