Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
Int J Mol Sci ; 25(7)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38612657

RESUMEN

Huntington's disease (HD) arises from the abnormal expansion of CAG repeats in the huntingtin gene (HTT), resulting in the production of the mutant huntingtin protein (mHTT) with a polyglutamine stretch in its N-terminus. The pathogenic mechanisms underlying HD are complex and not yet fully elucidated. However, mHTT forms aggregates and accumulates abnormally in neuronal nuclei and processes, leading to disruptions in multiple cellular functions. Although there is currently no effective curative treatment for HD, significant progress has been made in developing various therapeutic strategies to treat HD. In addition to drugs targeting the neuronal toxicity of mHTT, gene therapy approaches that aim to reduce the expression of the mutant HTT gene hold great promise for effective HD therapy. This review provides an overview of current HD treatments, discusses different therapeutic strategies, and aims to facilitate future therapeutic advancements in the field.


Asunto(s)
Enfermedad de Huntington , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/terapia , Terapia Genética , Proteínas Mutantes
2.
Neurobiol Dis ; 187: 106291, 2023 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-37716514

RESUMEN

Huntington's disease (HD) is an autosomal-dominant inherited neurodegenerative disease caused by a CAG repeat expansion in exon1 of the huntingtin gene (HTT). This expansion leads to the production of N-terminal mutant huntingtin protein (mHtt) that contains an expanded polyglutamine tract, which is toxic to neurons and causes neurodegeneration. While the production of N-terminal mHtt can be mediated by proteolytic cleavage of full-length mHtt, abnormal splicing of exon1-intron1 of mHtt has also been identified in the brains of HD mice and patients. However, the proportion of aberrantly spliced exon1 mHTT in relation to normal mHTT exon remains to be defined. In this study, HTT exon1 production was examined in the HD knock-in (KI) pig model, which more closely recapitulates neuropathology seen in HD patient brains than HD mouse models. The study revealed that aberrant spliced HTT exon1 is also present in the brains of HD pigs, but it is expressed at a much lower level than the normally spliced HTT exon products. These findings suggest that careful consideration is needed when assessing the contribution of aberrantly spliced mHTT exon1 to HD pathogenesis, and further rigorous investigation is required.

3.
BMC Microbiol ; 23(1): 369, 2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38030994

RESUMEN

BACKGROUND: Currently, clinical laboratories lack an effective method to differentiate between classical Klebsiella pneumoniae (cKP) and hypervirulent Klebsiella pneumoniae (hvKP) strains, leading to delays in diagnosing and treating hvKP infections. Previous studies have identified peg-344, iroB, iucA, prmpA, prmpA2, and siderophores (SP) yields greater than 30 µg/ml as reliable markers for distinguishing hvKP from cKp strains. However, these diagnostic tests were conducted on a relatively small study population and lacked sufficient clinical data support. In this study, hvKP strains were identified by biomarker analysis and the Galleria mellonella model. Combined with in vitro and in vivo experiments, the reliability of clinical identification method of hvKP was verified, which provided an experimental basis for timely diagnosis of hvKP infection. RESULTS: According to the clinical data, a total of 108 strains of hvKP were preliminary screened. Among them, 94 strains were further identified using PCR analysis of biomarkers and quantitative determination of SP. The high virulence of hvKP was subsequently confirmed through infection experiments on Galleria mellonella. Additionally, susceptibility testing revealed the identification of 58 carbapenem-resistant hvKP (CR-hvKP) strains and 36 carbapenem-sensitive hvKP (CS-hvKP) strains. By comparing molecular diagnostic indexes, molecular characteristics such as high SP production of CR-hvKP were found. CONCLUSION: The combination of clinical data and molecular diagnostic index analysis effectively enables the identification of hvKP, particularly CR-hvKP. This study provides a scientific basis for accurate clinical identification and timely treatment of hvKP.


Asunto(s)
Infecciones por Klebsiella , Mariposas Nocturnas , Humanos , Animales , Klebsiella pneumoniae/genética , Reproducibilidad de los Resultados , Virulencia , Carbapenémicos , Biomarcadores , Sideróforos , Infecciones por Klebsiella/epidemiología , Antibacterianos/uso terapéutico
4.
Int J Mol Sci ; 24(17)2023 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-37685866

RESUMEN

Huntington's disease (HD) is caused by an expansion of a CAG repeat in the gene that encodes the huntingtin protein (HTT). The exact function of HTT is still not fully understood, and previous studies have mainly focused on identifying proteins that interact with HTT to gain insights into its function. Numerous HTT-interacting proteins have been discovered, shedding light on the functions and structure of HTT. Most of these proteins interact with the N-terminal region of HTT. Among the various HTT-interacting proteins, huntingtin-associated protein 1 (HAP1) and HTT-interacting protein 1 (HIP1) have been extensively studied. Recent research has uncovered differences in the distribution of HAP1 in monkey and human brains compared with mice. This finding suggests that there may be species-specific variations in the regulation and function of HTT-interacting proteins. Understanding these differences could provide crucial insights into the development of HD. In this review, we will focus on the recent advancements in the study of HTT-interacting proteins, with particular attention to the differential distributions of HTT and HAP1 in larger animal models.


Asunto(s)
Encéfalo , Enfermedad de Huntington , Humanos , Animales , Ratones , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Modelos Animales , Especificidad de la Especie
5.
J Biol Chem ; 294(12): 4583-4595, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30705089

RESUMEN

Aquaporins (AQPs) are transmembrane proteins widely distributed in various organisms, and they facilitate bidirectional diffusion of water and uncharged solutes. The catalase-negative bacterium Streptococcus oligofermentans produces the highest H2O2 levels reported to date, which has to be exported to avoid oxidative stress. Here, we report that a S. oligofermentans aquaporin functions as a peroxiporin facilitating bidirectional transmembrane H2O2 transport. Knockout of this aquaporin homolog, So-AqpA, reduced H2O2 export by ∼50% and increased endogenous H2O2 retention, as indicated by the cellular H2O2 reporter HyPer. Heterologous expression of So-aqpA accelerated exogenous H2O2 influx into Saccharomyces cerevisiae and Escherichia coli cells, indicating that So-AqpA acts as an H2O2-transferring aquaporin. Alanine substitution revealed Phe-40 as a key residue for So-AqpA-mediated H2O2 transport. Northern blotting, qPCR, and luciferase reporter assays disclosed that H2O2 induces a >10-fold expression of So-aqpA Super-resolution imaging showed that H2O2 treatment increases So-AqpA protein molecules per cell by 1.6- to 3-fold. Inactivation of two redox-regulatory transcriptional repressors, PerR and MntR, reduced H2O2-induced So-aqpA expression to 1.8- and 4-fold, respectively. Electrophoretic mobility shift assays determined that MntR, but not PerR, binds to the So-aqpA promoter, indicating that MntR directly regulates H2O2-induced So-aqpA expression. Importantly, So-aqpA deletion decreased oxic growth and intraspecies competition and diminished the competitive advantages of S. oligofermentans over the caries pathogen Streptococcus mutans Of note, So-aqpA orthologs with the functionally important Phe-40 are present in all streptococci. Our work has uncovered an intrinsic, H2O2-inducible bacterial peroxiporin that has a key physiological role in H2O2 detoxification in S. oligofermentans.


Asunto(s)
Acuaporinas/metabolismo , Peróxido de Hidrógeno/metabolismo , Estrés Oxidativo , Streptococcus/metabolismo , Sustitución de Aminoácidos , Acuaporinas/química , Acuaporinas/genética , Transporte Biológico , Escherichia coli/genética , Eliminación de Gen , Saccharomyces cerevisiae/genética , Transcripción Genética
6.
Mol Microbiol ; 112(2): 552-568, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31074889

RESUMEN

Natural transformation increases the genetic diversity of bacteria, but is costly and must be strictly controlled. We previously found that deletion of ccpA, a key regulator of carbon catabolite repression (CCR), reduced transformation efficiency of Streptococcus oligofermentans, the current work further investigated the regulatory mechanisms of CcpA. The competence operon comCDE is subjected to basal and autoregulatory transcription. A luciferase reporter detected a transcriptional readthrough (TRT) from the upstream tRNAArg into the comCDE operon, which was induced by L -arginine. Insertion of the Escherichia coli T1T2 terminator downstream of tRNAArg abolished TRT, and reduced the basal comCDE transcription by 77% and also the transformation efficiency. Deletion of ccpA increased tRNAArg TRT and tRNAArg -comCDE polycistronic transcript by twofold. An in vitro transcription assay determined that CcpA promoted the transcription termination of tRNAArg TRT, and RNA EMSA and SPR assays detected equal binding affinity of CcpA to both the RNA and DNA of tRNAArg . These results indicate that CcpA controls the basal comCDE transcription by post-transcriptional actions. Overexpression of comDE or its phospho-mimicking mutant comDED58E reduced transformation efficiency, indicating that excessive ComE impairs competence development. CCR-regulated competence was further confirmed by higher tRNAArg TRT but lower transformation efficiency in galactose than in glucose.


Asunto(s)
Proteínas Bacterianas/metabolismo , Represión Catabólica , Regulación Bacteriana de la Expresión Génica , Proteínas Represoras/metabolismo , Streptococcus/crecimiento & desarrollo , Streptococcus/metabolismo , Proteínas Bacterianas/genética , Carbono/metabolismo , Regulación del Desarrollo de la Expresión Génica , Operón , Proteínas Represoras/genética , Streptococcus/genética , Transcripción Genética
7.
J Biol Chem ; 292(13): 5519-5531, 2017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28223356

RESUMEN

Manganese contributes to anti-oxidative stress particularly in catalase-devoid bacteria, and DtxR family metalloregulators, through sensing cellular Mn2+ content, regulate its homeostasis. Here, we show that metalloregulator MntR (So-MntR) functions dually as Mn2+ and H2O2 sensors in mediating H2O2 resistance by an oral streptococcus. H2O2 disrupted So-MntR binding to Mn2+ transporter mntABC promoter and induced disulfide-linked dimerization of the protein. Mass spectrometry identified Cys-11/Cys-156 and Cys-11/Cys-11 disulfide-linked peptides in H2O2-treated So-MntR. Site mutagenesis of Cys-11 and Cys-156 and particularly Cys-11 abolished H2O2-induced disulfide-linked dimers and weakened H2O2 damage on So-MntR binding, indicating that H2O2 inactivates So-MntR via disulfide-linked dimerization. So-MntR C123S mutant was extremely sensitive to H2O2 oxidization in dimerization/oligomerization, probably because the mutagenesis caused a conformational change that facilitates Cys-11/Cys-156 disulfide linkage. Intermolecular Cys-11/Cys-11 disulfide was detected in C123S/C156S double mutant. Redox Western blot detected So-MntR oligomers in air-exposed cells but remarkably decreased upon H2O2 pulsing, suggesting a proteolysis of the disulfide-linked So-MntR oligomers. Remarkably, elevated C11S and C156S but much lower C123S proteins were detected in H2O2-pulsed cells, confirming Cys-11 and Cys-156 contributed to H2O2-induced oligomerization and degradation. Accordingly, in the C11S and C156S mutants, expression of mntABC and cellular Mn2+ decreased, but H2O2 susceptibility increased. In the C123S mutant, increased mntABC expression, cellular Mn2+ content, and manganese-mediated H2O2 survival were determined. Given the wide distribution of Cys-11 in streptococcal DtxR-like metalloregulators, the disclosed redox regulatory function and mechanism of So-MntR can be employed by the DtxR family proteins in bacterial resistance to oxidative stress.


Asunto(s)
Proteínas Bacterianas/fisiología , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo , Proteínas Represoras/fisiología , Streptococcus/química , Transportadoras de Casetes de Unión a ATP , Dimerización , Homeostasis/efectos de los fármacos , Manganeso/metabolismo , Mutagénesis Sitio-Dirigida , Estrés Oxidativo/efectos de los fármacos , Streptococcus/metabolismo
8.
Appl Environ Microbiol ; 80(8): 2351-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24487543

RESUMEN

Metal homeostasis plays a critical role in antioxidative stress. Streptococcus oligofermentans, an oral commensal facultative anaerobe lacking catalase activity, produces and tolerates abundant H2O2, whereas Dpr (an Fe(2+)-chelating protein)-dependent H2O2 protection does not confer such high tolerance. Here, we report that inactivation of perR, a peroxide-responsive repressor that regulates zinc and iron homeostasis in Gram-positive bacteria, increased the survival of H2O2-pulsed S. oligofermentans 32-fold and elevated cellular manganese 4.5-fold. perR complementation recovered the wild-type phenotype. When grown in 0.1 to 0.25 mM MnCl2, S. oligofermentans increased survival after H2O2 stress 2.5- to 23-fold, and even greater survival was found for the perR mutant, indicating that PerR is involved in Mn(2+)-mediated H2O2 resistance in S. oligofermentans. Mutation of mntA could not be obtained in brain heart infusion (BHI) broth (containing ~0.4 µM Mn(2+)) unless it was supplemented with ≥2.5 µM MnCl2 and caused 82 to 95% reduction of the cellular Mn(2+) level, while mntABC overexpression increased cellular Mn(2+) 2.1- to 4.5-fold. Thus, MntABC was identified as a high-affinity Mn(2+) transporter in S. oligofermentans. mntA mutation reduced the survival of H2O2-pulsed S. oligofermentans 5.7-fold, while mntABC overexpression enhanced H2O2-challenged survival 12-fold, indicating that MntABC-mediated Mn(2+) uptake is pivotal to antioxidative stress in S. oligofermentans. perR mutation or H2O2 pulsing upregulated mntABC, while H2O2-induced upregulation diminished in the perR mutant. This suggests that perR represses mntABC expression but H2O2 can release the suppression. In conclusion, this work demonstrates that PerR regulates manganese homeostasis in S. oligofermentans, which is critical to H2O2 stress defenses and may be distributed across all oral streptococci lacking catalase.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Manganeso/metabolismo , Estrés Oxidativo , Proteínas Represoras/metabolismo , Streptococcus/genética , Streptococcus/metabolismo , Estrés Fisiológico , Proteínas Bacterianas/genética , Silenciador del Gen , Peróxido de Hidrógeno/toxicidad , Viabilidad Microbiana/efectos de los fármacos , Oxidantes/toxicidad , Proteínas Represoras/genética , Streptococcus/efectos de los fármacos
9.
Cell Death Discov ; 9(1): 251, 2023 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-37460539

RESUMEN

The ketogenic diet (KD) is a low carbohydrate and high-fat protein diet. It plays a protective role in neurodegenerative diseases by elevating the levels of ketone bodies in blood, regulating central and peripheral metabolism and mitochondrial functions, inhibiting neuroinflammation and oxidative stress, and altering the gut microbiota. However, studies on ketogenic therapy for Parkinson's disease (PD) are still in their infancy. Therefore, we examined the possible protective effect of KD in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model, examined the mouse gut microbiota and its metabolites, and performed transcriptomics and metabolomics on the substantia nigra of mice. Our results showed that a long-term medium-chain triglyceride KD (MCT-KD) significantly reduced MPTP-induced damage to dopaminergic (DA) neurons, exerted antioxidant stress through the PI3K/Akt/Nrf2 pathway, and reversed oxidative stress in DA neurons. The MCT-KD also reduced mitochondrial loss, promoted ATP production, and inhibited the activation of microglia to protect DA neurons in MPTP-induced PD mice. MCT-KD altered the gut microbiota and consequently changed the metabolism of substantia nigra neurons through gut microbiota metabolites. Compared to the MPTP group, MCT-KD increased the abundance of gut microbiota, including Blautia and Romboutsia. MCT-KD also affects purine metabolism in the substantia nigra pars compacta (SNpc) by altering fecal metabolites. This study shows that MCT-KD has multiple protective effects against PD.

10.
Cell Rep ; 42(12): 113443, 2023 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-37979175

RESUMEN

Our previous work has established a knockin (KI) pig model of Huntington's disease (HD) that can replicate the typical pathological features of HD, including selective striatal neuronal loss, reactive gliosis, and axonal degeneration. However, HD KI mice exhibit milder neuropathological phenotypes and lack overt neurodegeneration. By performing RNA sequencing to compare the gene expression profiles between HD KI pigs and mice, we find that genes related to interleukin-17 (IL-17) signaling are upregulated in the HD pig brains compared to the mouse brains. Delivery of IL-17 into the brain striatum of HD KI mice causes greater reactive gliosis and synaptic deficiency compared to HD KI mice that received PBS. These findings suggest that the upregulation of genes related to IL-17 signaling in HD pig brains contributes to severe glial pathology in HD and identify this as a potential therapeutic target for treating HD.


Asunto(s)
Enfermedad de Huntington , Animales , Ratones , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Gliosis/patología , Enfermedad de Huntington/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Ratones Transgénicos , Neuronas/metabolismo , Porcinos
11.
J Bacteriol ; 194(15): 3824-32, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22609925

RESUMEN

Streptococcus oligofermentans is an oral commensal that inhibits the growth of the caries pathogen Streptococcus mutans by producing copious amounts of H(2)O(2) and that grows faster than S. mutans on galactose. In this study, we identified a novel eight-gene galactose (gal) operon in S. oligofermentans that was comprised of lacABCD, lacX, and three genes encoding a galactose-specific transporter. Disruption of lacA caused more growth reduction on galactose than mutation of galK, a gene in the Leloir pathway, indicating that the principal role of this operon is in galactose metabolism. Diauxic growth was observed in cultures containing glucose and galactose, and a luciferase reporter fusion to the putative gal promoter demonstrated 12-fold repression of the operon expression by glucose but was induced by galactose, suggesting a carbon catabolite repression (CCR) control in galactose utilization. Interestingly, none of the single-gene mutations in the well-known CCR regulators ccpA and manL affected diauxic growth, although the operon expression was upregulated in these mutants in glucose. A double mutation of ccpA and manL eliminated glucose repression of galactose utilization, suggesting that these genes have parallel functions in regulating gal operon expression and mediating CCR. Electrophoretic mobility shift assays demonstrated binding of CcpA to the putative catabolite response element motif in the promoter regions of the gal operon and manL, suggesting that CcpA regulates CCR through direct regulation of the transcription of the gal operon and manL. This provides the first example of oral streptococci using two parallel CcpA-dependent CCR pathways in controlling carbohydrate metabolism.


Asunto(s)
Represión Catabólica , Galactosa/metabolismo , Regulación Bacteriana de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Streptococcus/genética , Streptococcus/metabolismo , Sitios de Unión , Medios de Cultivo/química , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Eliminación de Gen , Perfilación de la Expresión Génica , Genes Bacterianos , Genes Reporteros , Glucosa/metabolismo , Luciferasas/análisis , Luciferasas/genética , Operón , Regiones Promotoras Genéticas , Unión Proteica , Streptococcus/crecimiento & desarrollo
12.
Appl Environ Microbiol ; 78(7): 2120-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22287002

RESUMEN

Complex interspecies interactions occur constantly between oral commensals and the opportunistic pathogen Streptococcus mutans in dental plaque. Previously, we showed that oral commensal Streptococcus oligofermentans possesses multiple enzymes for H(2)O(2) production, especially lactate oxidase (Lox), allowing it to out-compete S. mutans. In this study, through extensive biochemical and genetic studies, we identified a pyruvate oxidase (pox) gene in S. oligofermentans. A pox deletion mutant completely lost Pox activity, while ectopically expressed pox restored activity. Pox was determined to produce most of the H(2)O(2) in the earlier growth phase and log phase, while Lox mainly contributed to H(2)O(2) production in stationary phase. Both pox and lox were expressed throughout the growth phase, while expression of the lox gene increased by about 2.5-fold when cells entered stationary phase. Since lactate accumulation occurred to a large degree in stationary phase, the differential Pox- and Lox-generated H(2)O(2) can be attributed to differential gene expression and substrate availability. Interestingly, inactivation of pox causes a dramatic reduction in H(2)O(2) production from lactate, suggesting a synergistic action of the two oxidases in converting lactate into H(2)O(2). In an in vitro two-species biofilm experiment, the pox mutant of S. oligofermentans failed to inhibit S. mutans even though lox was active. In summary, S. oligofermentans develops a Pox-Lox synergy strategy to maximize its H(2)O(2) formation so as to win the interspecies competition.


Asunto(s)
Antibiosis , Oxigenasas de Función Mixta/metabolismo , Piruvato Oxidasa/metabolismo , Streptococcus mutans/enzimología , Streptococcus mutans/crecimiento & desarrollo , Streptococcus/enzimología , Streptococcus/fisiología , Biopelículas/crecimiento & desarrollo , Eliminación de Gen , Peróxido de Hidrógeno/metabolismo , Oxigenasas de Función Mixta/genética , Datos de Secuencia Molecular , Boca/microbiología , Piruvato Oxidasa/genética , Análisis de Secuencia de ADN , Especificidad de la Especie , Streptococcus/clasificación , Streptococcus/genética , Streptococcus/crecimiento & desarrollo , Streptococcus mutans/genética
13.
Cells ; 11(21)2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36359783

RESUMEN

Huntington's disease (HD) is an autosomal-dominant inherited progressive neurodegenerative disorder. It is caused by a CAG repeat expansion in the Huntingtin gene that is translated to an expanded polyglutamine (PolyQ) repeat in huntingtin protein. HD is characterized by mood swings, involuntary movement, and cognitive decline in the late disease stage. HD patients often die 15-20 years after disease onset. Currently, there is no cure for HD. Due to the striking neuronal loss in HD, most studies focused on the investigation of the predominantly neuronal degeneration in specific brain regions. However, the pathology of the white matter area in the brains of HD patients was also reported by clinical imaging studies, which showed white matter abnormalities even before the clinical onset of HD. Since oligodendrocytes form myelin sheaths around the axons in the brain, white matter lesions are likely attributed to alterations in myelin and oligodendrocyte-associated changes in HD. In this review, we summarized the evidence for white matter, myelin, and oligodendrocytes alterations that were previously observed in HD patients and animal models. We also discussed potential mechanisms for white matter changes and possible treatment to prevent glial dysfunction in HD.


Asunto(s)
Enfermedad de Huntington , Sustancia Blanca , Animales , Enfermedad de Huntington/metabolismo , Sustancia Blanca/patología , Proteína Huntingtina/genética , Encéfalo/metabolismo , Vaina de Mielina/metabolismo
14.
Microbiol Spectr ; 10(5): e0249622, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36106896

RESUMEN

The opportunistic pathogen Streptococcus pneumoniae (pneumococcus) is a human nasopharyngeal commensal, and host N-glycan metabolism promotes its colonization and invasion. It has been reported that glucose represses, while fetuin, a glycoconjugated model protein, induces, the genes involved in N-glycan degradation through the two-component system TCS07. However, the mechanisms of glucose repression and TCS07 induction remain unknown. Previously, we found that the pneumococcal aquaglyceroporin Pn-AqpC facilitates oxygen uptake, thereby contributing to the antioxidant potential and virulence. In this study, through Tandem Mass Tag (TMT) quantitative proteomics, we found that the deletion of Pn-aqpC caused a marked upregulation of 11 proteins involved in N-glycan degradation in glucose-grown pneumococcus R6. Both quantitative RT-PCR and GFP fluorescence reporters revealed that the upregulation of N-glycan genes was completely dependent on response regulator (RR) 07, but not on the histidine kinase HK07 of TCS07 or the phosphoryl-receiving aspartate residue of RR07 in ΔPn-aqpC, indicating that RR07 was activated in an HK07-independent manner when Pn-AqpC was absent. The deletion of Pn-aqpC also enhanced the expression of pyruvate formate lyase and increased formate production, probably due to reduced cellular oxygen content, indicating that a shunt of glucose catabolism to mixed acid fermentation occurs. Notably, formate induced the N-glycan degradation genes in glucose-grown R6, but the deletion of rr07 abolished this induction, indicating that formate activates RR07. However, the induction of N-glycan degradation proteins reduced the intraspecies competition of R6 in glucose. Therefore, although N-glycan degradation promotes pneumococcal pathogenesis, the glucose metabolites-based RR07 regulation reported here is of importance for balancing growth fitness and the pathogenicity of pneumococcus. IMPORTANCE Pneumococcus, a human opportunistic pathogen, is capable of metabolizing host complex N-glycans. N-glycan degradation promotes pneumococcus colonization in the nasopharynx as well as invasion into deeper tissues, thus significantly contributing to pathogenesis. It is known that the two-component system 07 induces the N-glycan metabolizing genes; however, how TCS07 is activated remains unknown. This study reveals that formate, the anaerobic fermentation metabolite of pneumococcus, is a novel activator of the response regulator (RR) 07. Although the high expression of N-glycan degradation genes promotes pneumococcal colonization in the nasopharynx and pathogenesis, this reduces pneumococcal growth fitness in glucose as indicated in this work. Notably, the presence of Pn-AqpC, an oxygen-transporting aquaglyceroporin, enables pneumococcus to maintain glucose homolactic acid fermentation, thus reducing formate production, maintaining RR07 inactivation, and controlling N-glycan degrading genes at a non-induced status. Thus, this study highlights a novel fermentation metabolism pattern linking TCS-regulated carbohydrate utilization strategies as a trade-off between the fitness and the pathogenicity of pneumococcus.


Asunto(s)
Acuagliceroporinas , Liasas , Humanos , Streptococcus pneumoniae/metabolismo , Fermentación , Histidina Quinasa/metabolismo , Ácido Aspártico/metabolismo , Antioxidantes/metabolismo , Polisacáridos , Formiatos/metabolismo , Glucosa/metabolismo , Fetuínas/metabolismo , Acuagliceroporinas/metabolismo , Piruvatos/metabolismo , Liasas/metabolismo , Oxígeno/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo
15.
Appl Environ Microbiol ; 77(6): 1957-65, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21239541

RESUMEN

Commensal oral streptococci play critical roles in oral biofilm formation and promote dental health by competing with, and antagonizing the growth of, pathogenic organisms, such as Streptococcus mutans. Efficient utilization of the spectrum of carbohydrates in the oral cavity by commensal streptococci is essential for their persistence, and yet very little is known about the regulation of carbohydrate catabolism by these organisms. Carbohydrate catabolite repression (CCR) in the abundant oral commensal Streptococcus gordonii strain DL-1 was investigated using the exo-ß-D-fructosidase gene (fruA) and a fructose/mannose sugar:phosphotransferase (PTS) enzyme II operon (levDEFG) as model systems. Functional studies confirmed the predicted roles of FruA and LevD in S. gordonii. ManL, the AB domain of a fructose/mannose-type enzyme II PTS permease, contributed to utilization of glucose, mannose, galactose, and fructose and exerted primary control over CCR of the fruA and levD operons. Unlike in S. mutans, ManL-dependent CCR was not sugar specific, and galactose was very effective at eliciting CCR in S. gordonii. Inactivation of the apparent ccpA homologue of S. gordonii actually enhanced CCR of fruA and levD, an effect likely due to its demonstrated role in repression of manL expression. Thus, there are some similarities and fundamental differences in CCR control mechanisms between the oral pathogen S. mutans and the oral commensal S. gordonii that may eventually be exploited to enhance the competitiveness of health-associated commensals in oral biofilms.


Asunto(s)
Proteínas Bacterianas/metabolismo , Glicósido Hidrolasas/metabolismo , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/metabolismo , Streptococcus gordonii/enzimología , Streptococcus gordonii/metabolismo , Proteínas Bacterianas/genética , Represión Catabólica/genética , Represión Catabólica/fisiología , Glicósido Hidrolasas/genética , Operón/genética , Sistema de Fosfotransferasa de Azúcar del Fosfoenolpiruvato/genética , Streptococcus gordonii/genética , Streptococcus gordonii/crecimiento & desarrollo
16.
Microbiol Spectr ; 9(2): e0116721, 2021 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-34704809

RESUMEN

When encountering oxidative stress, organisms selectively upregulate antioxidant genes and simultaneously suppress the translation of most other proteins. Eukaryotes employ multiple strategies to adjust translation at both the initiation and elongation stages; however, how prokaryotes modulate translation under oxidative stress remains unclear. Here, we report that upon hydrogen peroxide (H2O2) challenge, Streptococcus oligofermentans reduced translation via RNase Z (So-RNaseZ) oxidative degradation, thus hindering tRNA maturation. S. oligofermentans encodes all CCA-less tRNAs that require So-RNaseZ for 3' end maturation. A combination of nonreducing SDS-PAGE and liquid chromatography/tandem mass spectrometry (LC/MS-MS) assays demonstrated that H2O2 oxidation induced Cys38-Cys149 disulfide linkages in recombinant So-RNaseZ protein, and serine substitution of Cys38 or Cys149 abolished these disulfide linkages. Consistently, redox Western blotting also determined intramolecular disulfide-linked So-RNaseZ in H2O2-treated S. oligofermentans cells. The disulfide-linked So-RNaseZ and monomer were both subject to proteolysis, whereas C149S mutation alleviated oxidative degradation of So-RNaseZ, suggesting that H2O2-mediated disulfide linkages substantially contributed to So-RNaseZ degradation. Accordingly, Northern blotting determined that tRNA precursor accumulation and mature tRNA species decrease in H2O2-treated S. oligofermentans. Moreover, reduced overall protein synthesis, as indicated by puromycin incorporation, and retarded growth of S. oligofermentans occurred in an H2O2 concentration-dependent manner. Overexpression of So-RNaseZ not only elevated tRNA precursor processing and protein synthesis but also partly rescued H2O2-suppressed S. oligofermentans growth. Moreover, So-RNaseZ oxidative degradation-mediated translation repression elevated S. oligofermentans survival under high H2O2 stress. Therefore, this work found that So-RNaseZ oxidative degradation-impeded tRNA maturation contributes to streptococcal translation repression and provides the oxidative stress adaptability for S. oligofermentans. IMPORTANCE Translation regulation is a common strategy used by organisms to reduce oxidative damage. Catalase-negative streptococci produce as well as tolerate high levels of H2O2. This work reports a novel translation regulation mechanism employed by Streptococcus oligofermentans in response to H2O2 challenge, in which the key tRNA endonuclease So-RNaseZ is oxidized to form Cys38-Cys149 disulfide linkages and both the disulfide-linked So-RNaseZ and monomers are subject to proteolysis; thus, tRNA maturation, protein translation, and growth are all suppressed. Notably, So-RNaseZ oxidative degradation-mediated translation repression offers oxidative adaptability to S. oligofermentans and enhances its survival against high H2O2 challenge. So-RNaseZ orthologs and H2O2-sensitive cysteines (Cys38 and Cys149) are widely distributed in Streptococcus and Lactococcus species genomes, which also encode all CCA-less tRNAs and lack catalase. Therefore, RNase Z oxidative degradation-based translation regulation could be widely employed by these lactic acid bacteria, including pathogenic streptococci, to cope with H2O2.


Asunto(s)
Endorribonucleasas/metabolismo , Peróxido de Hidrógeno/toxicidad , Estrés Oxidativo/genética , Biosíntesis de Proteínas/genética , ARN de Transferencia/biosíntesis , Streptococcus/metabolismo , Antioxidantes/metabolismo , Disulfuros/química , Regulación Bacteriana de la Expresión Génica/genética , ARN de Transferencia/genética , Streptococcus/genética , Streptococcus/crecimiento & desarrollo
17.
mBio ; 12(4): e0130921, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34399618

RESUMEN

Aquaporins, integral membrane proteins widely distributed in organisms, facilitate the transport of water, glycerol, and other small uncharged solutes across cellular membranes and play important physiological roles in eukaryotes. However, characterizations and physiological functions of the prokaryotic aquaporins remain largely unknown. Here, we report that Streptococcus pneumoniae (pneumococcus) AqpC (Pn-AqpC), representing a new aquaporin subfamily possessing a distinct substrate-selective channel, functions as an oxygen porin by facilitating oxygen movement across the cell membrane and contributes significantly to pneumococcal virulence. The use of a phosphorescent oxygen probe showed that Pn-AqpC facilitates oxygen permeation into pneumococcal and Pn-AqpC-expressing yeast cells. Reconstituting Pn-AqpC into liposomes prepared with pneumococcal and Escherichia coli cellular membranes further verified that Pn-AqpC transports O2 but not water or glycerol. Alanine substitution showed that Pro232 in the substrate channel is key for Pn-AqpC in O2 transport. The deletion of Pn-aqpC significantly reduced H2O2 production and resistance to H2O2 and NO of pneumococci, whereas low-H2O2 treatment helped the ΔPn-aqpC mutant resist higher levels of H2O2 and even NO, indicating that Pn-AqpC-facilitated O2 permeation contributes to pneumococcal resistance to H2O2 and NO. Remarkably, the lack of Pn-aqpC alleviated cell autolysis, thus reducing pneumolysin (Ply) release and decreasing the hemolysis of pneumococci. Accordingly, the ΔPn-aqpC mutant markedly reduced survival in macrophages, decreased damage to macrophages, and significantly reduced lethality in mice. Therefore, the oxygen porin Pn-AqpC, through modulating H2O2 production and pneumolysin release, the two major pneumococcal virulence factors, controls the virulence of pneumococcus. Pn-AqpC orthologs are widely distributed in various pneumococcal serotypes, highlighting that the oxygen porin is important for pneumococcal pathogenicity. IMPORTANCE Pneumococcus is the leading cause of community-acquired pneumonia, bacteremia, and meningitis. This work reports that a novel aquaporin subfamily represented by pneumococcal Pn-AqpC functions as an oxygen porin facilitating O2 influx into cells. Importantly, by mediating O2 influx, Pn-AqpC controls the production and release of H2O2 and Ply, the two major pneumococcal virulence factors. Moreover, by enhancing endogenous H2O2 production, Pn-AqpC significantly increases pneumococcal resistance to H2O2 and even NO, the major bactericidal chemical produced by macrophages. Consequently, the deletion of Pn-aqpC markedly decreased pneumococcal survival in macrophages and reduced damage to macrophages. Accordingly, the ΔPn-aqpC mutant displays significantly attenuated virulence in a murine pneumonia model. Given that Pn-AqpC orthologs are widely distributed in all pneumococcal serotypes, this new subfamily of aquaporins is identified as novel virulence-related proteins.


Asunto(s)
Acuaporinas/genética , Acuaporinas/metabolismo , Proteínas Bacterianas/metabolismo , Oxígeno/metabolismo , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Factores de Virulencia/metabolismo , Animales , Proteínas Bacterianas/genética , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Peróxido de Hidrógeno/metabolismo , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Células RAW 264.7 , Virulencia , Factores de Virulencia/genética
18.
Risk Manag Healthc Policy ; 14: 655-663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33623455

RESUMEN

BACKGROUND: In China, the prevalence of HUA in the Pearl River Delta (PRD) region of Guangdong Province has not been extensively investigated. Therefore, this study investigated the prevalence of HUA and its related factors among people aged 20-99 years in nine cities in the PRD. MATERIALS AND METHODS: We selected 6491 health check participants from 9 cities in the PRD and collected participants' anthropometric and biochemical test results for a cross-sectional study. We included 6491 participants and assessed their blood pressure (BP), body mass index (BMI), total cholesterol (TC), triglycerides (TG), glucose (Glu) and serum uric acid (UA) to analyze the regional prevalence of HUA and its related factors. HUA was indicated when fasting serum UA level was >420 µmol/L in men and >360 µmol/L in women. RESULTS: Overall prevalence of HUA in our cohort was 34.05%; prevalence was higher in men than in women (41.53% vs 26.14%, P < 0.001). Characteristics associated with HUA were hypertension (odds ratio (OR), 5.506; 95% confidence interval (CI), 4.402-6.889), higher body mass index (BMI; OR: 1.746; 95% CI: 1.560-1.954), age 31-40 years (OR: 0.829; 95% CI: 0.706-0.973), age 61-70 years (OR: 1.434; 95% CI: 1.194-1.722) and age ≥71 years (OR: 1.742; 95% CI: 1.397-2.173). In all subjects, serum UA was positively correlated with Glu, TG and TC. After we adjusted for age, BMI and BP, multivariate logistic regression analysis showed that HUA risk factors were high TC (OR: 1.770; 95% CI: 1.459-2.147) and TG (OR: 1.961; 95% CI: 1.632-2.357) in men; and high Glu (OR: 1.508; 95% CI: 1.084-2.099), TC (OR: 1.341; 95% CI: 1.084-1.660) and TG (OR: 1.680; 95% CI: 1.290-2.187) in women. CONCLUSION: The prevalence of HUA was relatively high in the PRD of Guangdong Province. Relevant governmental bodies should focus on early diagnosis, early treatment and early intervention.

19.
mSystems ; 5(2)2020 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184366

RESUMEN

Preexposure to a low concentration of H2O2 significantly increases the survivability of catalase-negative streptococci in the presence of a higher concentration of H2O2 However, the mechanisms of this adaptation remain unknown. Here, using a redox proteomics assay, we identified 57 and 35 cysteine-oxidized proteins in Streptococcus oligofermentans bacteria that were anaerobically cultured and then pulsed with 40 µM H2O2 and that were statically grown in a 40-ml culture, respectively. The oxidized proteins included the peroxide-responsive repressor PerR, the manganese uptake repressor MntR, thioredoxin system proteins Trx and Tpx, and most glycolytic proteins. Cysteine oxidations of these proteins were verified through redox Western blotting, immunoprecipitation, and liquid chromatography-tandem mass spectrometry assays. In particular, Zn2+-coordinated Cys139 and Cys142 mutations eliminated the H2O2 oxidation of PerR, and inductively coupled plasma mass spectrometry detected significantly decreased amounts of Zn2+ in H2O2-treated PerR, demonstrating that cysteine oxidation results in Zn2+ loss. An electrophoretic mobility shift assay (EMSA) determined that the DNA binding of Mn2+-bound PerR protein (PerR:Zn,Mn) was abolished by H2O2 treatment but was restored by dithiothreitol reduction, verifying that H2O2 inactivates streptococcal PerR:Zn,Mn through cysteine oxidation, analogous to the findings for MntR. Quantitative PCR and EMSA demonstrated that tpx, mntA, mntR, and dpr belonged to the PerR regulons but that only dpr was directly regulated by PerR; mntA was also controlled by MntR. Deletion of mntR significantly reduced the low-H2O2-concentration-induced adaptation of S. oligofermentans to a higher H2O2 concentration, while the absence of PerR completely abolished the self-protection. Therefore, a low H2O2 concentration resulted in the cysteine-reversible oxidations of PerR and MntR to derepress their regulons, which function in cellular metal and redox homeostasis and which endow streptococci with the antioxidative capability. This work reveals a novel Cys redox-based H2O2 defense strategy employed by catalase-negative streptococci in Mn2+-rich cellular environments.IMPORTANCE The catalase-negative streptococci produce as well as tolerate high levels of H2O2 This work reports the molecular mechanisms of low-H2O2-concentration-induced adaptation to higher H2O2 stress in a Streptococcus species, in which the peroxide-responsive repressor PerR and its redox regulons play the major role. Distinct from the Bacillus subtilis PerR, which is inactivated by H2O2 through histidine oxidation by the Fe2+-triggered Fenton reaction, the streptococcal PerR is inactivated by H2O2 oxidation of the structural Zn2+ binding cysteine residues and thus derepresses the expression of genes defending against oxidative stress. The reversible cysteine oxidation could provide flexibility for PerR regulation in streptococci, and the mechanism might be widely used by lactic acid bacteria, including pathogenic streptococci, containing high levels of cellular manganese, in coping with oxidative stress. The adaptation mechanism could also be applied in oral hygiene by facilitating the fitness and adaptability of the oral commensal streptococci to suppress the pathogens.

20.
Wei Sheng Wu Xue Bao ; 49(10): 1341-6, 2009 Oct.
Artículo en Zh | MEDLINE | ID: mdl-20069880

RESUMEN

OBJECTIVE: Streptococcus oligofermentans was a newly characterized streptococcal species, which we isolated from dental plaques of carious-free humans. Our previous work indicated that in aerobic environments, it produced, as well as survived high concentration of hydrogen peroxide (4.4 mmol/L). This study analyzed the function of dpr gene in hydrogen peroxide tolerance of Streptococcus oligofermentans. METHODS: Cloned and expressed the dpr gene, and characterized Dpr protein. We constructed the dpr mutant strain of S. oligfermentans and detected its susceptivity to hydrogen peroxide. Furthermore, S. oligofermentans dpr gene was cloned into S. mutans, and its susceptivity to hydrogen peroxide was tested. RESULTS: S. oligofermentans dpr mutant strain was much more susceptive to hydrogen peroxide than the wild type, and introduction of its dpr gene into S. mutans could enhance the later' s hydrogen peroxide tolerant ability. Dpr proteins could bind irons, but not to DNA. [Conclusion] dpr gene of S. oligofermentans has significant role in hydrogen peroxide tolerance of the host strain, while Dpr protein may have other oxidant-damage protective mechanisms except for iron-binding.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de Unión al ADN/metabolismo , Peróxido de Hidrógeno/metabolismo , Streptococcus/metabolismo , Proteínas Bacterianas/genética , Proteínas de Unión al ADN/genética , Regulación Bacteriana de la Expresión Génica , Streptococcus/genética , Streptococcus mutans/genética , Streptococcus mutans/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda