Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 704
Filtrar
1.
J Bioenerg Biomembr ; 56(5): 573-587, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39254861

RESUMO

Cysteine desulfurase (NFS1) is highly expressed in a variety of tumors, which is closely related to ferroptosis of tumor cells and affects prognosis. The relationship between NFS1 and the development of gastric cancer (GC) remains unknown. Here we showed that NFS1 expression was significantly higher in GC tissues compared to adjacent normal tissues. Patients with high expression of NFS1 in GC tissues had a lower overall survival rate than those with low expression. NFS1 was highly expressed in cultured GC cells compared to normal gastric cells. Knockdown of NFS1 expression reduced the viability, migration and invasion of GC cells. In cultured GC cells, NFS1 deficiency promoted ferroptosis. Mechanistically, NFS1 inhibited ferroptosis by upregulating the signal transduction and activator of transcription 3 (STAT3) signaling pathway in cultured GC cells. NFS1 knockdown using siRNA inhibited the STAT3 pathway, reduced the expression of glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11), and elevated intracellular levels of reactive oxygen species (ROS), ferrous ion (Fe2+), and malondialdehyde (MDA) in cultured GC cells. A specific STAT3 activator significantly reversed the inhibitory effect of NFS1 deficiency on ferroptosis in cultured GC cells. These in vitro results were further confirmed by experiments in vivo using a mouse xenograft tumor model. Collectively, THESE RESULTS INDICATE THAT NFS1 is overexpressed in human GC tissues and correlated with prognosis. NFS1 inhibits ferroptosis by activating the STAT3 pathway in GC cells. These results suggest that NFS1 may be a potential prognostic biomarker and therapeutic target to treat GC.


Assuntos
Ferroptose , Fator de Transcrição STAT3 , Neoplasias Gástricas , Ferroptose/fisiologia , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Humanos , Fator de Transcrição STAT3/metabolismo , Camundongos , Animais , Transdução de Sinais , Linhagem Celular Tumoral , Liases de Carbono-Enxofre/metabolismo , Masculino , Feminino , Camundongos Nus
2.
Arch Microbiol ; 206(10): 407, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39297992

RESUMO

Biofilm is the primary cause of persistent infections caused by Streptococcus suis (S. suis). Metabolism and AI-2 quorum sensing are intricately linked to S. suis biofilm formation. Although the role of the AI-2 quorum sensing luxS gene in S. suis biofilm has been reported, its specific regulatory mechanism remains unclear. This study explored the differences in biofilm formation and monosaccharide metabolism among the wild type (WT), luxS mutant (ΔluxS) and complement strain (CΔluxS), and Galleria mellonella larvae were used to access the effect of luxS gene deletion on the virulence of S. suis in different monosaccharide medias. The results indicated that deletion of the luxS gene further compromised the monosaccharide metabolism of S. suis, impacting its growth in media with fructose, galactose, rhamnose, and mannose as the sole carbon sources. However, no significant impact was observed in media with glucose and N-acetylglucosamine. This deletion also weakened EPS synthesis, thereby diminishing the biofilm formation capacity of S. suis. Additionally, the downregulation of adhesion gene expression due to luxS gene deletion was found to be independent of the monosaccharide medias of S. suis.


Assuntos
Proteínas de Bactérias , Biofilmes , Liases de Carbono-Enxofre , Monossacarídeos , Percepção de Quorum , Streptococcus suis , Biofilmes/crescimento & desenvolvimento , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Streptococcus suis/genética , Streptococcus suis/metabolismo , Streptococcus suis/crescimento & desenvolvimento , Percepção de Quorum/genética , Monossacarídeos/metabolismo , Animais , Regulação Bacteriana da Expressão Gênica , Deleção de Genes , Virulência/genética , Lactonas/metabolismo , Larva/microbiologia , Homosserina/análogos & derivados , Homosserina/metabolismo
3.
J Biol Chem ; 300(9): 107641, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39122000

RESUMO

Fe-S clusters are critical cofactors for redox chemistry in all organisms. The cysteine desulfurase, SufS, provides sulfur in the SUF Fe-S cluster bioassembly pathway. SufS is a dimeric, pyridoxal 5'-phosphate-dependent enzyme that uses cysteine as a substrate to generate alanine and a covalent persulfide on an active site cysteine residue. SufS enzymes are activated by an accessory transpersulfurase protein, either SufE or SufU depending on the organism, which accepts the persulfide product and delivers it to downstream partners for Fe-S assembly. Here, using Escherichia coli proteins, we present the first X-ray crystal structure of a SufS/SufE complex. There is a 1:1 stoichiometry with each monomeric unit of the EcSufS dimer bound to one EcSufE subunit, though one EcSufE is rotated ∼7° closer to the EcSufS active site. EcSufE makes clear interactions with the α16 helix of EcSufS and site-directed mutants of several α16 residues were deficient in EcSufE binding. Analysis of the EcSufE structure showed a loss of electron density at the EcSufS/EcSufE interface for a flexible loop containing the highly conserved residue R119. An R119A EcSufE variant binds EcSufS but is not active in cysteine desulfurase assays and fails to support Fe-S cluster bioassembly in vivo. 35S-transfer assays suggest that R119A EcSufE can receive a persulfide, suggesting the residue may function in a release mechanism. The structure of the EcSufS/EcSufE complex allows for comparison with other cysteine desulfurases to understand mechanisms of protected persulfide transfer across protein interfaces.


Assuntos
Liases de Carbono-Enxofre , Proteínas de Escherichia coli , Escherichia coli , Proteínas Ferro-Enxofre , Sulfetos , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Sulfetos/química , Sulfetos/metabolismo , Escherichia coli/metabolismo , Escherichia coli/genética , Cristalografia por Raios X , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/química , Proteínas Ferro-Enxofre/metabolismo , Proteínas Ferro-Enxofre/química , Proteínas Ferro-Enxofre/genética , Domínio Catalítico , Modelos Moleculares
4.
Nucleic Acids Res ; 52(17): 10543-10562, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39166491

RESUMO

In this study, we present an extensive analysis of a widespread group of bacterial tRNA de-modifying enzymes, dubbed RudS, which consist of a TudS desulfidase fused to a Domain of Unknown Function 1722 (DUF1722). RudS enzymes exhibit specific de-modification activity towards the 4-thiouridine modification (s4U) in tRNA molecules, as indicated by our experimental findings. The heterologous overexpression of RudS genes in Escherichia coli significantly reduces the tRNA 4-thiouridine content and diminishes UVA-induced growth delay, indicating the enzyme's role in regulating photosensitive tRNA s4U modification. Through a combination of protein modeling, docking studies, and molecular dynamics simulations, we have identified amino acid residues involved in catalysis and tRNA binding. Experimental validation through targeted mutagenesis confirms the TudS domain as the catalytic core of RudS, with the DUF1722 domain facilitating tRNA binding in the anticodon region. Our results suggest that RudS tRNA modification eraser proteins may play a role in regulating tRNA during prokaryotic stress responses.


Assuntos
Escherichia coli , RNA de Transferência , Tiouridina , Tiouridina/metabolismo , Tiouridina/análogos & derivados , Tiouridina/química , RNA de Transferência/metabolismo , RNA de Transferência/genética , Escherichia coli/genética , Escherichia coli/enzimologia , Simulação de Dinâmica Molecular , Simulação de Acoplamento Molecular , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/química , Domínio Catalítico , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/química , Anticódon/genética
5.
Biochim Biophys Acta Mol Cell Res ; 1871(7): 119794, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39033933

RESUMO

Sulfur-containing biomolecules such as [FeS] clusters, thiamin, biotin, molybdenum cofactor, and sulfur-containing tRNA nucleosides are essential for various biochemical reactions. The amino acid l-cysteine serves as the major sulfur source for the biosynthetic pathways of these sulfur-containing cofactors in prokaryotic and eukaryotic systems. The first reaction in the sulfur mobilization involves a class of pyridoxal-5'-phosphate (PLP) dependent enzymes catalyzing a Cys:sulfur acceptor sulfurtransferase reaction. The first half of the catalytic reaction involves a PLP-dependent CS bond cleavage, resulting in a persulfide enzyme intermediate. The second half of the reaction involves the subsequent transfer of the thiol group to a specific acceptor molecule, which is responsible for the physiological role of the enzyme. Structural and biochemical analysis of these Cys sulfurtransferase enzymes shows that specific protein-protein interactions with sulfur acceptors modulate their catalytic reactivity and restrict their biochemical functions.


Assuntos
Cisteína , Fosfato de Piridoxal , Enxofre , Sulfurtransferases , Enxofre/metabolismo , Enxofre/química , Cisteína/metabolismo , Cisteína/química , Sulfurtransferases/metabolismo , Sulfurtransferases/química , Fosfato de Piridoxal/metabolismo , Humanos , Cofatores de Molibdênio , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/química
6.
Cancer Genomics Proteomics ; 21(4): 395-398, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38944421

RESUMO

BACKGROUND/AIM: It has been recently demonstrated that a methionine-restricted diet increases the response to immune checkpoint inhibitors (ICIs) via an increase in PD-L1 in a syngeneic mouse colorectal-cancer model. Our laboratory has developed recombinant methioninase (rMETase) to restrict methionine. The aim of the present study was to determine if rMETase can increase PD-L1 expression in a human colorectal cancer cell line in vitro. MATERIALS AND METHODS: We evaluated the half-maximal inhibitory concentration (IC50) value of rMETase on HCT-116 human colorectal cancer cells. HCT-116 cells were treated with rMETase at the IC50 Western immunoblotting was used to compare PD-L1 expression in HCT-116 cells treated with and without rMETase. RESULTS: The IC50 value of rMETase on HCT-116 was 0.79 U/ml. Methionine restriction using rMETase increased PD-L1 expression compared to the untreated control (p<0.05). CONCLUSION: Methionine restriction with rMETase up-regulates PD-L1 expression in human colorectal cancer cells and the combination of rMETase and ICIs may have the potential to improve immunotherapy in human colorectal cancer.


Assuntos
Antígeno B7-H1 , Liases de Carbono-Enxofre , Neoplasias Colorretais , Metionina , Proteínas Recombinantes , Humanos , Liases de Carbono-Enxofre/metabolismo , Metionina/farmacologia , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Proteínas Recombinantes/farmacologia , Células HCT116
7.
Microbiol Spectr ; 12(8): e0055624, 2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-38916309

RESUMO

All sulfur transfer pathways have generally a l-cysteine desulfurase as an initial sulfur-mobilizing enzyme in common, which serves as a sulfur donor for the biosynthesis of numerous sulfur-containing biomolecules in the cell. In Escherichia coli, the housekeeping l-cysteine desulfurase IscS has several interaction partners, which bind at different sites of the protein. So far, the interaction sites of IscU, Fdx, CyaY, and IscX involved in iron-sulfur (Fe-S) cluster assembly have been mapped, in addition to TusA, which is required for molybdenum cofactor biosynthesis and mnm5s2U34 tRNA modifications, and ThiI, which is involved in thiamine biosynthesis and s4U8 tRNA modifications. Previous studies predicted that the sulfur acceptor proteins bind to IscS one at a time. E. coli TusA has, however, been suggested to be involved in Fe-S cluster assembly, as fewer Fe-S clusters were detected in a ∆tusA mutant. The basis for this reduction in Fe-S cluster content is unknown. In this work, we investigated the role of TusA in iron-sulfur cluster assembly and iron homeostasis. We show that the absence of TusA reduces the translation of fur, thereby leading to pleiotropic cellular effects, which we dissect in detail in this study.IMPORTANCEIron-sulfur clusters are evolutionarily ancient prosthetic groups. The ferric uptake regulator plays a major role in controlling the expression of iron homeostasis genes in bacteria. We show that a ∆tusA mutant is impaired in the assembly of Fe-S clusters and accumulates iron. TusA, therefore, reduces fur mRNA translation leading to pleiotropic cellular effects.


Assuntos
Proteínas de Escherichia coli , Escherichia coli , Homeostase , Proteínas Ferro-Enxofre , Ferro , Proteínas Repressoras , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Ferro/metabolismo , Proteínas Ferro-Enxofre/metabolismo , Proteínas Ferro-Enxofre/genética , Proteínas Repressoras/metabolismo , Proteínas Repressoras/genética , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/genética , Regulação Bacteriana da Expressão Gênica , Enxofre/metabolismo , Biossíntese de Proteínas , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Pteridinas/metabolismo , Cofatores de Molibdênio
8.
J Phys Chem B ; 128(24): 5823-5839, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38848492

RESUMO

The reaction of benzylsuccinate synthase, the radical-based addition of toluene to a fumarate cosubstrate, is initiated by hydrogen transfer from a conserved cysteine to the nearby glycyl radical in the active center of the enzyme. In this study, we analyze this step by comprehensive computer modeling, predicting (i) the influence of bound substrates or products, (ii) the energy profiles of forward- and backward hydrogen-transfer reactions, (iii) their kinetic constants and potential mechanisms, (iv) enantiospecificity differences, and (v) kinetic isotope effects. Moreover, we support several of the computational predictions experimentally, providing evidence for the predicted H/D-exchange reactions into the product and at the glycyl radical site. Our data indicate that the hydrogen transfer reactions between the active site glycyl and cysteine are principally reversible, but their rates differ strongly depending on their stereochemical orientation, transfer of protium or deuterium, and the presence or absence of substrates or products in the active site. This is particularly evident for the isotope exchange of the remaining protium atom of the glycyl radical to deuterium, which appears dependent on substrate or product binding, explaining why the exchange is observed in some, but not all, glycyl-radical enzymes.


Assuntos
Biocatálise , Cinética , Liases de Carbono-Enxofre/química , Liases de Carbono-Enxofre/metabolismo , Domínio Catalítico , Modelos Moleculares , Cisteína/química , Cisteína/metabolismo , Hidrogênio/química , Radicais Livres/química , Radicais Livres/metabolismo , Carbono-Carbono Liases
9.
Int J Mol Sci ; 25(12)2024 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-38928118

RESUMO

ß C-S lyases (ß-CSLs; EC 4.4.1.8) are enzymes catalyzing the dissociation of ß carbon-sulfur bonds of cysteine S-conjugates to produce odorant metabolites with a free thiol group. These enzymes are increasingly studied for their role in flavor generation in a variety of food products, whether these processes occur directly in plants, by microbial ß-CSLs during fermentation, or in the mouth under the action of the oral microbiota. Microbial ß-CSLs react with sulfur aroma precursors present in beverages, vegetables, fruits, or aromatic herbs like hop but also potentially with some precursors formed through Maillard reactions in cooked foods such as meat or coffee. ß-CSLs from microorganisms like yeasts and lactic acid bacteria have been studied for their role in the release of polyfunctional thiols in wine and beer during fermentation. In addition, ß-CSLs from microorganisms of the human oral cavity were shown to metabolize similar precursors and to produce aroma in the mouth with an impact on retro-olfaction. This review summarizes the current knowledge on ß-CSLs involved in flavor generation with a focus on enzymes from microbial species present either in the fermentative processes or in the oral cavity. This paper highlights the importance of this enzyme family in the food continuum, from production to consumption, and offers new perspectives concerning the utilization of ß-CSLs as a flavor enhancer.


Assuntos
Fermentação , Aromatizantes , Humanos , Aromatizantes/metabolismo , Liases de Carbono-Enxofre/metabolismo , Bactérias/enzimologia , Bactérias/metabolismo , Paladar
10.
Biochemistry ; 63(12): 1569-1577, 2024 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-38813769

RESUMO

The Escherichia coli cysteine desulfurase SufS (EcSufS) is a dimeric, PLP-dependent enzyme responsible for sulfur mobilization in the SUF Fe-S cluster bioassembly pathway. The enzyme uses cysteine as a sulfur source and generates alanine and a covalent persulfide located on an active site of cysteine. Optimal in vitro activity of EcSufS requires the presence of the transpersulfurase protein, EcSufE, and a strong reductant. Here, presteady-state and single-turnover kinetics are used to investigate the mechanism of EcSufS activation by EcSufE. In the absence of EcSufE, EcSufS exhibits a presteady-state burst of product production with an amplitude of ∼0.4 active site equivalents, consistent with a half-sites reactivity. KinTek Explorer was used to isolate the first turnover of alanine formation and fit the data with a simplified kinetic mechanism with steps for alanine formation (k3) and a net rate constant for the downstream steps (k5). Using this treatment, microscopic rate constants of 2.3 ± 0.5 s-1 and 0.10 ± 0.01 s-1 were determined for k3 and k5, respectively. The inclusion of EcSufE in the reaction results in a similar rate constant for k3 but induces a 10-fold enhancement of k5 to 1.1 ± 0.2 s-1, such that both steps are partially rate-determining. The most likely downstream step where EcSufE could exert influence on EcSufS activity is the removal of the persulfide intermediate. Importantly, this step appears to serve as a limiting feature in the half-sites activity such that activating persulfide transfer allows for rapid shifting between active sites. Single-turnover assays show that the presence of EcSufE slightly slowed the rates of alanine-forming steps, suggesting it does not activate steps in the desulfurase half reaction.


Assuntos
Liases de Carbono-Enxofre , Proteínas de Escherichia coli , Escherichia coli , Sulfetos , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/química , Sulfetos/metabolismo , Sulfetos/química , Escherichia coli/metabolismo , Escherichia coli/enzimologia , Escherichia coli/genética , Cinética , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/química , Alanina/metabolismo , Alanina/química , Domínio Catalítico , Cisteína/metabolismo , Cisteína/química , Proteínas Ferro-Enxofre/metabolismo , Proteínas Ferro-Enxofre/química
11.
J Agric Food Chem ; 72(23): 13228-13239, 2024 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-38810088

RESUMO

Limited alliinase resources cause difficulties in the biosynthesis of thiosulfinates (e.g., allicin), restricting their applications in the agricultural and food industries. To effectively biosynthesize thiosulfinates, this study aimed to excavate bacterial alliinase resources and elucidate their catalytic properties. Two bacterial cystathionine ß-lyases (MetCs) possessing high alliinase activity (>60 U mg -1) toward L-(-)-alliin were identified from Allium sativum rhizosphere isolates. Metagenomic exploration revealed that cystathionine ß-lyase from Bacillus cereus (BcPatB) possessed high activity toward both L-(±)-alliin and L-(+)-alliin (208.6 and 225.1 U mg -1), respectively. Although these enzymes all preferred l-cysteine S-conjugate sulfoxides as substrates, BcPatB had a closer phylogenetic relationship with Allium alliinases and shared several similar features with A. sativum alliinase. Interestingly, the Trp30Ile31Ala32Asp33 Met34 motif in a cuspate loop of BcPatB, especially sites 31 and 32 at the top of the motif, was modeled to locate near the sulfoxide of L-(+)-alliin and is important for substrate stereospecificity. Moreover, the stereoselectivity and activity of mutants I31V and A32G were higher toward L-(+)-alliin than those of mutant I31L/D33E toward L-(-)-alliin. Using bacterial alliinases and chemically synthesized substrates, we obtained thiosulfinates with high antimicrobial and antinematode activities that could provide insights into the protection of crops and food.


Assuntos
Proteínas de Bactérias , Alho , Sequência de Aminoácidos , Bacillus cereus/enzimologia , Bacillus cereus/genética , Bactérias/classificação , Bactérias/enzimologia , Bactérias/genética , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/química , Cisteína/análogos & derivados , Dissulfetos/química , Dissulfetos/metabolismo , Alho/enzimologia , Alho/microbiologia , Cinética , Filogenia , Estereoisomerismo , Especificidade por Substrato , Ácidos Sulfínicos/química , Ácidos Sulfínicos/metabolismo
12.
Environ Res ; 256: 119244, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38810822

RESUMO

Industrial wastewater is a major environmental concern due to its high copper content, which poses significant toxicity to microbial life. Autoinducer-2 (AI-2) can participate in the inter- and intra-species communication and regulate the physiological functions of different bacterial species by producing AI-2 signal molecules. However, there are few research reports on the luxS gene and lsr operon functions for AI-2 in bacteria with a certain tolerance to copper. This study delves into the potential of quorum sensing mechanisms, particularly the AI-2 system, for enhancing microbial resistance to copper toxicity in Klebsiella michiganensis (KM). We detail the critical roles of the luxS gene in AI-2 synthesis and the lsr operon in AI-2 uptake, demonstrating their collective impact on enhancing copper resistance. Our findings show that mutations in the lsr operon, alongside the knockout of the luxS gene in KM strain (KMΔluxSΔlsr), significantly impair the strain's motility (p < 0.0001) and biofilm formation (p < 0.01), underscoring the operon's role in AI-2 transport. These genetic insights are pivotal for developing bioremediation strategies aimed at mitigating copper pollution in wastewater. By elucidating the mechanisms through which KM modulates copper resistance, this study highlights the broader ecological significance of leveraging microbial quorum sensing pathways for sustainable wastewater management.


Assuntos
Proteínas de Bactérias , Liases de Carbono-Enxofre , Cobre , Klebsiella , Óperon , Percepção de Quorum , Cobre/toxicidade , Percepção de Quorum/efeitos dos fármacos , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Klebsiella/genética , Klebsiella/efeitos dos fármacos , Klebsiella/metabolismo , Homosserina/análogos & derivados , Homosserina/metabolismo , Lactonas/metabolismo
13.
Biochim Biophys Acta Rev Cancer ; 1879(4): 189122, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38796027

RESUMO

Cancer cells are addicted to L-methionine (L-Met) and have a much greater requirement for L-Met than normal cells due to excess transmethylation, termed the Hoffman effect. By targeting this vulnerability through dietary restriction of L-Met, researchers have been able to achieve promising results in inhibiting tumor growth and eradicating cancer cells. Methioninase (EC 4.4.1.11; METase) catalyzes the transformation of L-Met into α-ketobutyrate, ammonia, and methanethiol. The use of METase was initially limited due to its poor stability in vivo, high immunogenicity, and enzyme-induced inactivating antibodies. These issues could be partially resolved by PEGylation, encapsulation in erythrocytes, and various site-directed mutagenesis. The big breakthrough came when it was discovered that METase is effectively administered orally. The enzyme L-asparaginase is approved by the FDA for treatment of acute lymphoblastic leukemia. METase has more potential as a therapeutic since addiction to L-Met is a general and fundamental hallmark of cancer.


Assuntos
Liases de Carbono-Enxofre , Neoplasias , Liases de Carbono-Enxofre/uso terapêutico , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Metionina/metabolismo , Animais , Antineoplásicos/uso terapêutico , Antineoplásicos/farmacologia
14.
J Biol Chem ; 300(6): 107371, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38750791

RESUMO

The sulfite-reducing bacterium Bilophila wadsworthia, a common human intestinal pathobiont, is unique in its ability to metabolize a wide variety of sulfonates to generate sulfite as a terminal electron acceptor (TEA). The resulting formation of H2S is implicated in inflammation and colon cancer. l-cysteate, an oxidation product of l-cysteine, is among the sulfonates metabolized by B. wadsworthia, although the enzymes involved remain unknown. Here we report a pathway for l-cysteate dissimilation in B. wadsworthia RZATAU, involving isomerization of l-cysteate to d-cysteate by a cysteate racemase (BwCuyB), followed by cleavage into pyruvate, ammonia and sulfite by a d-cysteate sulfo-lyase (BwCuyA). The strong selectivity of BwCuyA for d-cysteate over l-cysteate was rationalized by protein structural modeling. A homolog of BwCuyA in the marine bacterium Silicibacter pomeroyi (SpCuyA) was previously reported to be a l-cysteate sulfo-lyase, but our experiments confirm that SpCuyA too displays a strong selectivity for d-cysteate. Growth of B. wadsworthia with cysteate as the electron acceptor is accompanied by production of H2S and induction of BwCuyA. Close homologs of BwCuyA and BwCuyB are present in diverse bacteria, including many sulfate- and sulfite-reducing bacteria, suggesting their involvement in cysteate degradation in different biological environments.


Assuntos
Cisteína , Cisteína/metabolismo , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Bilophila/metabolismo , Bilophila/enzimologia , Racemases e Epimerases/metabolismo , Oxirredução , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/química , Sulfitos/metabolismo , Humanos
15.
Sci Total Environ ; 933: 173057, 2024 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-38729372

RESUMO

Dimethylsulfoniopropionate (DMSP), a key organic sulfur compound in marine and subseafloor sediments, is degraded by phytoplankton and bacteria, resulting in the release of the climate-active volatile gas dimethylsulfide (DMS). However, it remains unclear if dominant eukaryotic fungi in subseafloor sediments possess specific abilities and metabolic mechanisms for DMSP degradation and DMS formation. Our study provides the first evidence that fungi from coal-bearing sediments ∼2 km below the seafloor, such as Aspergillus spp., Chaetomium globosum, Cladosporium sphaerospermum, and Penicillium funiculosum, can degrade DMSP and produce DMS. In Aspergillus sydowii 29R-4-F02, which exhibited the highest DMSP-dependent DMS production rate (16.95 pmol/µg protein/min), two DMSP lyase genes, dddP and dddW, were identified. Remarkably, the dddW gene, previously observed only in bacteria, was found to be crucial for fungal DMSP cleavage. These findings not only extend the list of fungi capable of degrading DMSP, but also enhance our understanding of DMSP lyase diversity and the role of fungi in DMSP decomposition in subseafloor sedimentary ecosystems.


Assuntos
Fungos , Compostos de Sulfônio , Compostos de Sulfônio/metabolismo , Fungos/metabolismo , Sedimentos Geológicos/microbiologia , Sulfetos/metabolismo , Biodegradação Ambiental , Liases de Carbono-Enxofre/metabolismo
16.
Biochim Biophys Acta Mol Cell Res ; 1871(5): 119731, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38631442

RESUMO

Molybdenum cofactor (Moco) biosynthesis is a complex process that involves the coordinated function of several proteins. In the recent years it has become evident that the availability of Fe-S clusters play an important role for the biosynthesis of Moco. First, the MoaA protein binds two [4Fe-4S] clusters per monomer. Second, the expression of the moaABCDE and moeAB operons is regulated by FNR, which senses the availability of oxygen via a functional [4Fe-4S] cluster. Finally, the conversion of cyclic pyranopterin monophosphate to molybdopterin requires the availability of the L-cysteine desulfurase IscS, which is an enzyme involved in the transfer of sulfur to various acceptor proteins with a main role in the assembly of Fe-S clusters. In this review, we dissect the dependence of the production of active molybdoenzymes in detail, starting from the regulation of gene expression and further explaining sulfur delivery and Fe-S cluster insertion into target enzymes. Further, Fe-S cluster assembly is also linked to iron availability. While the abundance of selected molybdoenzymes is largely decreased under iron-limiting conditions, we explain that the expression of the genes is dependent on an active FNR protein. FNR is a very important transcription factor that represents the master-switch for the expression of target genes in response to anaerobiosis. Moco biosynthesis is further directly dependent on the presence of ArcA and also on an active Fur protein.


Assuntos
Coenzimas , Proteínas Ferro-Enxofre , Metaloproteínas , Cofatores de Molibdênio , Pteridinas , Metaloproteínas/metabolismo , Metaloproteínas/genética , Metaloproteínas/biossíntese , Proteínas Ferro-Enxofre/metabolismo , Proteínas Ferro-Enxofre/genética , Coenzimas/metabolismo , Coenzimas/biossíntese , Coenzimas/genética , Pteridinas/metabolismo , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Ferro/metabolismo , Enxofre/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/genética , Regulação Bacteriana da Expressão Gênica , Óperon , Isomerases
17.
Biomolecules ; 14(4)2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38672469

RESUMO

Porcine extraintestinal pathogenic Escherichia coli (ExPEC) is a pathogenic bacterium that causes huge economic losses to the pig farming industry and considerably threatens human health. The quorum sensing (QS) system plays a crucial role in the survival and pathogenesis of pathogenic bacteria. Hence, it is a viable approach to prevent ExPEC infection by compromising the QS system, particularly the LuxS/AI-2 system. In this study, we investigated the effects of baicalin on the LuxS/AI-2 system of ExPEC. Baicalin at concentrations of 25, 50, and 100 µg/mL significantly diminished the survival ability of ExPEC in hostile environments and could inhibit the biofilm formation and autoagglutination ability in ExPEC. Moreover, baicalin dose-dependently decreased the production of AI-2 and down-regulated the expression level of luxS in PCN033. These results suggest that baicalin can weaken the virulence of PCN033 by inhibiting the LuxS/AI-2 system. After the gene luxS was deleted, AI-2 production in PCN033 was almost completely eliminated, similar to the effect of baicalin on the production of AI-2 in PCN033. This indicates that baicalin reduced the production of AI-2 by inhibiting the expression level of luxS in ExPEC. In addition, the animal experiment further showed the potential of baicalin as a LuxS/AI-2 system inhibitor to prevent ExPEC infection. This study highlights the potential of baicalin as a natural quorum-sensing inhibitor for therapeutic applications in preventing ExPEC infection by targeting the LuxS/AI-2 system.


Assuntos
Proteínas de Bactérias , Liases de Carbono-Enxofre , Escherichia coli Extraintestinal Patogênica , Flavonoides , Homosserina , Homosserina/análogos & derivados , Percepção de Quorum , Percepção de Quorum/efeitos dos fármacos , Flavonoides/farmacologia , Animais , Liases de Carbono-Enxofre/genética , Liases de Carbono-Enxofre/metabolismo , Suínos , Virulência/efeitos dos fármacos , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/genética , Homosserina/metabolismo , Escherichia coli Extraintestinal Patogênica/efeitos dos fármacos , Escherichia coli Extraintestinal Patogênica/patogenicidade , Escherichia coli Extraintestinal Patogênica/genética , Biofilmes/efeitos dos fármacos , Biofilmes/crescimento & desenvolvimento , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/microbiologia , Lactonas/farmacologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Doenças dos Suínos/microbiologia , Doenças dos Suínos/tratamento farmacológico
18.
Biomolecules ; 14(4)2024 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-38672486

RESUMO

The Dph1•Dph2 heterodimer from yeast is a radical SAM (RS) enzyme that generates the 3-amino-3-carboxy-propyl (ACP) precursor for diphthamide, a clinically relevant modification on eukaryotic elongation factor 2 (eEF2). ACP formation requires SAM cleavage and atypical Cys-bound Fe-S clusters in each Dph1 and Dph2 subunit. Intriguingly, the first Cys residue in each motif is found next to another ill-defined cysteine that we show is conserved across eukaryotes. As judged from structural modeling, the orientation of these tandem cysteine motifs (TCMs) suggests a candidate Fe-S cluster ligand role. Hence, we generated, by site-directed DPH1 and DPH2 mutagenesis, Dph1•Dph2 variants with cysteines from each TCM replaced individually or in combination by serines. Assays diagnostic for diphthamide formation in vivo reveal that while single substitutions in the TCM of Dph2 cause mild defects, double mutations almost entirely inactivate the RS enzyme. Based on enhanced Dph1 and Dph2 subunit instability in response to cycloheximide chases, the variants with Cys substitutions in their cofactor motifs are particularly prone to protein degradation. In sum, we identify a fourth functionally cooperative Cys residue within the Fe-S motif of Dph2 and show that the Cys-based cofactor binding motifs in Dph1 and Dph2 are critical for the structural integrity of the dimeric RS enzyme in vivo.


Assuntos
Motivos de Aminoácidos , Cisteína , Histidina/análogos & derivados , Proteínas Repressoras , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Cisteína/metabolismo , Cisteína/genética , Cisteína/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimologia , Multimerização Proteica , Liases de Carbono-Enxofre/metabolismo , Liases de Carbono-Enxofre/química , Liases de Carbono-Enxofre/genética , Mutagênese Sítio-Dirigida
19.
Nat Commun ; 15(1): 3269, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38627381

RESUMO

Maturation of iron-sulfur proteins in eukaryotes is initiated in mitochondria by the core iron-sulfur cluster assembly (ISC) complex, consisting of the cysteine desulfurase sub-complex NFS1-ISD11-ACP1, the scaffold protein ISCU2, the electron donor ferredoxin FDX2, and frataxin, a protein dysfunctional in Friedreich's ataxia. The core ISC complex synthesizes [2Fe-2S] clusters de novo from Fe and a persulfide (SSH) bound at conserved cluster assembly site residues. Here, we elucidate the poorly understood Fe-dependent mechanism of persulfide transfer from cysteine desulfurase NFS1 to ISCU2. High-resolution cryo-EM structures obtained from anaerobically prepared samples provide snapshots that both visualize different stages of persulfide transfer from Cys381NFS1 to Cys138ISCU2 and clarify the molecular role of frataxin in optimally positioning assembly site residues for fast sulfur transfer. Biochemical analyses assign ISCU2 residues essential for sulfur transfer, and reveal that Cys138ISCU2 rapidly receives the persulfide without a detectable intermediate. Mössbauer spectroscopy assessing the Fe coordination of various sulfur transfer intermediates shows a dynamic equilibrium between pre- and post-sulfur-transfer states shifted by frataxin. Collectively, our study defines crucial mechanistic stages of physiological [2Fe-2S] cluster assembly and clarifies frataxin's molecular role in this fundamental process.


Assuntos
Frataxina , Proteínas Ferro-Enxofre , Proteínas Ferro-Enxofre/metabolismo , Sulfetos/metabolismo , Enxofre/metabolismo , Liases de Carbono-Enxofre/metabolismo , Proteínas de Ligação ao Ferro/metabolismo
20.
Int J Biol Macromol ; 265(Pt 2): 130997, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38508568

RESUMO

Cancer remains a global health challenge, demanding novel therapeutic options due to the debilitating side effects of conventional treatments on healthy tissues. The review highlights the potential of L-methioninase, a pyridoxal-5-phosphate (PLP)-dependent enzyme, as a promising avenue in alternative cancer therapy. L-methioninase offers a unique advantage, its ability to selectively target and inhibit the growth of cancer cells without harming healthy cells. This selectivity arises because tumor cells lack an essential enzyme called methionine synthase, which healthy cells use to make the vital amino acid L-methionine. Several sources harbor L-methioninase, including bacteria, fungi, plants, and protozoa. Future research efforts can explore and exploit this diverse range of sources to improve the therapeutic potential of L-methioninase in the fight against cancer. Despite challenges, research actively explores microbial L-methioninase for its anticancer potential. This review examines the enzyme's side effects, advancements in combination therapies, recombinant technologies, polymer conjugation and novel delivery methods like nanoparticles, while highlighting the success of oral administration in preclinical trials. Beyond its promising role in cancer therapy, L-methioninase holds potential applications in food science, antioxidants, and various health concerns like diabetes, cardiovascular issues, and neurodegenerative diseases. This review provides a piece of current knowledge and future prospects of L-methioninase, exploring its diverse therapeutic potential.


Assuntos
Liases de Carbono-Enxofre , Neoplasias , Humanos , Liases de Carbono-Enxofre/metabolismo , Neoplasias/tratamento farmacológico , Terapia Combinada , Fungos/metabolismo , Metionina/metabolismo , Proteínas Recombinantes/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA